Review article
Free radicals in anticancer drug pharmacology

https://doi.org/10.1016/0009-2797(89)90117-8Get rights and content

Abstract

This review examines the formation of free radical intermediates from a number of clinically active antitumor agents including quinone-containing antibiotics and etoposide. An attempt is also made to relate the formation of these reactive intermediates to biochemical and pharmacological basis for tumor cell kill and resistance. The formation of these intermediates in some tumor cells has been detected by both direct ESR and spin-trapping technique. The detection of free radicals in biological systems, however, depends upon cellular bioenvironments, e.g. reducing conditions, and the presence and/or absence of activation and detoxification mechanisms. Evidence shows that certain antitumor drugs generate free radicals in vitro and in vivo and that these reactive species kill tumor cells by causing damage to DNA, membranes or enzymes.

References (115)

  • B.K. Sinha et al.

    Binding of Adriamycin to cellular macromolecules in vivo

    Biochem. Pharmacol.

    (1980)
  • E. Goormaghtigh et al.

    Evidence of a specific complex between Adriamycin and negatively charged phospholipids

    Biochim. Biophys. Acta

    (1980)
  • M. Potmesil et al.

    Two mechanisms of Adriamycin-DNA interactions in L1210 cells

    Biochem. Pharmacol.

    (1984)
  • J.H. Doroshow

    Prevention of doxorubin-induced killing of MCF-7 human breast cancer cells by oxygen radical scavengers and iron chelating agents

    Biochem. Biophys. Res. Commun.

    (1986)
  • B.K. Sinha et al.

    Adriamycin-stimulated hydroxyl radical formation in human tumor cells

    Biochem. Pharmacol.

    (1987)
  • G. Batist et al.

    Overexpression of a novel anionic glutathione transferase in multidrug-resistant human breast cancer cells

    J. Biol. Chem.

    (1986)
  • W.S. Thayer

    Serum lipid peroxides in rats treated chronically with Adriamycin

    Biochem. Pharmacol.

    (1984)
  • R.A. Floyd et al.

    Use of salicylate with high pressure liquid chromatography and electrochemical detection (LCED) as a sensitive measure of hydroxyl radicals in Adriamycin treated rats

    J. Free-Rad. Biol. Med.

    (1986)
  • H. Nakano et al.

    Inhibition by the protein ceruloplasmin of lipid peroxidation stimulated by an Fe3+-ADP-Adriamycin complex

    FEBS Lett.

    (1984)
  • L. Gianni et al.

    Characterization of the cycle of iron-mediated electron transfer from Adriamycin to molecular oxygen

    J. Biol. Chem.

    (1985)
  • L. Gianni et al.

    Characterization of the cycle of iron-mediated electron transfer from Adriamycin to molecular oxygen

    J. Biol. Chem.

    (1985)
  • J.R.F. Muindi et al.

    Hydroxyl radical production and DNA damage by anthracycline-iron complex

    FEBS Lett.

    (1984)
  • C.E. Myers et al.

    5-Iminodaunomycin: an anthracycline with unique properties

    J. Biol. Chem.

    (1987)
  • E.J.F. Demant et al.

    Binding of transferrin-iron by Adriamycin at acidic pH

    FEBS Lett.

    (1986)
  • M. Gosalvez et al.

    Quelamycin, a new derivative of Adriamycin with several possible therapeutic advantages

    Eur. J. Cancer

    (1978)
  • P.L. Gutierrez et al.

    Free radicals in quinone-containing antitumor agents: the nature of the diaziquone [3,6-diaziridinyl-2,5-bis(carboethoxyamino)-1,4-benzoquinone] free radicals

    Biochim. Biophys. Acta

    (1983)
  • P.L. Gutierrez et al.

    Cellular activation of diaziquinone [3,6-diaziridinyl-2,5-bis(carboethoxyamino)-1,4-benzoquinone] to its free radical species

    Biochem. Pharmacol.

    (1985)
  • M.M. Mossoba et al.

    Diaziquonone as a potential agent for photoirradiation therapy: formation of the semiquinone and hydroxyl radicals by visible light

    Biochem. Biophys. Res. Commun.

    (1985)
  • B.K. Sinha et al.

    Irreversible binding of etoposide (VP-16-213) to deoxyribonucleic acid and proteins

    Biochem. Pharmacol.

    (1984)
  • B.K. Sinha et al.

    Microsomal interactions and inhibition of lipid peroxidation by etoposide (VP-16-213): implications for mode action

    Biochem. Pharmacol.

    (1985)
  • 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.

    (1987)
  • D. Ross et al.

    The reaction and subsequent fate of glutathionyl radical in biological systems

    J. Biol. Chem.

    (1985)
  • A.G. Katki et al.

    Interactions of the antitumor drug, etoposide with reduced thiols in vitro and in vivo

    Chem.-Biol. Interact.

    (1987)
  • B.K. Sinha et al.

    Iron-dependent hydroxyl radical formation and DNA damage from a novel metabolite of the clinically active antitumor drug VP-16

    FEBS Lett.

    (1988)
  • R.J. Weinkam et al.

    Metabolic activation of procarbazine

    Life Sci.

    (1978)
  • F.N. Dost et al.

    Methane formation in vivo from N-isopropyl-(2-methylhydrazino)-p-toluamide hydrochloride, a tumor-inhibiting methylhydrazine derivative

    Biochem. Pharmacol.

    (1967)
  • R.P. Mason

    Free radical intermediates in the metabolism of toxic chemicals

  • J.W. Lown

    Molecular mechanism of action of anticancer agents involving free radical intermediates

    Adv. Free-Rad. Biol. Med.

    (1985)
  • N.R. Bachur et al.

    A general mechanism for microsomal activation of quinone anticancer agents

    Cancer Res.

    (1978)
  • E.G. Mimnaugh et al.

    Adriamycin-enhanced lipid peroxidation in isolated rat nuclei

    Cancer Res.

    (1985)
  • C.A. Pritsos et al.

    Generation of reactive oxygen radicals through bioactivation of mitomycin antibiotics

    Cancer Res.

    (1986)
  • S.R. Keys et al.

    Role of NADPH-cytochrome c reductase and DT-diaphorase in the bioactivation of mitomycin C

    Cancer Res.

    (1984)
  • S. Pan et al.

    Reductive activation of mitomycin C and mitomycin C metabolites catalyzed by NADPH-cytochrome P-450 reductase and xanthine oxidase

    J. Biol. Chem.

    (1985)
  • J.W. Lown et al.

    Further studies on the generation of reactive oxygen species from activated anthracyclines and the relationship to cytotoxic action and cardiotoxic effects

    Biochem. Pharmacol.

    (1982)
  • M. Tomaz et al.

    The mode of interactions of mitomycin C with deoxyribonucleic acids and other polynucleotides in vitro

    Biochemistry

    (1974)
  • Y. Sugiura et al.

    Formation of superoxide and hdyroxyl radicals in iron (II)-bleomycin-oxygen systems: electron spin resonance detection by spin trapping

    J. Antiobiot. (Tokyo)

    (1978)
  • E.A. Sausville et al.

    Effects of chelating agents and metal ions on the degradation of DNA by bleomycin

    Biochemistry

    (1978)
  • H. Kuramochi et al.

    An active intermediate formed in the reaction of bleomycin-Fe (II) complex with oxygen

    J. Antibiot. (Tokyo)

    (1981)
  • B.K. Sinha

    Binding specificity of activated anthracycline anticancer agents to nucleic acids

    Chem.-Biol. Interact.

    (1980)
  • B.K. Sinha et al.

    Enzymatic activation and binding off Adriamycin to nuclear DNA

    Cancer Res.

    (1984)
  • Cited by (114)

    • Natural products as multidrug resistance modulators in cancer

      2019, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Beside antiapoptotic defense one more system that can hamper the activity of anticancer drugs is the antioxidant defense system, as most of these drugs induce free radical generation and this oxidative stress results in damage on the level of proteins, nucleic acids, and cell membranes, but some pre-existing systems in the body which normally protects the cells against these free radicals also protects the cancerous cells from the damage by these radicals. These systems include the cytochromes, superoxide dismutase, catalase, glutathione peroxidase, and antioxidants [4,14ā€“18]. As evident from the name the pump resistance is the one which depends on membrane-bound active efflux pumps.

    • Nitric oxide reverses drug resistance by inhibiting ATPase activity of p-glycoprotein in human multi-drug resistant cancer cells

      2018, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      While ADR is known to cause many biological/pharmacological effects in tumor cells, there are two main mechanisms that have been suggested to play important roles in tumor cell killing [37,40,43]. First, ADR induces both single- and double-strand breaks via acting as a topo II poison [27,41,42] and secondly, it generates reactive hydroxyl radicals following reductive activation at or near DNA, inducing DNA damage leading to tumor cell death [33ā€“35,45]. We have examined both mechanisms in the presence of PPNO as an explanation for the reversal of ADR resistance.

    • ROS signalling in the biology of cancer

      2018, Seminars in Cell and Developmental Biology
      Citation Excerpt :

      Daunorubicin, an anthracycline is used in the treatment of AML, acute lymphoblastic leukaemia (ALL) and acute promyelocytic leukaemia (APL). It reacts with cytochrome p450 reductase [242ā€“244] in the presence of reduced NADPH to form semiquinone radical intermediates [245,246] that react with O2 to form O2āˆ’ [245,247ā€“251]. This leads to increased activation of neutral sphingomyelinase enzyme and increased ceramide resulting in activation of the JNK pathway leading to apoptosis [252,253].

    • Nitric oxide inhibits topoisomerase II activity and induces resistance to topoisomerase II-poisons in human tumor cells

      2016, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      The most interesting and somewhat surprising finding is that PPNO treatment had no significant effect on DOX-induced cleavable complex formation nor did it have any significant effects on DOX cytotoxicity in these cells lines. While the mechanisms of DOX-induced tumor cell killing are extremely complicated and various mechanisms have been reported [16,54ā€“56], DOX is also considered to be a topo II poison. Under conditions where both the catalytic and relaxation activity of topo II were inhibited by PPNO, resulting in significant resistance to VP-16, no effect on DOX cytotoxicity was observed in either HT-29 or MCF-7 tumor cells.

    View all citing articles on Scopus
    View full text