. , Les radicaux alcoxy (RO) et peroxy (ROO)

. , Production au niveau des membranes du réticulum endoplasmique et de la membrane nucléaire

. , Production de radicaux libres liée au facteurs environnementaux

. , Production de radicaux libres en situation pathologique

. , L'agression radicalaire et ses sources biologiques

. .. , 1. 3. La glutathion peroxydase séléno-dépendante ( GPx)

M. Marty, F. Calvo, M. Espie, J. M. Lagner, I. Florentin et al., Phannacologie clinique: bases thérapeutiques. Expansion Scientifique Française. 2° édition p, pp.1894-1945, 1988.

J. Chauvergne and B. Hoerni, Chimiothérapie anticancéreuse. 2° édition, 1992.

J. Calop and G. Carraz, Anticancéreux et cancérogènes, pp.181-229, 1988.

A. Jacquemin-sablon, Phannacologie moléculaire. Mécanismes d'action des médiateurs et des médicaments· 2ème édition Arnette, pp.603-636, 1993.

J. P. Annand, J. P. Cano, G. Fabre, A. Gouyette, R. Rhaman et al., Tubiana M. Thérapeutique des cancers. Edition Flammarion, pp.193-287, 1986.

L. Jeune, C. Paule, and B. , Chimiothérapie anticancéreuse, In: Impact internat. Thérapeutique, vol.20, pp.381-387, 1993.

M. Clavel, Chimiothérapies anticancéreuses. Classification et mode d'action, Rev .Prat, vol.41, pp.59-2766, 1991.

G. Powis and M. P. Hacker, The toxicity ofanticancer drugs, 1991.

M. Samoun, A. C. Potocka, R. Pillière, G. Delonne, J. Prinseau et al., , vol.20, pp.458-460, 1991.

L. H. Fraisser, S. Kanekal, and P. Kehrerg, Cyclophosphamide toxicity, characterising and avoiding the problem, Drugs, vol.42, pp.781-795, 1991.

J. Emerit, J. Fechner, A. Galli, J. P. Clavel, and F. Congy, Les radicaux libres dérivés de l'oxygène et la peroxydation lipidique. Rôle en biologie cellulaire et en physiopathologie, Presse Medicale, vol.15, pp.751-754, 1986.

P. M. Reilly, H. J. Schiller, and G. B. Bulkley, Phannacologic approach to tissue injury mediated by free radicals and other reactive oxygen metabolites, Am. J; Surg, pp.488-503, 1990.

J. Emerit and A. M. Michelson, Les radicaux libres en médecine et en biologie, Sem.Hop, vol.58, pp.2670-2675, 1982.

J. P. Clavel, J. Emerit, and A. Thuillier, Lipoperoxydation et radicaux libres. Rôle en biologie cellulaire et pathologie, Path. Biol, vol.33, pp.61-69, 1985.

B. A. Freedman and J. D. Crapo, Biology of disease. Free radicals and tissue injury, Lab. lnvest, vol.47, pp.412-426, 1982.

J. Chouvet, Place des radicaux libres dans la physiopathologie des arthropathies des hémodialysés, Thèse Pharmacie. Université J. Fourier Grenoble, 1989.

E. Anggard, Nitric oxide: mediator, murderer and medicine, Lancet, vol.343, pp.1199-1206, 1994.

A. L. Lehninger, D. L. Nelson, and M. C. Cox, Principes de biochimie. 2nde édition Flammarion. 19-Brawn K., Fridovich 1. (1980) Superoxide radical and superoxide dismutase threat and defense, Acta Physiol.Scand, vol.492, pp.9-18, 1994.

G. M. Cohen, . D'arcy, and M. Doherty, Free radical mediated cell toxicity by redox cycling chemicals, Br. J. Cancer, vol.55, pp.46-52, 1987.

C. E. Vaca, J. Wilhelm, and M. Harm-ringdahl, Interaction of lipid peroxidation products with DNA. A review, Mut. Res, vol.195, pp.137-149, 1988.

F. T. Slater, Free radical mechanisms in tissue injury, Biochem. J, pp.1-15, 1984.

A. Favier, Intérêt des marqueurs de la peroxydation lipidique, Compte rendus des XIII emes journées nationales de biologie. Lyon, pp.45-53, 1990.

. P. Wolffs, A. Gainer, and D. T. Dean, Free radicals, lipids and protein degradation. TIBS, 11 29-31. 25-Imlay, Science, vol.240, pp.1302-1309, 1986.

J. F. Ward, J. W. Evans, C. L. Limoli, and P. M. Calabro-jone, Radiation and hydrogen peroxide-induced free radical damage to DNA, Br. J. Cancer, vol.55, pp.105-112, 1987.

J. F. Mouret, Formation d'adénine-N-oxyde sur l'ADN par action de l'eau oxygénée ou de l'acide métachloroperbenzoïque. Thèse Pharmacie, Université J. Fourier Grenoble, 1987.

G. Jadot, Les superoxydes dismutases: biochimie, pharmacologie, thérapeutique, 1988.

S. Sato, M. Iwaizumi, K. Handa, and Y. Tamura, Electron spin resonance study on the mode of generation of free radicals of daunomycin, adriamycin, and carboquone in NAD(P)H microsome systeme, Gann, vol.68, pp.603-608, 1977.

J. Butler and B. M. Hoey, Are reduced quinones necessarily involved in the antitumor activity of quinone drags, Br. J. Cancer, vol.55, pp.53-59, 1987.

J. Butler, B. M. Hoey, and A. J. Swallow, Generation ofhydroxyl radical by the anticancer quinone drags, carbazilquinone, mitomycin C, aclacinomycin A and adriamycin, in the presence of NADPH-cytochrome P 450 reductase, Biochem. Pharmacol, vol.182, pp.3551-3656, 1982.

D. D. Pietronigro, J. E. Mc-ginness, M. J. Koren, R. Crippa, M. L. Seligman et al., ~1979) Spontaneous generation ofadriamycin semi-quinone at physiologie pH, Physiol.Chem.Phys, vol.11, pp.405-413

P. J. Thornalley, W. H. Bannister, and J. V. Bannister, Reduction of oxygen by NADH/NADH dehydrogenase in the presence of adriamycin, Free. Res. Commun, vol.2, pp.163-171, 1986.

E. G. Mimnaugh, E. G. Theodore, and M. A. Trush, Stimulation of mouse heart and liver microsomal lipid peroxidation by anthracycline anticancer drugs. Characterization and effects of reactive oxygen scavengers, J. Pharm. Exp. Ther, vol.226, pp.806-816, 1983.

C. Winterboum, Evidence for the production ofhydroxyl radicals from the adriamycin semiquinone and H2ü2, Febs lett, vol.136, pp.89-94, 1981.

D. A. Bates and . C. Winterbourn, Deoxyribose breakdown by the adriamycin semiquinone and H2 02 Evidence for hydroxyl radical participation, Febs. Lett, vol.145, pp.137-142, 1982.

H. Beraldo, A. Garnier-suillerot, L. Tosi, and F. Lavelle, Iron III-adriamycin and iron III-daunorubicin complexes physicochemical characteristics, interaction with DNA, and antitumor activity, Biochemistry, vol.24, pp.284-289, 1985.

C. E. Thomas and S. D. Aust, Release of iron from ferritin by cardiotoxic anthracycline antibiotics, Arch. Biochem. biophys, vol.248, pp.684-689, 1986.

L. Gianni, J. L. Zweier, A. Levy, and C. E. Myers, Characterisation of the cycle of iron-mediated electron transfer from adriamycin to molecular oxygen, J.Biol.Chem, vol.260, pp.6820-6826, 1985.

J. M. Gutteridge, Lipid peroxidation and possible hydroxyl radical formation stimulated by the selfreduction of a doxorubicin iron(III) complex, Biochem.Pharmacol, vol.33, pp.1725-1728, 1984.

J. R. Muindi, B. K. Sinha, L. Gianni, and C. E. Myers, Hydroxyl radical production and DNA damage induced by anthracycline-iron complexes, Febs Lett, vol.172, pp.226-230, 1984.

K. B. Wallace, Non-enzymatic oxygen activation and stimulation of lipid peroxidation by doxorubicincopper, Tox. Appl. Pharmacol, vol.86, pp.69-79, 1984.

B. K. Sinha, M. A. Trush, K. A. Kennedy, and E. G. Mimnaugh, Enzymatic activation and binding of Adriamycin to nuclear DNA, Cancer Res, vol.27, p.24, 1984.

M. E. Scheulen, H. Kappus, A. Nienhaus, and G. C. Schmidt, Covalent protein binding of reactive adriamycin metabolites in rat liver and rat heart microsomes, J.Cancer Res.Clin, vol.103, pp.39-48, 1982.

M. G. Ghezzi, M. G. Donelli, C. Pantarotto, R. Facchinetti, and S. Garattini, Evidence for covalent binding of Adriamycin to rat liver microsomal protein, Biochem. Pharmacol, vol.30, pp.175-182, 1981.

B. K. Sinha, Free radicals in anticancer drug pharmacology, Chem. Bio!. Interaction, vol.69, pp.293-317, 1989.

J. H. Doroshow, Prevention of doxorubicin-induced killing of MCF 7 human breast cancer cells by oxygen radical scavengers and iron chelating agents, Biochem. Biophys. Res. Commun, vol.135, pp.330-335, 1986.

J. H. Doroshow, Role of hydrogen peroxide and hydroxyl radical formation in the killing of Ehrlich tumor cells by anticancer quinones, Proc. Natl. Acad. Sei, vol.83, pp.4514-4518, 1986.

B. K. Sinha, A. G. Katki, B. Batist, K. H. Cowan, and C. E. Myers, Adriamycin stimulated hydroxyl radical formation in human breast tumor cells, Biochem. Pharmacol, vol.36, pp.793-796, 1987.

A. Bozzi, . Mavelli-1, B. Mondovi, R. Strom, and G. Rotilio, Differential cytotoxicity of daunomycin in tumor cells is related to glutathione-dependent hydrogen peroxide metabolism, Biochem. J, vol.194, pp.369-372, 1981.

A. Russo and J. B. Mitchell, Potentiation and protection of doxorubicin cytotoxicity by cellular glutathione modulation, Cancer Treat. Rep, vol.69, pp.1293-1296, 1985.

V. Berlin and W. A. Haseltine, Reduction of adriamycin to a semi-quinone free radical by NADPHcytochrome P450 reductase produces DNA cleavage in a reaction mediated by molecular oxygen, J. Bio!. Chem, vol.10, pp.4747-4756, 1981.

C. W. Haidle, M. Kinney, and S. H. , Adriamycin-mediated introduction of a limited number of single strand breaks into supercoiled DNA, Cancer Biochem. Biophys, vol.8, pp.327-335, 1986.

J. W. Akman, J. H. Doroshow, T. G. Burbe, and M. Dizdaroglu, 56-Svingen B.A., Powis G. (1981) Pulse radiolysis studies on antitumor quinones: radical lifetimes, reactive with oxygen and one-electron reduction potentials, Arch. Biochem. Biophys, vol.31, pp.3296-3304, 1980.

K. Ogura and R. , Effects of vitamins on lipid peroxidation and suppression of DNA synthetis induced by adriamycin in Ehrlich cells, J. Nutr. Sei. Vitaminol, vol.31, pp.129-134, 1985.

J. W. Lown, H. H. Chen, J. A. Plambeck, and E. M. Acton, Further studies on the generation of reactive oxygen species from activated anthracyclines and the relationship to cytotoxic action and cardiotoxic effects, Biochem. Pharmacol, vol.31, pp.575-581, 1982.

G. L. Tong, D. W. Henry, E. M. Acton, A. C. 63-dickinson, M. G. Boutin et al., OH generation by adriamycin semi-quinone and H202 and explanation for the cardiotoxicity of anthracycline antibiotics, Res. Commun. Chem. Path. Pharm, vol.22, pp.445-459, 1979.

J. H. Doroshow and K. J. Davies, Redox cycling of anthracycline by cardiac mitochondria. Formation of superoxyde anion, hydrogen peroxyde and hydroxyl radical, J.Biol.Chem, vol.261, pp.3068-3074, 1986.

J. H. Doroshow, Effect of anthracycline antibiotics on oxygen radical formation in rat heart, Cancer Res, vol.43, pp.460-472, 1983.

J. A. Davies, J. H. Doroshow, P. Hochstein, R. A. Floyd, R. Henderson et al., Use of salicylate with high pressure liquid chromatography and electrochemical detection as a sensitive measure of hydroxyl free radical in adriamycintreated rats, J. Free Radical Biol.Med, vol.153, pp.13-18, 1983.

E. G. 72-mimnaugh, M. A. Trush, and T. E. Gram, Enhancement of reactive oxygen-dependant mitochondrial membrane lipid peroxidation by the anticancer drug adriamycin, Biochem. Pharmacol, vol.30, pp.847-856, 1981.

S. W. 4-thayer, Evaluation of tissus indicators of oxydative stress, in rats treated chronically with adriamycin, Biochem. Pharmacol, vol.37, pp.2189-2194, 1988.

N. Koukay, S. Mouhieddine, M. J. Richard, J. Arnaud, J. De-leiris et al., Influence of selenium on lipid peroxidation and cardiac functions in chronically adriamycin-treated rats, Antioxydants in therapy and preventive medecine, vol.39, pp.769-774, 1990.

E. G. Mimnaugh, Z. H. Siddik, R. Drew, B. I. Sikic, and T. E. Gram, Failure of the antioxydant Vitamin E to protect against Adriamycin-induced cardiotoxicity in the rabbit, Toxicol. Appl. Pharmacol, vol.49, pp.2033-2038, 1979.

E. H. Herman and V. J. Ferrans, Influence ofvitamin E and ICRF-187 on chronic doxorubicin cardiotoxicity in miniature swine, Lab. lnvest, vol.49, pp.69-77, 1983.

S. S. Legha and Y. M. Wang, Clinical and pharmacologie investigation of the effect ofo-tocopherol on adriamycin toxicity, Ann. N.Y. Acad. Sei, vol.393, pp.411-418, 1982.

J. A. Whittaker and S. A. Al-ismail, Effect of digoxine and vitamine E in preventing cardiac damage caused by doxorubicin in acute myeloîd leukaemia, British Med. J, vol.288, pp.283-284, 1984.

K. Fugita, K. Shinpo, K. Yamada, T. Sato, H. Niimi et al., Prevention of doxorubicin myocardial toxicity in mice by reduced glutathion, Cancer Res, vol.42, pp.2551-2556, 1982.

D. V. 84-unverferth, C. V. Leier, S. P. Balcerzak, and R. L. Hamlin, Usefulness of a free radical scavenger in preventing doxorubicin-induced heart failure in dogs, Am. J. Cardiol, vol.56, pp.157-161, 1985.

E. H. Herman and V. J. Ferrans, Reduction of chronic doxorubicin cardiotoxicity in dogs by pretreatrnent with 1,2 bis(3-5 dioxopiperazinyl) propane (ICRF 187), Res. Commun. Chem. Pathol. Pharmacol, vol.41, pp.85-97, 1981.

P. Alderton, J. Gross, M. D. Green, J. L. Speyer, M. D. Green et al., Reductive activation of mitomycin C and mitomycin metabolites catalysed by NADPH cytochrome P-450 reductase and xanthine oxidase, Biochem. Biophys. Acta, vol.3, pp.959-966, 1978.

M. A. Trush, E. G. Mimnaugh, E. Ginsburg, T. E. Gram, T. Komiyama et al., Interaction of anticancer quinone drugs, aclacinomycin A, adriamycin, carbazilquinone and mitomycin C with NADPH cytochrome P 450 reductase, xanthine-oxydase and oxygen, Biochem. Pharmacol, vol.31, pp.651-664, 1982.

J. M. Gutteridge, G. J. Quinlan, and S. Wilkins, Mitomycin C induced deoxyribose degradation inhibited by superoxyde dismutase. A reaction involving iron, hydroxyle and semiquinone radicals, Cancer Res, vol.167, pp.3528-3532, 1984.

C. A. Pristos, S. R. Keyes, and A. C. Sartorelli, Role of oxygen radicals in the toxicity of mitomycin antibiotics to EMT6 tumor cell, Proc. Amer. Assoc. Cancer Res, vol.27, pp.1760-1764, 1986.

M. C. Krishna, . De-graffw, S. Tamura, F. J. Gonzales, A. Samum et al., Mechanisms ofhypoxic and aerobic cytotoxicity ofmitomycin C in chinese hamster V79 cells. Cancer Res. 6622-6628, Biochem. Pharmacol, vol.30, pp.2881-2884, 1981.

B. K. Sinha, A. G. Motten, and K. W. Hanck, The electrochemical reduction of l,4-bis-(2-(2-hydroxyethyl)amino )ethylamino-anthracenedione and daunomycin biochemical signifiance in superoxide formation, Chem. Bio!. Interact, vol.43, pp.371-377, 1983.

E. D. Karasch and R. F. Novak, Inhibition of adriamycin-stimulated microsomal lipid peroxidation by mitoxantrone and amenantrone, two new anthracenedione antineoplastic agents, Preclinical Pharmacol. and Exp. Therap, vol.12, pp.1346-1352, 1982.

E. D. Karasch and R. F. Novak, Spectroscopie evidence for anthracenedione antineoplastic agent selfassociation and complex formation with flavin nucleotides, Proc. Amer. Assoc. Cancer Res, vol.234, p.75, 1978.

W. D. Flitter and R. P. Mason, The enzymatic reduction of actinomycin D to a free radical species, Arch. Biochem. Biophys, vol.267, pp.632-639, 1988.

M. G. Cox, B. K. Sinha, H. Kasai, H. Naganawa, H. Takita et al., Interaction of bleomycin with nucleic acids, preferential binding to guanine base and electrostatic effect of the terminal amine, es sciences pharmaceutiques. Faculté pharmacie. Lilles. 109-Takita T, vol.267, pp.1073-1077, 1978.

E. A. Sausville, J. Peisach, and S. B. Horwitz, Effect of chelating agents and metal ions on the degradation ofDNA by bleomycin, Biochemistry, vol.17, pp.2740-2746, 1978.

P. C. Dedon and H. J. Goldberg, Free radical mechanisms involved in the formation of sequence-dependent bistranded DNA lesions by the antitumor antibiotics bleomycin, neocarzinostatin, calicheamicin. Res. Tox, vol.5, pp.311-332, 1992.

Y. Suguira, K. Kikuchi, M. E. Scheulen, H. Kappus, D. Thyssen et al., Formation of superoxide and hydroxyl radicals in iron (II) bleomycin oxygene system. ESR detection by spin trapping, Biochem. Pharmacol, vol.12, pp.8608-8615, 1978.

, Bleomycin-dependent damage to the bases in DNA is a minor side reaction, Biochemistry, vol.30, pp.2444-2448, 1991.

C. J. Doelman, A. Bast, J. G. Hay, P. L. Haslam, M. Turner-warwick et al., The effect of iron and desferrioxamine on the lung injury induced by intravenous bleomycin and hyperoxia, Free Rad. Res. Commun, vol.9, pp.315-329, 1987.

J. Hay, S. Shahzeidi, and G. Laurent, Alteration in pulmonary protective enzymes following systemic bleomycin treatment in mice, Biochem.Pharmacol, vol.65, pp.1111-1116, 1988.

S. N. Giri, Z. L. Chen, W. R. Youker, and M. J. Schiedt, Effects ofintratracheal administration ofbleomycin on GSH shuttle enzymes, catalase, lipid peroxydation and collagen content in the lung of hamster, Toxicol. Appl. pharmacol, vol.71, pp.132-141, 1983.

C. Bailly, J. C. Beauvillain, J. L. Bernier, and J. P. Henichart, Plasma membrane perturbation of KB3 cells induced by bleomycin iron complex, Cancer Res, vol.50, pp.385-392, 1990.

H. Kikuchi and T. Tetsuka, On the mecanism oflipooxygenase-like action ofbleomycin-iron complexes, J. Antib, vol.45, pp.548-555, 1992.

D. B. Chandler, J. C. Barton, D. D. Briggs, T. W. Butler, J. I. Kennedy et al., Peroxidation of arachidonic acid by bleomycin. A possible mechanism of bleomycin pulmonary toxicity, Proc. Amer. Assoc. Cancer Res, vol.46, p.9994, 1988.

M. Jordana, C. Richard, L. B. Irving, and J. Gauldie, Spontaneous in vitro release of alveolar macrophage cytokines after the intratracheal instillation of bleomycin in rats. Characterisation and kinetic studies, Am. Rev. Respir. Dis, vol.137, pp.1135-1140, 1988.

D. O. Sloman, D. M. Costabella, M. Roth, G. Werlen, B. S. Polla et al., Lack of metabolism as the biochemical basis of bleomycin-induced pulmonary toxicity, Proc. Natl. Acad. Sei, vol.3, pp.2386-2389, 1983.

W. M. Haschek, K. E. Baer, and J. E. Rutherford, Effects of dimethyl sulfoxyde (DMSO) on pulmonary fibrosis in rats and mice, Toxicology, vol.54, pp.197-205

H. E. Wand, A. Nicholson, and N. Berend, Failure of systemic N-acetylcysteine to protect the rat lung against bleomycin toxicity, Pathology, vol.19, pp.353-360, 1987.

S. Shahzeidi, B. Sarnstrand, P. K. Jeffery, R. J. Mc-anulty, G. J. Laurent et al., Oral N-Acetylcysteine reduces bleomycin-induced collagen deposition in the lungs ofmice, Toxicol. Appl. Pharmacol, vol.4, pp.127-138, 1985.

B. K. Sinha and M. A. Trush, Free radical of VP-16 and inhibition of anthracyclin-induced lipid peroxidation, Biochem. Pharmacol, vol.22, pp.3495-3498, 1983.

N. Haim, J. Nemec, J. Roman, B. K. Sinha, N. Haim et al., In vitro metabolism of etoposide (VP-16-213) by liver microsomes and irreversible binding of reactive intermediates to microsomal proteins, Biochem. Pharmacol, vol.36, pp.2131-2139, 1987.

C. Auclair, K. Hyland, and C. Paoletti, Autoxidation of the antitumor drug 9-hydroxyellipticine and its derivatives, J. Med. Chem, vol.26, pp.1438-1444, 1983.

B. Dugue, C. Auclair, and B. Meunier, Covalent binding of elliptinium acetate (NCS 264137) to nucleic acid ofL2110 cells in culture, Cancer Res, vol.46, pp.3828-3833, 1988.

C. Dadoun and G. Raguenez-viotte, Celliptium induced nephrotoxicity and lipid peroxydation in rat renal cortex, Cancer Chemother. Pharmacol, vol.27, pp.178-186, 1990.

N. Thomas, G. Raguenez-viotte, and M. Dieber-rotheneder, Time course study of lipid peroxidation induced by N2-methyl-9-hydroxyellipticinium acetate or celliptium in rat renal cortex, Pharmacol. Toxicol, vol.69, pp.112-116, 1991.

D. Gergel, :. V. Misik, and K. Ondrias, Cis-platine-induced lipid peroxydation and decrease of gluconeogenesis in rat kidney cortex: different effects of antioxydants and radical scavengers, Int. J. Immunopharmacol, vol.41, pp.709-714, 1986.

Y. Sadzuka, T. Shogi, and Y. Takino, Effect of cisplatin on the activity of enzymes wich protect against lipid peroxidation, Biochem. Pharmacol, vol.43, pp.1872-1875, 1992.

J. G. Zangh and W. E. Lindup, Role ofmitochondria in cisplatin-induced oxidative damage exhibited by rat renal cortical slices, Biochem. Pharmacol, vol.45, pp.2215-2222, 1993.

J. G. Zhang and W. E. Lindup, Cisplatin nephrotoxicity: decreases in mitochondrial protein sulfphydryl concentration and calcium uptake by mitochondria from rat renal cortical slice, Biochem. Pharmacol, vol.47, pp.1127-1135, 1994.

S. Nakano and M. Gemba, Potentation of cisplatin-induced lipid peroxidation in rat kidney cortical slices by glutathion depletion, Jpn J. Pharmacol, vol.50, pp.87-92, 1989.

G. Bompart, Cisplatin-induced changes in cytochrome P 450, lipid peroxydation and drug-metabolizing enzyme activities in rat kidney cortex, Toxicol. Lett, vol.48, pp.193-199, 1989.

B. K. Sinha, Metabolic activation of procarbazine. Evidence for carbon-centered free radical intermediates, Biochem. Pharmacol, vol.17, pp.2777-2781, 1984.

K. Edo, M. Mizugaki, Y. Koide, H. Seto, K. Otake et al., Mode of reversible binding of neocarzinostatin chromophore to DNA. Evidence ofbinding via the minor groove, Tetrahedron Lett, vol.26, pp.6913-6920, 1985.

D. H. Chin and I. H. Golberg, Production of superoxide free radical by neocarzinostatin and its possible role in DNA damage, Biochemistry, vol.25, pp.1009-1015, 1986.

S. M. Keyse and M. T. Rex, Herne oxygenase is the major 32 kDa stress protein induced in human skin fibroblast by UV A radiation, hydrogen peroxide and sodium arsenite, Proc. N atl. Acad. Sei. USA, vol.86, pp.1043-1046, 1987.

S. M. Keyse and R. M. Tyrrel, Induction of the heme oxygenase in human skin fibroblasts by hydrogen peroxide and UV A (365 nm) radiation: evidence for the involvement of the hydroxyl radical, Carcinogenesis, vol.11, pp.1551-1554, 1988.

R. A. Donati, D. O. Slosman, and B. Poila, Oxidative injury and the heat shock response, Biochem. Pharmacol, vol.40, pp.2571-2577, 1990.

S. T. Nowak, H. Kato, Y. Liu, K. Kogure, and K. Kato, Induction of 27 kDa heat shock protein following cerebral ischemia in rat model ofischemic tolerance, Free Radical Res. Commun, vol.45, issue.167, pp.129-139, 1985.

D. R. Spitz, W. C. Dewey, G. C. Li, J. Ananthan, A. L. Golberg et al., Increased survival after treatments with anticancer agents of Chinese hamster cell expressing the human 27 ,000 heat shock protein, The small heat shock protein HSP27 is correlated with growth and drug resistance in human breast cancer lines, vol.131, pp.4443-4448, 1986.

S. S. Donalson, L. F. Cordon, and G. M. Hahn, Protective effect ofhyperthermia against the cytotoxicity of actinomycin Don Chinese hamster cells, Cancer Treat. Rep, vol.62, pp.1489-1495, 1978.

S. A. Huber, Heat shock protein induction in Adriamycin and picomavirus infected cardiocytes, Lab. Invest, vol.62, pp.218-224, 1992.

S. Schunck, H. Rainer, B. Bielka, and H. , Cisplatin induces the small heat shock protein HSP25 and thermotolerance in Ehrlich ascites tumor cells, Biochem. Biophys. Res. Comm, vol.180, pp.243-248, 1991.

P. L. Moseley, S. J. York, and J. York, Bleomycin induces the HSP 70 heat shock promoter in cultured cells, Am. J. Resp. Cell. Mol. Bio!, vol.1, pp.89-93, 1989.

J. M. Croop, P. Gros, D. E. Housman, H. T. Albeson, D. E. Housman et al., Radiation survival paramaters ofantineoplastic drug sensitive and resistant human ovarian cancer cell lines and their modification by buthionine sulfoximine, Critical Review. Mol. Bio!, vol.81, pp.2110-2115, 1984.

C. Meijer, N. H. Mulder, G. A. Hosper, D. R. Uges, E. G. De-vries et al., Cellular resistance to oxidative stress is accompagned by resistance to cisplatin. The signifiance of increased catalase activity and total glutathione content in hydrogen peroxide-resistant fibroblasts, J. Cell. Physiol, vol.62, issue.186, pp.72-79, 1990.

V. Gupta, S. V. Singh, H. Ahmad, R. D. Medh, Y. C. Awasthi et al., Reduced levels of drug-induced DNA cross-linking in nitrogen mustard resistant chinese hamster ovary cells expressing elevated glutathione-S-transferase activity, Proc. Nat!. Acad. Sei. USA, vol.38, pp.8511-8515, 1987.

Y. Y. Wang, B. A. Teicher, T. C. Shea, S. A. Holden, K. W. Robse et al., Cross resistance and glutathione-S-transferase n levels among four human melanoma cell lines selected for alkylating agentresistance, Cancer Res, vol.49, pp.6185-6192, 1988.

M. T. Smith, C. J. Evans, D. Seltzer, P. Castro, V. M. Tahir et al., Denitrosation of a 1-3 bis (2-chloroethyl)-1-nitrosourea by class µ glutathion transferase and its role in cellular resistance in rat brain tumor cells Cancer Res, vol.49, pp.2651-2655, 1989.

G. Batist, A. Tulpule, B. K. Sinha, A. G. Katki, C. E. Myers et al., Overexpression of a novel anionic glutathione transferase in multi-drug resistant human breast cancer cell, J. Biol. Chem, vol.33, pp.15544-15549, 1986.

S. V. Singh, S. Nair, H. Ahmad, Y. C. Awasthi, and A. Krishan, Glutathion S transferases and glutathion peroxidases in doxorubicin-resistant murine leukemic P388 cells, Biochem. Pharmacol, vol.20, pp.3505-3510, 1989.

F. Y. Lee, J. Sciandra, and D. W. Sieman, A study of the mechanism of resistance to adriamycin. Glutathione metabolism, P-glycoprotein expression, and drug transport, Biochem. Pharmacol, vol.21, pp.3697-3705, 1989.

B. K. Sinha, A. G. Katki, G. Batist, K. H. Cowan, and C. E. Myers, Differential formation ofhydroxyl radical by adriamycin in sensitive and resistant MCF7 human breast tumor cells. Implication for the mechanism of action, Biochemistry, vol.26, pp.3776-3781, 1987.

W. L. Bigbee, A. J. Wyrobek, R. G. Langlois, R. H. Jensen, and R. B. Everson, The effect of chemotherapy on the in vivo frequency of glycophorin A "null" variant erythrocytes, Mutation Res, vol.240, pp.165-175, 1990.

F. P. Perera, R. J. Motzer, D. Tang, E. Reed, R. Parker et al., Multiple biological markers in germ cell tumor patients treated with platinum-based chemotherapy, Cancer Res, vol.52, pp.3558-3565, 1992.

S. G. Grant and W. L. Bigbee, Bone marrow somatic mutation after genotoxic cancer therapy, vol.343, pp.1507-1508, 1994.

M. W. Gary and J. H. Weisburger, Chemical carcinogenesis, Casarett and Doull's Toxicology, pp.127-199, 1991.

P. Bienvenu and D. Gasparutto, Stimulation de la peroxydation lipidique in vitro par la vitamine KI, la chlorméthine et l'orellanine, au moyen d'un système à base de microsomes hépatiques et d'un générateur de NADPH, 1993.

H. Masuda, T. Tanaka, and U. Takahama, Cisplatin generates superoxide anion by interaction with DNA in a cell-free system, Biochem. Biophys. Res. Comms, vol.203, pp.1175-1180, 1994.