Vous êtes sur la page 1sur 14

See

discussions, stats, and author profiles for this publication at:


https://www.researchgate.net/publication/20953130

Doxorubicin (adriamycin): A critical


review of free radical-dependent
mechanisms of cytotoxicity

Article in Pharmacology [?] Therapeutics February 1990


DOI: 10.1016/0163-7258(90)90088-J Source: PubMed

CITATIONS READS

353 417

4 authors:

Hiskias Keizer Herbert M Pinedo


Stepan Specialty Products VU University Medical Center
33 PUBLICATIONS 1,606 CITATIONS 958 PUBLICATIONS 32,604 CITATIONS

SEE PROFILE SEE PROFILE

Gerrit Jan Schuurhuis Hans Joenje


VU University Medical Center VU University Medical Center
223 PUBLICATIONS 5,356 CITATIONS 259 PUBLICATIONS 15,076 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Discrimination of healthy and leukemic stem cells in AML patients View project

All content following this page was uploaded by Gerrit Jan Schuurhuis on 07 November 2015.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
Pharmac. Ther. Vol. 47, pp. 219-231, 1990 0163-7258/90 $0.00 + 0.50
Printed in Great Britain. All rights reserved 1990 Pergamon Press plc

Associate Editor: D. SHUGAR

DOXORUBICIN (ADRIAMYCIN): A CRITICAL REVIEW OF


FREE RADICAL-DEPENDENT MECHANISMS OF
CYTOTOXICITY

H. G. KEIZER,*~ H. M. PINEDO,t G. J. SCHUURHUISt a n d H. JOENJE*


*Institute of Human Genetics and tDepartment of Oncology, Free University, P.O. Box 7161, 1007 MC
Amsterdam, The Netherlands

Abstract--The antineoplastic drug doxorubicin is capable of generating a variety of free radical species
in subcellular systems and this capacity has been considered critical for its antitumor action. However,
for most tumor cell lines this mechanism of cytotoxicity does not appear to play a major role. Free
radical-independent cytotoxicity mechanisms, taking place in the nuclear compartment of the cell, may
more likely be involved in the antitumor effect of doxorubicin.

CONTENTS
1. Introduction 219
2. Free Radical-Dependent Toxicity Mechanisms of Doxorubicin 220
2.1. Mechanism of free radical production due to redox-cycling 220
2.2. DNA damage due to redox-cycling 220
2.3. Lipid peroxidation induced by redox-cycling 221
2.4. Mechanism of free radical formation by the doxorubicin-iron complex 222
2.5. Doxorubicin-iron complex-mediated DNA damage 222
2.6. Doxorubicin-iron complex-mediated lipid peroxidation 223
3. The Relative Importance of Free Radicals in the Mechanism of Cytotoxicity of Doxorubicin 223
3.1. Free radicals generated by redox-cycling 223
3.2. Free radicals generated by the doxorubicin-iron complex 224
3.3. Doxorubicin-dependent hydroxyl radical formation by intact tumor cells 224
3.4. Effects of radical scavengers on the cytotoxicity of doxorubicin 225
3.5. Role of free radicals in doxorubicin-induced cardiotoxicity 226
3.5.1. Free radical chemistry of doxorubicin in cardiac tissue 226
3.5.2. Free radical damage in cardiac tissue 226
4. Free Radical-Independent Mechanisms of Cytotoxicity 227
5. Summary and Conclusions 228
Acknowledgements 228
References 228

1. I N T R O D U C T I O N by a cumulative dose-related cardiomyopathy that


manifests itself as congestive heart failure (Young
The anthracycline antibiotic doxorubicin (Adri- et al., 1981).
amycin), originally isolated from the fungus Much effort has been devoted to unraveling the
Streptomyces peucetius (Arcamone et al., 1972), is a mechanism of antitumor action of doxorubicin.
chemotherapeutic agent with strong activity against a Currently the belief is widespread that free radical
wide range of human malignant neoplasms including formation is critically involved in the mechanism
acute leukemia, non-Hodgkin lymphomas, breast of cytotoxicity of doxorubicin against tumor cells.
cancer, Hodgkin's disease and sarcomas (Young However, most of the evidence favoring a free
et al., 1981). Apart from side-effects that are common radical-dependent model has been obtained by study-
to many cancer chemotherapeutics, i.e. hematopoietic ing subcellular fractions, often in combination with
suppression, nausea and vomiting, and alopecia, the extremely high concentrations of doxorubicin that
clinical usefulness of doxorubicin is limited largely are not encountered in clinical practice.
Below we review the mechanism of free radical
++Present address: Department of Toxicology, Duphar formation from doxorubicin and discuss the likeli-
B.V., P.O. Box 900, 1380 DA Weesp, The Netherlands. hood of these radicals playing a role in its antitumor
To whom correspondence should be addressed. effect. We conclude that, in general, this role must be

219
220 H.G. KEIZERet al.

a minor one, even though doxorubicin-induced car- known to occur in the presence of iron (Gianni et al.,
diotoxicity may well be mediated by free radical 1985; Zweier, 1985) when no reducing system is
formation. Finally, an attractive alternative mecha- present. When the oxidized doxorubicin semi-
nism, based on the interference of doxorubicin with quinone-iron complex reacts with oxygen, this will
topoisomerase II, is discussed. ultimately lead to the formation of a fully oxidized
form of doxorubicin, with formation of oxygen
radicals. Both radical-producing systems are dis-
cussed below.
2. FREE RADICAL-DEPENDENT TOXICITY
MECHANISMS OF DOXORUBICIN
2.1. MECHANISMOF FREE RADICALPRODUCTIONDUE
The chemical structure of doxorubicin is depicted TO REDOX-CYCLING
in Fig. 1. From a 'free radical' point of view both
rings B and C are of special interest. One-electron- Reductive redox-cycling of doxorubicin was
reduction of ring C leads to the formation of a described by Goodman and Hochstein (1977), who
semiquinone free radical (Fig. 2). This radical is observed that cytochrome P-450 reductase causes
relatively stable under anoxic conditions, but under NADPH-dependent oxygen consumption in the
oxic conditions its unpaired electron is donated to presence of doxorubicin, in excess of the amount of
oxygen (02) forming superoxide radicals (Doroshow, drug present. Later studies indicated that not only
1983a, b; Bachur et al., 1982; Svingen and Powis, NADPH cytochrome P-450 reductase (Pan et al.,
1981). Suitable flavoproteins catalyze the formation 1981), but also NADH dehydrogenase (Doroshow,
of reduced semiquinone radicals by accepting electrons 1983b; Davies et al., 1983a, b) and xanthine oxidase
from NADH or NADPH and donating them to (Doroshow, 1983a; Pan and Bachur, 1980), catalyze
doxorubicin. By reducing oxygen to superoxide the the one-electron-reduction of doxorubicin. As a con-
parental doxorubicin molecule is regenerated. This sequence, redox-cycling of doxorubicin can occur in
sequence of reactions, known as 'redox-cycling', is cytoplasm, mitochondria, endoplasmic reticulum
potentially harmful to cells since relatively little (Doroshow, 1983a), and nucleus (Bachur et al., 1982).
doxorubicin would suffice to catalyze the formation The nucleus-catalyzed redox-cycling led Bachur et al.
of numerous superoxide radicals. Formation of an (1978) to postulate the concept of site-specific free
oxidized semiquinone in ring B of doxorubicin is radical formation in the nucleus: doxorubicin binds
selectively to nuclear DNA and can be metabolically
activated in the nucleus to produce free radical-
o OH o
1 ~t2 11 to 13 mediated damage to DNA.
The possible importance of redox-cycling is
stressed by the observation that not only isolated
] 4 I] = I * ' %, _." enzymes, but also intact cells, can support one-
H3CO O OH O| .;~c., electron-reduction of doxorubicin and subsequent
Y' doxorubicin-induced oxygen consumption (Sato
et al., 1977). Furthermore, one-electron-reduction of
2,'N'~ Rg 3
doxorubicin might also occur in the clinical situation,
since an important metabolite found in patients is
Anthracydine R1 R2 R3 R4 R5 the C7-deoxy-aglycone form of doxorubicin (Mross
et al., 1988), which is a metabolite known to be
Adciamycin CH2OH NH 2 OH H H
formed from doxorubicin semiquinone radicals under
Oaun~ubicin CH 3 NH 2 OH H H anoxic conditions (Guti6rrez et al., 1983; Pan et al.,
1981). In short, doxorubicin may be involved in a
'redox-cycling' process, which is potentially impor-
tant for its cytotoxicity.
FIG. I. The chemical structure of doxorubicin. Chemically
doxorubicin (adriamycin) consists of a water-insoluble te-
tracycline aglycone (structures ABCD) and a water-soluble
reducing daunosamine sugar. Several parts of the molecule 2.2. DNA DAMAGEDUE TO REDOx-CYCLING
can be involved in the metabolism of doxorubicin. Ring C Redox-cycling is rapid in the presence of oxygen
is a quinone group which can be activated into a
semiquinone radical after one-electron-reduction. Ring B is and slow under hypoxic conditions. Thus, in the
a hydroquinone which can also be activated into a presence of oxygen, large amounts of superoxide may
semiquinone radical after one-electron-oxidation. The C9 be formed, whereas in its absence the semiquinone
side chain is probably important in the iron-catalyzed radical will accumulate. As indicated below, different
autoxidation of the molecule. Differences in the C9 side toxicity mechanisms might be operative, depending
chain form the only difference between doxorubicin and its on the amount of oxygen present.
analog daunorubicin. The iron-chelation site is probably An oxygen-dependent mechanism of DNA damage
formed by the oxygen atoms of CI1 and C12. Iron at this has been described by Berlin and Haseltine (1981),
site can be reduced by the doxorubicin molecule either by
who observed extensive DNA strand scission in a
oxidation of the hydroquinone or by oxidation of the C9
side chain. The sugar group is important for the water system containing NADPH, NADPH cytochrome
solubility of doxorubicin; during anaerobic metabolic reduc- P-450 reductase and DNA, and by Gutteridge and
tion of doxorubicin it is split off and the C7-deoxyaglycone Toeg (1982), who found deoxyribose breakdown in a
is formed. From Gianni et al. (1988). (Reproduced with system containing deoxyribose, doxorubicin semi-
permission of the authors.) quinone radicals and iron. The authors proposed that
Doxorubicin (Adriamycin) 221

_ 9" 9" o. on
Orug-macromolecule DNA,RNA~ * ~ ~ C O R t~;rJ~*,~COR
complexes ' Proteins C H ~ " O H 4 ~ ~ , ~ " O H
so o- o. c's0 6 0H
(CT-Ouinone methide) T (C7-Deoxyaglycone)
/ DNA- nicking

0 OH DNA /
~ C O R /f
\ f \ / - ' ,~j r " o ,
y F~I( %0 o o. 6-s,or ~ ( - - / ~L,p,~,
.,o,.J\, J qo. y\ .,o,00.,o,,o.,,o.
~ "OH ~ "~- 02
CH30 O" OH C)-sugor
(Semiquinone)

Oruo-mocromolecule DNA,RNA ~ j ~ z ~ , , ~ COR


complexes * Proteins ~ L ~ ~ OH
CHso 0 OH
CT-Free rodicol

FIG. 2. Free radical and alkylating intermediates in the metabolism of doxorubicin. Doxorubicin can be
reduced by cellular flavoproteins to form its one-electron reduced semiquinone form. In the presence of
oxygen the semiquinone free radical is oxidized back into the parental quinone form under formation of
superoxide radicals. In the absence of oxygen the semiquinone is unstable; it loses its sugar moiety and
an intermediate C7 free radical can be formed. This radical can bind covalently to cellular macromolecules
or become again reduced forming a relatively stable product, the C7-deoxyaglycone,which is in fact a
doubly reduced doxorubicin molecule that has lost its sugar moiety. A tautomer of C7-deoxyaglyconeis
the C7-quinone-methide, which is a potent DNA alkylating species and potentially toxic for tumor cells.
From Sinha et al. (1984). Reproduced with permission of the authors and the copyright holder, Cancer
Research, Inc., Philadelphia.

the DNA damaging species is the hydroxyl radical Another DNA-damaging mechanism that may
formed from the following sequence of reactions: operate at low partial pressure o f oxygen has been
described by Winterbourn and collaborators. They
202- + 2H+ ~H202 + 02
showed that in the absence of oxygen the semi-
02 + F e 3 + ~ O : + Fe 2+ quinone of doxorubicin reacts with H202 causing
breakdown of deoxyribose (Bates and Winterbourn,
H202 + Fe 2+ --*'OH + O H - + Fe 3+.
1982). This reaction was catalyzed by very low con-
The sum of the latter two reactions centrations of iron: submicromolar concentrations
are sufficient for a maximal effect (Winterbourn et al.,
H202 + O~- ~ ' O H + O H - + 02
1985). Both the low iron requirement of the reaction
is known as the iron-catalyzed Haber-Weiss reaction. and the optimal reaction rate at an oxygen tension of
That superoxide-generating systems can indeed approximately 4 mm Hg, may mimic the situation in
damage DNA by this series of reactions was shown a tumor cell, since almost all intracellular iron is
by Rowley and Halliwell (1983), who were able to believed to be in a complexed form, while oxygen
inhibit DNA damage in a superoxide-producing tensions of 3-6 mm Hg are considered to be common
system with superoxide dismutase, catalase, iron (Winterbourn et al., 1985). These data underscore the
chelators and hydroxyl radical scavengers. potential importance of this reaction.
In the absence o f oxygen the semiquinone is
converted to its C7-deoxyaglycone metabolite, which
2.3. LIPID PEROXIDATION INDUCED BY
in itself is not pharmacologically active (Averbuch
REDOX-CYCLING
et al., 1985). However, its tautomer, the C7-quinone
methide is a DNA-alkylating species (Moore, 1977). Unsaturated fatty acids are peroxidized when
Furthermore, an intermediate in the formation of exposed to hydroxyl radicals in the presence of a
C7-deoxyaglycone from the doxorubicin semi- metal catalyst (Gutteridge, 1982). This process might
quinone is the C7 free radical, which can also bind damage cellular membranes and eventually kill cells.
covalently to DNA (Sinha and Chignell, 1979; Sinha Doxorubicin can cause lipid peroxidation after
and Lewis Gregory, 1981; Sinha et al., 1984). The metabolic activation by heart, liver or kidney micro-
structures of these reactive species are depicted in somes (Glazer et al., 1982; Mimnaugh et al., 1982;
Fig. 2. Bachur et al., 1977, 1978), mitochondria (Mimnaugh
222 H.G. KEIZERet al.

et al., 1985b), or nuclei (Mimnaugh et al., 1985a).


In the presence of oxygen, doxorubicin-dependent
lipid peroxidation is generally inhibited by superoxide NADP~-.~ 1.,- DOX-Fe ~* ,,~...,-.~GSSG

dismutase, catalase, glutathione, iron chelators


and hydroxyl radical scavengers (Mimnaugh et al.,
1985a, b, 1983), suggesting that OH radicals gener- NADPH - ' / ~'- DOX.Fe ~+ GSH

ated by the iron-catalyzed Haber-Weiss reaction are


involved. The rate of lipid peroxidation in the pres- a+
ence of iron and superoxide, produced by reduced I:~X-Fe
anthracyclines, was much higher for derivatives of
doxorubicin that bind to microsomes than for non-
binding derivatives (Sterrenberg et al., 1985). Doll,g"
Analogous to D N A damage, membrane lipid per-
oxidation can also be induced in the absence of
OH ~,"~
oxygen with H202 present (Winterbourn et al., 1985).
Without H:O2 very low concentrations of oxygen are oox.~=g
required for optimal lipid peroxidation, probably to
provide H202 from the dismutation of superoxide,
which is formed by the reaction of 02 with doxoru-
[
9-COOH-DOX
bicin semiquinone radicals. Lipid peroxidation
occurred without added iron, but increased when iron
was added in the 0.1-1pM range. OH radical FIG. 3. Free radical production from the doxorubicin iron
scavengers did not inhibit lipid peroxidation in this complex. The doxorubicin-iron II complex (DOX-Fe 2+)
system. This is probably due to binding of doxoru- can react with molecular oxygen (O2) or hydrogen peroxide
bicin to the liposomes used in this test, since the (H202) leading to the formation of superoxide (O~) or
reactivity of "OH is so high that, when generated in hydroxyl radicals ('OH), respectively, while the complex is
a 'site-specific' way, competition by scavengers is very oxidized to doxorubicin-iron III (DOX-Fe3+). DOX-Fe 3+
inefficient (Winterbourn et al., 1985). can be reduced enzymatically by cytochrome P-450 reduc-
tase (Fp) or nonenzymatically via reaction with reduced
In conclusion, under both hypoxic and normoxic glutathione (GSH), forming back DOX Fe2+, which can
conditions, lipid peroxidation can be induced by the again react with 02 or H202. In the presence of a reducing
reduced doxorubicin semiquinone radical, which in system no metabolites are formed and radical production
both cases is probably mediated by the OH radical. can proceed indefinitely. In the absence of a reducing
system, DOX-Fe 3+ can reduce its iron intramolecularly by
oxidizing its hydroquinone moiety leading to the formation
2.4. MECHANISM OF FREE RADICAL FORMATION BY
of an oxidized doxorubicin semiquinone radical or by
THE DOXORUBICIN-IRON COMPLEX oxidizing its C9 side chain. This intramolecular reduction of
The doxorubicin-iron complex can support the iron is probably catalyzed by DNA. Electrons from the C9
chain of doxorubicin can keep the iron of the complex
formation of free radicals by two distinct mecha- reduced and thus support free radical production until the
nisms, one dependent on the presence of a reducing side chain is fully oxidized. The end product of such
system, and another forming radicals from the oxidation is 9-COOH-doxorubicin.
doxorubicin-iron complex itself, in the absence of a
reducing system. Both mechanisms are schematically doxorubicin free radical chelate with Fe 2+ (Gianni
depicted in Fig. 3. et al., 1985; Zweier et al., 1986; Zweier, 1985). The
The upper part of Fig. 3 illustrates free radical formation of this doxorubicin-Fe 2+ complex is
formation in the presence of a reducing system. probably catalyzed by D N A (Muindi et al., 1985). In
Doxorubicin-Fe 3 is reduced to doxorubicin-Fe 2+ the presence of oxygen this complex is oxidized, with
either enzymatically by N A D H cytochrome P-450 formation of superoxide radicals (Gianni et al., 1985),
reductase (Sugioka and Nakano, 1982) or by thiols while it can also react with H202 via formation of
like glutathione (GSH) or cysteine (Zweier, 1985). hydroxyl radicals (Zweier, 1985; Muindi et al., 1984,
Doxorubicin-Fe 2 can react with oxygen to form 1985). Further oxidation of the side chain will
superoxide radicals (O;-) which dismutate to form ultimately lead to the formation of the oxidized meta-
hydrogen peroxide (H202). Hydrogen peroxide can bolite of doxorubicin 9-dehydroxyacetyl-9-carboxyl
also react with doxorubicin-Fe 2 and form hydroxyl doxorubicin (9-COOH-doxorubicin) (Gianni et al.,
radicals (Eliot et aL, 1984; Myers et al., 1982). This 1988) (see Fig. 3).
GSH-driven hydroxyl radical formation is a cyclic
process comparable to redox-cycling in that no
doxorubicin metabolites are being formed (Zweier, 2.5. DOXORUBICIN--IRON COMPLEX-MEDIATED D N A
1985; Myers et aL, 1982) and, in the presence of DAMAGE
sufficient GSH, the cycling process can proceed in-
definitely. Doxorubicin-iron complexes bind tightly to D N A
In the absence of a reducing system, doxoru- (Eliot et aL, 1984). However, contrary to intercalated
bicin-Fe 3+ can reduce its chelated iron by an intra- doxorubicin (Mimnaugh et al., 1985b; Sinha and
molecular redox reaction, either by oxidation of the Chignell, 1979; Sato et al., 1977; Berlin and Haseltine,
side chain on C9 (Zweier et aL, 1986; Gianni et al., 1981; Rowley and Halliwell, 1983), the doxoru-
1988; see also Fig. l) or the hydroquinone moiety at bicin-iron complex preserves its ability to catalyze the
ring B (Zweier, 1983; Gianni et al., 1985), forming a formation of oxygen free radicals in the presence of
Doxorubicin (Adriamycin) 223

double-stranded DNA (Eliot et aL, 1984). Thus, the might be different, as in some cell types more than
doxorubicin-iron complex-driven hydroxyl radical 99.8% of the doxorubicin is present in a DNA-bound
formation can proceed in close proximity to DNA form (Gigli et al., 1988), whereas binding to double
and has therefore the potential to damage DNA stranded DNA is known to preclude chemical (Sinha
efficiently, especially since DNA seems to catalyze and Chignell, 1979), enzymatic (Berlin and Haseltine,
hydroxyl radical formation by this complex (Muindi 1981; Rowley and Halliwell, 1983), and microsomal
et al., 1984, 1985). Hydroxyl radicals are probably (Sato et al., 1977) one-electron-reduction of doxoru-
involved in damaging of DNA since the generation of bicin. As a consequence of this, redox-cycling-depen-
hydroxyl radicals by the Dox-Fe 3 complex corre- dent DNA strand scission (Berlin and Haseltine,
lates with its ability to cleave DNA (Muindi et al., 1981), lipid peroxidation (Mimnaugh et al., 1985a)
1984) and also since catalase, iron chelators and and generation of superoxide from doxorubicin by
hydroxyl radical scavengers are protective in this microsomes (Kalyanaraman et al., 1980) is largely
system (Eliot et al., 1984). Relatively high concentra- prevented in the presence of double-stranded DNA.
tions of hydroxyl radical scavengers were required for That Bachur et al. (1982) nevertheless found redox
protection, indicating that these radicals were indeed activity of doxorubicin in intact nuclei may be due to
generated in a site-specific way. the extremely high concentrations of doxorubicin
( > 500 11M) used.
2.6. DOXORUBICIN-IRONCOMPLEX-MEDIATEDLIPID Even though radical formation by redox-cycling
PEROXIDATION may not take place in the nucleus, semiquinone or
oxygen radicals may well be formed at some cytoplas-
In the presence of GSH, doxorubicin efficiently mic site, e.g. the endoplasmic reticulum (Bachur
damages erythrocyte ghost membranes (Myers et al., et al., 1978), and reach the nucleus by diffusion.
1982). This reaction was inhibited by superoxide However, the semiquinone radical is relatively un-
dismutase, catalase and hydroxyl radical scavengers, stable, with an estimated diffusion radius of 0.6 #m
indicating the involvement of the hydroxyl radical. under anaerobic and 0.1 # m under aerobic conditions
Even in the absence of reducing agents, doxoru- (Svingen and Powis, 1981), while an average cell has
bicin-iron complexes appear to have the potential to a diameter in the range of 5-20/~m. This suggests that
damage biomembranes, since such complexes pro- only a minor fraction of the semiquinone radicals
voke lipid peroxidation (Sugioka and Nakano, 1982; produced in the cytoplasm may be able to reach
Gutteridge, 1982, 1983, 1984; Gianni et al., 1988). the nucleus. Theoretically, the probability for
In this case, however, superoxide dismutase, catalase semiquinone radicals to reach the nucleus is six-fold
or hydroxyl radical scavengers did not protect higher under anoxic than under aerobic conditions.
(Sugioka and Nakano, 1982; Gutteridge, 1983; Yet, the sensitivity of cells to doxorubicin is essen-
Gianni et al., 1988). The reduction of iron by the tially similar under both anoxic and aerobic condi-
side chain of doxorubicin was probably involved in tions (Kennedy et al., 1983), which seems to argue
this type of membrane damage, since daunorubicin, against involvement of semiquinone formation in the
a structural analog of doxorubicin, lacking a readily cytotoxicity of doxorubicin.
oxidizable side chain, was much less efficient (Gianni Alternatively, superoxide and hydrogen peroxide
et al., 1988). Thus, both in the absence and presence formed in the cytoplasm by redox-cycling might
of reducing agents, the doxorubicin-iron complex diffuse into the nucleus and damage the DNA.
can damage membranes, probably due to a hydroxyl However, such a mechanism seems unlikely, since
radical-dependent mechanism. increasing the concentration of intracellular SOD and
catalase, either artificially (Cervantes et al., 1988) or
by selection for resistance to hyperoxia (Keizer et al.,
3. THE RELATIVE IMPORTANCE OF FREE 1988), has been shown not to protect against the
RADICALS IN THE MECHANISM OF cytotoxicity of doxorubicin. Furthermore, a recent
CYTOTOXICITY OF DOXORUBICIN spin-trapping study failed to provide evidence for an
involvement of ESR-observable oxygen-derived radi-
As summarized in the previous section, roughly cals in the antitumor activity of doxorubicin (Alegria
two mechanisms of doxorubicin-mediated free radi- et al., 1989).
cal formation can be distinguished, one dependent on In addition to DNA damage, free radical damage
the formation of semiquinone radicals generated to cellular membranes by lipid peroxidation may also
during flavoprotein-mediated redox-cycling, and the be involved in the antitumor effect of doxorubicin. In
other dependent on the doxorubicin-iron complex. fact, doxorubicin-induced lipid peroxidaton has been
Below we discuss the likelihood of such mechanisms observed in hepatocytes (Meredith and Reed, 1983;
playing a role in the tumoricidal and cardiotoxic Babson et al., 1981) and cardiocytes (Julicher et al.,
effects of doxorubicin. 1985). However, to observe this effect, relatively high
concentrations of doxorubicin (100pM) were re-
3.1. FREE RADICALSGENERATEDBY REDOX-CYCLING quired; in addition, the experiments were done with
glutathione-depleted cells under an atmosphere of
With respect to redox-cycling, Bachur et al. (1978, pure oxygen, conditions known to accelerate the
1982) have suggested that free radical formation can process of lipid peroxidation. Bearing in mind that
occur site-specifically in the nucleus, since isolated hepatocytes and cardiocytes are probably better
nuclei bind doxorubicin and can also support suited to support redox-cycling of doxorubicin, com-
semiquinone formation from doxorubicin (Bachur pared to tumor cells, such data can not be taken to
et al., 1982). However, the situation in intact cells indicate that lipid peroxidation is involved in the
224 H.G. KEIZERet al.

antitumor effect of doxorubicin, especially since lipid in intact cells under clinically relevant conditions
peroxidation has not been reported for tumor (Gelvan and Samuni, 1988).
cells treated with clinically relevant concentrations of
doxorubicin. 3.3. DOXORUBICIN-DEPENDENT HYDROXYL RADICAL
To investigate the role of flavoprotein-mediated FORMATION BY INTACT TUMOR CELLS
redox-cycling of doxorubicin in its cytotoxicity
against SW-1573 human lung tumor cells, Keizer As appears from the data summarized in Section
et al. (1989a) made use of a set of inhibitors of 2.1, in all models for doxorubicin-induced free radical
flavoprotein-mediated reduction of doxorubicin and damage, the ultimate damaging species is presumed
observed that none of these agents protected against to be the hydroxyl radical. Therefore several groups
the cytotoxicity of doxorubicin, although the cyto- of investigators have attempted to detect 'OH
toxicity of mitomycin C, which is known to depend formation in intact tumor cells after treatment with
on ftavoprotein activity, was strongly inhibited. doxorubicin.
From the evidence presented above we must con- Using spin-trapping, Sinha et al. (1987a, b) de-
clude that the situation inside the intact tumor cell is tected hydroxyl radical formation in MCF7 human
not favorable for redox-cycling, and that the involve- breast cancer cells; however, the doxorubicin concen-
ment of redox-cycling in the cytotoxicity of doxoru- trations used were three orders of magnitude higher
bicin in tumor cells has not been proven. than that needed to cause 50% inhibition of
In the next section we discuss the probability that growth (IC50). Furthermore, in a doxorubicin-
doxorubicin-iron complex-dependent free radical resistant variant, hydroxyl radical formation was still
formation is involved in the mechanism of cytotoxic- undetectable, even at concentrations that were 100-
ity of doxorubicin. fold higher than the IC50 value for this variant. This
indicates that hydroxyl radicals measured under such
3.2. FREE RADICALS GENERATED BY THE experimental conditions are not necessarily involved
DOXORUBICIN-IRON COMPLEX in the cytotoxic effect of doxorubicin. The observa-
tions that extracellularly added NADPH strongly
T h i o l - i n d e p e n d e n t free radical formation is very stimulated hydroxyl radical formation, whereas both
unlikely to occur in intact cells, since intracellular superoxide dismutase and catalase were inhibitory,
thiols are known to be abundantly present, which is suggest that at least part of the hydroxyl radicals were
expected to prevent autoxidation of doxorubicin generated outside the cells (Sinha et al., 1987a, b; cf.
(Zweier, 1985). In addition the 9-COOH-doxorubicin Alegria et al., 1989). It follows that enzymes at the
metabolite is not detected in patients treated with outer cell surface might reduce doxorubicin, thus
doxorubicin, Although tumor cells in vivo were not leading to the formation of superoxide radicals and
protected by extracellularly added thiols (Freeman et hydrogen peroxide through redox-cycling. Since iron
al., 1980; Doroshow et al., 1981; Yoda et al., 1986), scavengers inhibited hydroxyl radical formation
it cannot be excluded that this mechanism of free (Sinha et al., 1987a, b) an iron-catalyzed Haber
radical formation occurs in extracellular fluids, where Weiss reaction might thus be responsible for hydroxyl
glutathione levels are relatively low. radical formation in this system; alternatively extra-
Thiol-dependent free radical formation from cellular reduction of doxorubicin-iron complexes
doxorubicin-iron complexes is a more attractive might produce these radicals.
model for hydroxyl radical-mediated D N A damage, Using methane formation from DMSO as an assay
since doxorobucin-iron complexes bind efficiently to for hydroxyl radical formation, Doroshow (1986b)
DNA, where they can still be reduced by thiols and obtained essentially similar data for intact Ehrlich
produce hydroxyl radicals in a site-specific way at ascites tumor cells. However, since catalase com-
target. However, thiol depletion does not protect pletely inhibited hydroxyl radical formation,
tumor cells against the cytotoxicity of doxorubicin Doroshow's results indicate that the hydroxyl radi-
(Ramu et al., 1984; Hamilton et al., 1985), nor does cals detected were generated outside the cells.
an increased thiol content potentiate its cytotoxicity Hydroxyl radicals generated at the outer surface of
(Doroshow et al., 1981). The only system described cells might play a role in the mechanism of tumor cell
thus far, in which thiols increase doxorubicin- killing by immobilized doxorubicin. In this case
mediated damage, is the microsomal fraction doxorubicin is complexed to a synthetic carrier, so
from kidney, in which thiols potentiate lipid peroxi- that the drug cannot enter the cells; nevertheless such
dation (Mimnaugh, 1986). Thus kidney damage due a complex is highly cytotoxic (Tritton and Yee, 1982;
to doxorubicin may be partially caused by thiol- Rogers and T6k~s, 1984; Panneerselvam et al., 1987;
dependent free radical formation (Mimnaugh, 1986; Rogers et al., 1983). At sites where the cells make
Mimnaugh et al., 1986). For tumor cell killing, contact with the immobilized doxorubicin, the local
however, the occurrence of this mechanism remains doxorubicin concentrations may be high enough to
to be proven. support hydroxyl radical formation after reduction of
In conclusion, of all free radical-dependent mecha- doxorubicin by cell surface-bound reducing enzymes.
nisms, thiol-dependent free radical formation from That redox activity outside the cells, as well as
the doxorubicin-iron complex seems to be the most hydroxyl radical formation, is necessary for the cyto-
attractive model for tumor cell killing by doxoru- toxicity of immobilized doxorubicin, is suggested by
bicin, even though it is far from certain whether such the observations that hydroxyl radical scavengers
a mechanism actually takes place in the intact tumor together with superoxide dismutase and catalase
cell, since doxorubicin-iron complexes have not been inhibited cytotoxicity (Panneerselvam et al., 1987).
demonstrated in intact cells, and may not even exist Furthermore, immobilized iminodaunorubicin, a
Doxorubicin (Adriamycin) 225

derivative of doxorubicin that is less readily reduced sal mechanism of cytotoxicity. Furthermore, murine
metabolically, was much less cytotoxic than immobi- S180 cells were not protected against doxorubicin by
lized doxorubicin (Panneerselvam et al., 1987). Such the iron chelating drug ICRF 187 (Wadler et al.,
data underline the possibility that under certain 1986). An important question now is which cell
conditions hydroxyl radicals formed outside the cells model is representative for the most common mecha-
can kill tumor cells. However, this mechanism of nism of tumor cell killing by doxorubicin.
cytotoxicity may be quite distinct from the usual In this context it is important to note that both
toxicity mechanism operating against tumor cells, Ehrlich ascites cells and MCF7 cells are exceptional
as is further suggested by the observation that a with respect to the level of their antioxidant de-
pharmacologically inactive analog, 4-demethoxy-7,9- fense capacity. Ehrlich ascites cells are known to be
di-epi-daunorubicin, after coupling to a macromolec- catalase-deficient and to have very low levels of
ular carrier, acquired significant cytostatic activity to glutathione-dependent free radical-detoxifying en-
both doxorubicin-resistant and -sensitive L1210 cells zymes (Bozzi et al., 1981), while MCF7 cells contain
(Rodgers and T6k6s, 1984). unusually high levels of doxorubicin-reducing en-
Although OH radical formation has only been zymes (Sinha et al., 1987b), along with a relatively
detected at relatively high doxorubicin concentra- low glutathione-S-transferase activity (Broxterman et
tions at the outer cell surface, we cannot entirely al., 1989), an important peroxide detoxification en-
rule out the possibility that OH radicals are also zyme. That endogenous antioxidant levels in MCF7
formed inside the cells at clinically achievable concen- and Ehrlich ascites cells may indeed be limiting for
trations of doxorubicin, since OH radicals formed cellular tolerance to doxorubicin is suggested by the
inside the cell might react with cellular constituents fact that PZ51, an agent which can be taken up by
too rapidly to be detected by the relatively insensitive cells and function as an intracellular peroxidase in the
competitive detection methods that are based on presence of glutathione, protects both MCF7 and
spin-trapping and reaction with DMSO. To investi- Ehrlich ascites cells against the cytotoxicity of dox-
gate the role of OH radical formation in the cyto- orubicin (Doroshow, 1986a, b). The observation that
toxicity of doxorubicin at clinically achievable doxorubicin-resistant variants of MCF7 cells contain
concentrations, the effect of several free radical scav- increased levels of glutathione-S-transferase and glu-
engers on the cytotoxicity of doxorubicin has been tathione peroxidase (Kramer et al., 1988) further
studied. The findings from these studies are reviewed supports this concept. In several other cell lines
in the next section. (Ramu et al., 1984; Meijer et al., 1987; Ross et al.,
1988; Keizer et al., 1989b), the level of antioxidant
defense does not increase during selection for doxoru-
3.4. EFFECTSOF RADICALSCAVENGERSON THE bicin resistance, which strongly suggests that in these
cell lines the antioxidant defense does not limit
CYTOTOXICITYOF DOXORUBICIN
cellular tolerance to doxorubicin treatment. The same
As indicated in Table 1, extracellularly added was concluded for Chinese hamster ovary cells, since
superoxide dismutase or catalase protected MCF7 an oxygen-resistant variant having increased levels of
(Sinha et al., 1987b; Doroshow, 1986a) and Ehrlich superoxide dismutases, catalase, glutathione and glu-
ascites tumor (Doroshow, 1986b) cells against the tathione peroxidase was as sensitive to doxorubicin as
cytotoxicity of doxorubicin. However, no protection the parental CHO line (Keizer et al., 1988).
by superoxide dismutase and catalase was observed in In this respect an important observation was made
A2780 and A2780 AD cells (Cervantes et al., 1988). by Potmesil et al. (1984), who observed that doxoru-
Apparently, since superoxide dismutase and catalase bicin causes two types of DNA strand breaks in
are presumed not to enter cells, MCF7 and Ehrlich L1210 cells, i.e. protein-associated single-strand
ascites cells are killed to some extent by activated breaks and direct single-strand breaks. Up to a
oxygen species generated outside the cells by a mech- doxorubicin concentration of 2.8 pM, which kills
anism that is probably dependent upon iron, since more than 99.99% of the cells (Ross and Smith,
iron chelators were also protective, (Ooroshow, 1982), only protein-associated strand breaks were
1986a, b; see also Alegria et al., 1989). That A2780 observed (Potmesil et al., 1983). At higher concentra-
cells are not killed by a similar mechanism, as sug- tions protein-associated strand breakage decreased,
gested from the lack of effect of superoxide dismutase whereas direct strand breakage increased (Potmesil
and catalase, indicates that this may not be a univer- et al., 1983). Interestingly, direct strand breakage

TABLE 1. Effect o f Free Radical Scavengers on the Cytotoxicity o f Doxorubicin


Cell growth in the presence of doxorubicin (%)
Cell type Control + SOD + Catalase 'OH scavengers + PZ 51 Reference
Ehrlich acites 51 72 75 87-104 94 Doroshow(1986b)
MCF 7 49 93 87 100 74 Doroshow(1986a)
MCF 7 40 80 60 ND ND Sinha et al. (1987b)
A 2780 31 28 31 28 70 ND Cervanteset al. (1988)
A 2780 AD 43-46 46 42 47-80 ND Cervanteset al. (1988)
Effect of superoxide dismutase (SOD), catalase, several OH radical scavengers and a synthetic glutathione peroxidase
(PZ 51) on the cytotoxicity of doxorubicin, as reported in the literature. Cells were treated with doxorubicin concentrations
leading to a relative growth of 31-51% compared to untreated controls. The relative growth in the presence of antioxidant
is indicated. ND, not determined.

JPT 47/2--F
226 H.G. KEIZERet al.

occurred only in the presence of oxygen (Potmesil 3.5. ROLE OF FREE RADICALSIN
et al., 1983) and was inhibited by extracellularly DOXORUBICIN-INDUCED CARDIOTOXICITY
added superoxide dismutase or catalase (Potmesil
et al., 1984). These results clearly indicate that free Free radical production in cardiac cells due to
radicals generated at the cell surface have the potency one-electron-reduction of doxorubicin might occur at
to damage the cellular DNA. However, since lethality the nuclear envelope (Bachur et al., 1982), in mito-
of L1210 cells was almost 100% without occurrence chondria (NADH dehydrogenase), cytosol (xanthine
of this type of D N A damage, at doxorubicin concen- oxidase) or sarcoplasmic reticulum (NADPH cyto-
trations which even exceed the clinically achievable chrome P-450 reductase) (Doroshow, 1983a). In this
concentrations in plasma (Potmesil et al., 1984; respect cardiac cells are not exceptional. Neverthe-
Erttmann et al., 1988), a free radical-dependent less, the occurrence of cardiotoxicity is an important
mechanism of toxicity is apparently not very impor- dose-limiting factor in doxorubicin treatment of
tant in the killing of L1210 cells by doxorubicin. cancer patients (Green et al., 1984). The selective
Nevertheless, for cell lines with a relative lack of toxicity of doxorubicin to heart cells might simply
antioxidant enzymes, like Ehrlich ascites and MCF7 reflect an unusually high level of drug accumulation
cells, this type of damage might play a more impor- in these ceils, as reported by several authors
tant role in the cytotoxicity of doxorubicin. (Lampidis et al., 1981; Johnson et al., 1986). In
A rather consistent finding is that scavengers of addition, as discussed below, free radical damage
hydroxyl radicals protect cells against the cytotoxicity might be important for the development of
of doxorubicin (Table 1). However, protection is cardiotoxicity.
typically found only at extremely high scavenger
concentrations, often in the range between 100 and 3.5.1. Free Radical C h e m & t r y o f Doxorubicin in
250 mM (Doroshow, 1986a, b; Cervantes et al., 1988). Cardiac Tissue
Of course, only high "OH scavenger concentrations
are expected to be able to protect against hydroxyl Heart muscle cells are extremely rich in mitochon-
radical damage in view of the extreme reactivity of dria, which might render them particularly vulnerable
this radical. However, in the same concentration to free radicals generated at these organelles. In
range, sodium chloride also protected against the addition, unlike liver microsomes, where doxorubicin
cytotoxicity of doxorubicin (Iliakis and Lazar, 1987). semiquinone radicals react preferentially with molec-
Therefore, there are serious doubts about the specifi- ular oxygen to form relatively harmless superoxide
city of the hydroxyl radical scavengers when used at radicals, semiquinones formed in heart mitochondria
such high concentrations. Thus, although the effects appear to react rather with hydrogen peroxide with
of hydroxyl radical scavengers are consistent with a formation of the highly reactive hydroxyl radical
role for these radicals in the cytotoxicity of doxoru- (Nohl and Jordan, 1983). Compared to liver micro-
bicin, the critical involvement of hydroxyl radicals somes, sarcosomes from heart tissue are relatively
remains unproven. inefficient in reducing doxorubicin to its semiquinone
Not only in vitro but also in vivo effects of radical form (Nohl and Jordan, 1983), probably due to a
scavengers have been investigated on the tumoricidal relative lack of N A D P H cytochrome P-450 reduc-
effect of doxorubicin. Ascorbate (Fujita et al., 1982), tase. However, semiquinones generated at this site
N-acetylcysteine (Freeman et al., 1980; Doroshow et also tend to react preferentially with hydrogen per-
al., 1981 ), cysteine (Freeman et al., 1980), glutathione oxide, even in the presence of oxygen (Thornalley and
(Yoda et al., 1986) and alpha-tocopherol (Myers Dodd, 1985), while in tumor cells this reaction takes
et al., 1977) have been tested in vivo. None of these place only in the absence of oxygen (Bates and
treatments reduced the tumoricidal effect of doxoru- Winterbourn, 1982).
bicin against L1210 (Fujita et al., 1982; Yoda et al., Although intracellular hydroxyl radical forma-
1986), P388 (Freeman et al., 1980; Myers et al., 1977) tion in cardiac tissue could not be demonstrated
or Ehrlich ascites cells (Freeman et al., 1980), while (Rajagopalan et al., 1988), the fact that microscopi-
these treatments did reduce cardiotoxicity in the cally visible cardiac damage starts to occur at the
animals under investigation. Since free radical endoplasmic reticulum and mitochondria (Olson and
damage is likely to be responsible for cardiotoxicity Capen, 1977) suggests that free radical formation
in mice (see below), these data suggest that this type may be involved in the cardiotoxicity of doxorubicin.
of damage is not responsible for the in vivo antitumor
effect of doxorubicin.
3.5.2. Free Radical D a m a g e in Cardiac Tissue
Altogether these results suggest that in the clinical
situation only those cells could be damaged by a free In mice, both with acute (Myers et al., 1977) and
radical-dependent mechanism that are well oxy- chronic (Lazzarino et al., 1987) doxorubicin-induced
genated and exposed to relatively high concentrations cardiotoxicity, lipid peroxidation of heart tissue has
of doxorubicin or have a relative lack of antioxidant been observed, even though this process could not be
defense capacity. Probably this will include only a demonstrated in tumor cells (Myers et al., 1977) or
minor fraction of the tumor cells. However, in heart liver tissue (Lazzarino et al., 1987) of the same mice.
tissue such conditions are met during treatment with This suggests that cardiac tissue is specifically dam-
a single high dose of doxorubicin, and, in fact, acute aged by a free radical-dependent mechanism causing
cardiotoxicity is frequently observed. The possible lipid peroxidation. This is probably not only the
participation of free radicals in the development result of the relative abundance of mitochondria, and
of doxorubicin-induced cardiotoxicity is discussed efficient hydroxyl radical formation in cardiac tissue
below. as discussed above, but also of a relatively poor
Doxorubicin (Adriamycin) 227

antioxidant defense, since the heart has rather low binding derivatives of doxorubicin, like AD-32, may
levels of superoxide dismutase and catalase directly interact with the RNA polymerase to inhibit
(Doroshow et al., 1980) and a relatively low rate of transcription (Aubel-Sadron and Londos-Gagliardi,
glutathione turnover when compared to e.g. liver 1984).
tissue (Griffith and Meister, 1979). Furthermore, Recently, a novel mechanism by which doxoru-
during doxorubicin treatment, glutathione peroxi- bicin, as well as other intercalating drugs, may kill
dase tends to become depleted (Doroshow et al., tumor cells has been described; it is based on interfer-
1980). ence with the D N A breakage-reunion reaction
That a relative lack of antioxidant defense is indeed mediated by topoisomerase II (Tewey et al., 1984a).
involved in the development of cardiotoxicity in mice This mechanism would explain, why in tumor cells
is suggested by experiments showing that inhibition treated with clinically relevant doxorubicin concen-
of glutathione peroxidase (Doroshow et aL, 1980) or trations, both single- and double-strand protein-
decreasing glutathione levels in cardiac cells (Olson associated D N A breaks are observed (Potmesil et al.,
et al., 1980) increased the severity of doxorubicin- 1983, 1984). A major question now is whether this
induced cardiotoxicity, while agents that protect cells reaction plays a dominant role in the antitumor effect
against free radical damage, including N-acetylcys- of doxorubicin.
teine (Doroshow et al., 1981; Olson et aL, 1980), Evidence favoring this possibility is derived from a
cysteine (Olson et al., 1980), reduced glutathione comparison of treatments that inhibit topoisomerase
(Yoda et al., 1986) and alpha-tocopherol (Myers II-mediated D N A damage and affect doxorubicin
et al., 1977), protected against doxorubicin-induced cytotoxicity. Three conditions are known to inhibit
lipid peroxidation and cardiotoxicity. topoisomerase II in vitro : low temperature (Liu et al.,
In rats, the situation differs from that in mice, since 1983), high salt concentrations (Tewey et al., 1984b;
rat cardiac tissue appears to be unusually resistant to Liu et al., 1983) and doxorubicin concentrations
induction of lipid peroxidation (Mimnaugh et al., exceeding a critical value between 0.5 and 2.5 #M
1981; Julicher et al., 1985). As a consequence, lipid (Tewey et al., 1984b). For all three conditions there
peroxidation of rat heart tissue is barely detectable is evidence for a similar effect on doxorubicin cyto-
(Porta et al., 1983; Muliawan et al., 1980) and, in toxicity: (1) Incubation of cells at 0C totally abol-
cases where it was detected, did not parallel the ished doxorubicin cytotoxicity (Lane et al., 1987); (2)
development of cardiotoxicity (Porta et al., 1983). high salt dramatically protected against doxorubicin
Furthermore, neither N-acetylcysteine nor alpha- cytotoxicity at concentrations very similar to those
tocopherol protected rat myocardial cell cultures inhibiting topoisomerase II-dependent D N A damage
in vitro against the cytotoxicity of doxorubicin (Table 2; Liu et al., 1983; Iliakis and Lazar, 1987); (3)
(Newman et al., 1981). Therefore, in rats lipid perox- high concentrations of doxorubicin are known to
idation of cardiac tissue is probably not responsible inhibit the induction of protein-associated D N A
for the development of cardiotoxicity. On the other strand breaks (Potmesil et aL, 1983) which is expected
hand, lipid peroxides have been detected in serum of to inhibit its own cytotoxicity. Indeed, clonal survival
rats treated with doxorubicin (Thayer, 1984); these curves usually level off at relatively high concentra-
possibly originate from the liver (Mimnaugh et al., tions of doxorubicin (Barranco, 1984), possibly due
1981), since this organ is very well suited to support to this phenomenon. Together these data seem to be
free radical formation from doxorubicin. Hypotheti- consistent with a major role for interference with the
cally, these peroxides might contribute to cardiotoxi- breakage-reunion reaction of topoisomerase II in the
city, since heart cells are known to be very sensitive mechanism of cytotoxicity of doxorubicin.
to exposure to lipid peroxides (Noronha-Dutra and Such a mechanism is further supported by the
Steen, 1982). However, this possibility awaits further observation that topoisomerase II-dependent D N A
verification. damage is inhibited by ouabain (an inhibitor of
cellular Na/K ATPase), and that this inhibition
was closely correlated with decreased cytotoxicity of
4. FREE R A D I C A L - I N D E P E N D E N T doxorubicin in the presence of ouabain (Lawrence,
M E C H A N I S M S OF TOXICITY 1988). Interestingly, a decreased endogenous Na/

As doxorubicin is active against a wide variety of TABLE 2. Effect of Medium Tonicity on Doxorubicin Cyto-
cancers, while many tumor cell types, as pointed out toxicity and Topoisomerase I1 Activity
above, are not likely to be killed via a free radical- Interference with
dependent mechanism of cytotoxicity, efficient Medium % Clonal survival topoisomerase II
radical-independent mechanisms of cytotoxicity of tonicity after treatment activity
doxorubicin must exist. The most widely studied (mOsm) with doxorubicin (arbitrary units)
nonradical-dependent mechanisms of cytotoxicity are
300 0.1 --
based on the capacity of this drug to intercalate into 500 5.0 ++
double-stranded DNA. It has been known for 20 1000 70.0 +++*
years that anthracyclines can give rise to an intercala- 1300 100.0 ND
tion complex with D N A (for review see Aubel-
The tonicity of the medium can interfere with both
Sadron and Londos-Gagliardi, 1984). Possibly as a the cytotoxicity of doxorubicin (Liu et al., 1983) and the
consequence of this property, doxorubicin inhibits activity of topoisomerase II (Iliakis and Lazar, 1987).
both RNA and D N A synthesis, a process which Tonicity was increased by adding NaCI. The asterisk
might be involved in its antitumor effect (Aubel- indicates that interference was still incomplete. ND, not
Sadron and Londos-Gagliardi, 1984). Non-DNA- determined.
228 H. G. KEIZER et al.

K + ATPase activity was observed in doxorubicin- anisms of cytotoxicity must also exist. Since doxoru-
resistant L5178Y cells (Sugimoto et al., 1981). Fur- bicin at very low concentrations has been shown to
thermore, cell types found to be low in topoisomerase interfere with the activity of topoisomerase II, result-
lI activity were relatively resistant to doxorubicin ing in D N A strand breakage, while both D N A strand
(Potmesil et al., 1988). In a doxorubicin-resistant breakage and topoisomerase II activity are correlated
variant of P388 cells, reduced topoisomerase II with the cytotoxicity of doxorubicin, interference
activity was observed (Defile et al., 1989). Further a with topoisomerase II forms an attractive alternative
cell line selected for resistance to VP-16-213, another for free radical-dependent mechanisms of doxoru-
anticancer drug that is considered to have topoiso- bicin cytotoxicity against tumor cells.
merase II as an important target (Ross et al., 1984),
had a 10-fold lower topoisomerase II activity and was Acknowledgements--Research in the authors' laboratories is
cross-resistant to doxorubicin (Yalowich et al., 1987). supported by the Dutch Cancer Society.
In addition, cells hypersensitive to doxorubicin
contained an elevated activity of topoisomerase II
(Davies et al., 1988). Lastly, nuclei of doxorubicin-
resistant P388 cells showed less anthracycline- REFERENCES
induced D N A breaks after exposure to doxorubicin ALEGRIA, A. E., SAMUNI, A., MITCHELL, J. B., R1ESZ, P. and
than nuclei from sensitive cells, probably as a Russo, A. (1989) Free radicals induced by adriamycin-
consequence of reduced topoisomerase II activity sensitive and adriamycin-resistant cells: A spin-trapping
(Capranico et al., 1987). study. Biochemistry 28: 8653-8658.
In conclusion, interference with topoisomerase II ARCAMONE, F., CASSINELLI, G. and FRANCESCHI, G. (1972)
activity is an attractive mechanism in the tumoricidal Structure and physicochemical properties of adriamycin
action of doxorubicin, but its relative importance (doxorubicin). In: Int. Syrup. Adriamycin, pp. 9-22,
remains to be established. CARTER, S. K., DIMARCO, A. and GnIONE, M. (eds)
Springer, New York.
AUBEL-SADRON, G. and LONDOS-GAGLIARDI, D. (1984)
Daunorubicin and doxorubicin, anthracycline antibiotics,
5. S U M M A R Y AND CONCLUSIONS a physicochemical and biological review. Biochimie 66:
333-352.
Doxorubicin or doxorubicin-iron complexes can AVERBUCH, S. O., GAUD1ANO,G., KOCH, T. H. and BACHUR,
initiate the formation of hydroxyl radicals after re- N. R. 0985) Radical dimer rescue of toxicity and im-
duction of free doxorubicin or after reduction or auto- proved therapeutic index of adriamycin in tumor-bearing
oxidation of the doxorubicin-iron complex. These mice. Cancer Res. 45: 620045204.
hydroxyl radicals can damage cellular membranes by BABSON,J. R., ABELL,N. S. and REED,D. J. (1981) Protective
role of the glutathione redox cycle against adriamycin-
lipid peroxidation or damage cellular D N A . How- mediated toxicity in isolated hepatocytes. Biochem.
ever, the relative importance of such free radical Pharmac. 30: 2299-2304.
damage in the mechanism of killing tumor cells by BACHUR, N. R., GORDON, S. L. and GEE, M. V. (1977)
doxorubicin is not clear, because double-stranded Anthracycline antibiotic augmentation of microsomal
D N A , to which almost all intracellular doxorubicin electron transport and free radical formation. Molec.
is bound, inhibits free radical formation from dox- Pharmac. 13: 901-910.
orubicin while doxorubicin-iron complexes have BACHUR, N. R., GORDON, S. L. and GEE, M. V. (1978) A
never been detected in cells treated with doxorubicin. general mechanism for microsomal activation of quinone
To investigate the possible involvement of hy- anticancer agents to free radicals. Cancer Res. 38:
1745-1750.
droxyl radicals in the mechanism of cytotoxicity of
BACHUR, N. R., GEE, M. V. and FRIEDMAN, R. D. (1982)
doxorubicin, two approaches have been chosen. The Nuclear catalyzed antibiotic free radical formation.
first is to try to detect O H radical formation under Cancer Res. 42: 1078-1081.
conditions that kill the tumor cells and to investigate BARRANCO,S. C. (1984) Cellular and molecular effects of
whether O H radical formation correlates with the adriamycin on dividing and nondividing cells. Pharmac.
cytotoxicity of doxorubicin under various conditions. Ther. 24: 303-319.
The second is to investigate the effect of free radical BATES,D. A. and WINTERBOURN,C. C. (1982) Deoxyribose
scavengers on the cytotoxicity of doxorubicin. Under breakdown by the adriamycin semiquinone and H202:
conditions that kill tumor cells efficiently, no intra- evidence for hydroxyl radical participation. FEBS Lett.
145: 137-142.
cellular hydroxyl radical formation can be detected.
BERLIN, V. and HASELTINE, W. A. (1981) Reduction of
Only at relatively high doxorubicin concentrations adriamycin to a semiquinone-free radical by NADPH
was extracellular O H radical formation detectable. cytochrome P-450 reductase produces DNA cleavage in a
Experiments with free radical scavengers show that reaction mediated by molecular oxygen. J. biol. Chem.
certain cell types, including normal cardiac cells and 256: 4747-4756.
some tumor cell lines with a relative deficiency in their Bozzl, A., MAVELLI, I., MONDOVl, B., STROM, R. and
antioxidant defense capacity, are killed by these ROTILIO, G. (1981) Differential cytotoxicity of dauno-
extracellularly generated radicals. However, for the mycin in tumor cells is related to glutathione dependent
majority of tumor cells lines, free radical involvement hydrogen peroxide metabolism. Biochem. J. 194:369 372.
BROXTERMAN, H. J., PINEDO, H. M., KUIPER, C. M.,
could not be demonstrated in the mechanism of
SCHUtJRHU~S,G. J. and LANKELMA,J. (1989) Glycolysis in
cytotoxicity of doxorubicin. P-glycoprotein overexpressing human tumor cell lines.
As doxorubicin is active against a variety of solid Effect of resistance modifying agents. FEBS Lett. 247:
tumors and leukemias, while many tumor cell types 405-4 10.
are not killed by a free radical-dependent mechanism CAPRANICO, G., RIVA, A., TINELLI, S., DASDIA, T. and
of cytotoxicity, efficient nonradical-dependent mech- ZUNINO, F. (1987) Markedly reduced levels of anthracy-
Doxorubicin (Adriamycin) 229

cline-induced DNA strand breaks in resistant P388 GIANNI, U, VIGANO, L., LANZI, C., NIGGELER, M. and
leukemia cells and isolated nuclei. Cancer Res. 47: MALATESTA,V. (1988) Role of daunosamine and hydroxy-
3752-3756. acetyl side chain in reaction with iron and lipid peroxida-
CERVANTES, A., PINEDO, H. M., LANKELMA, J. and tion by anthracyclines. J. nam. Cancer Inst. 80:
SCHtJURHUIS, G. J. (1988) The role of oxygen-derived free ll04-1111.
radicals in the cytotoxicity of doxorubicin in multidrug GIGLI, M., DOGLIA, S. M., MILLOT, J. M., VALENTINI,L. and
resistant and sensitive human ovarian cancer cells. Cancer MANFAIT,M. (1988) Quantitative study of doxorubicin in
Left. 41: 169-177. living cell nuclei by microspectrofluorometry. Biochim.
DAVIES, K. J. A., DOROSHOW, J. H., CHAN, T. M. and biophys. Acta 950: 13-20.
HOCHSTEIN, P. (1983a) Mitochondrial reduction of an- GLAZER, R. I., HARTMAN, K. D. and RICHARDSON, C. L.
thracyclines: Oxygen radical production at the expense of (1982) Cytokinetic and biochemical effects of 5-
ATP. In: O x y Radicals and their Scavenger Systems, iminodaunorubicin in human colon carcinoma in culture.
Vol. H: Cellular and Medical Aspects, pp. 313-316, Cancer Res. 42: 117-121.
GREENWALD,R. A. and COHEN,G. (eds) Elsevier, New GOODMAN, J. and HOCHSTEIN, P. (1977) Generation of free
York. radicals and lipid peroxidation by redox cycling of
DAVIES, K. J. A., DOROSHOW, J. H. and HOCHSTEIN, P. adriamycin and daunomycin. Biochem. biophys. Res.
(1983b) Mitochondrial NADH dehydrogenase-catalyzed Commun. 77: 797-803.
oxygen radical production by adriamycin, and the relative GREEN, M. D., SPEYER, J. L. and MUGGIA, F. M. (1984)
inactivity of 5-iminodaunorubicin. FEBS Lett. 153: Cardiotoxicity of anthracyclines. Eur. J. Cancer clin.
227-230. Oncol. 20: 293-296.
DAVIES, S. M., ROBSON,C. N., DAVIES,S. L. and HICKSON, I. GRIFFITH, O. W. and MEISTER,A. (1979) Glutathione: inter-
D. (1988) Nuclear topoisomerase II levels correlate with organ translocation, turnover, and metabolism. Proc.
the sensitivity of mammalian cells to intercalating agents natn. Acad. Sci. U.S.A. 76: 5606-5610.
and epipodophylotoxins. 3. biol. Chem. 263:17724-17729. GUTI~R~Z, P. L., GEE, M. V. and BACHUR, N. R. (1983)
DEFFIE, A. M., BATRA, I. K. and GOLDBERG, G. J. (1989) Kinetics of anthracycline antibiotic free radical formation
Direct correlation between DNA topoisomerase II and reductive glycosidase activity. Archs Biochem. Bio-
activity and cytotoxicity in adriamycin-sensitive and phys. 223: 68-75.
-resistant P388 leukemia cell lines. Cancer Res. 49: 58~2. GUTTERIDOE, J. M. C. (1982) The role of superoxide and
DOROSHOW, J. H. (I 983a) Effect of anthracycline antibiotics hydroxyl radicals in phospholipid peroxidation catalysed
on oxygen radical formation in rat heart. Cancer Res. 43: by iron salts. FEBS Lett. 150: 454~58.
460-472. GUTTERIDGE, J. M. C. (1983) Adriamycin-iron catalysed
DOROSHOW, J. H. (1983b) Anthracycline antibiotic- phospholipid peroxidation: a reaction not involving
stimulated superoxide, hydrogen peroxide, and hydroxyl reduced adriamycin or hydroxyl radicals. Biochem. Phar-
radical production by NADH dehydrogenase. Cancer mac. 32: 1949-1952.
Res. 43: 4543-4551. GUTTERIDGE, J. M. C. (1984) Lipid peroxidation and possi-
DOROSHOW, J. H. (1986a) Prevention of doxorubicin- ble hydroxyl radical formation stimulation by the self-
induced killing of MCF-7 human breast cancer cells by reduction of a doxorubicin-iron (III) complex. Biochem.
oxygen radical scavengers and iron chelating agents. Pharmac. 33: 1725-1728.
Biochem. biophys. Res. Commun. 135: 330-335. GUTTERIDGE, J. M. C. and TOEG, D. (1982) Adriamycin-
DOROSHOW, J. H. (1986b) Role of hydrogen peroxide and dependent damage to deoxyribose: a reaction involving
hydroxyl radical formation in the killing of Ehrlich tumor iron, hydroxyl and semiquinone free radicals. FEBS Lett.
cells by anticancer quinones. Proc. natn. Acad. Sci. U.S.A. 149: 228-232.
83: 4514-4518. HAMILTON, T. C., WINKER, M. A., LOUIE, K. G., BATIST,G.,
DOROSHOW, J. H., LOCKER, G. Y. and MYERS, C. E. (1980) BEHRENS, B. C., TSURUO,T., GROTZINGER,K. R., MCKOY,
Enzymatic defenses of the mouse heart against reactive W. M., YOUNG, R. C. and OZOLS, R. F. (1985) Augmenta-
oxygen metabolites; alterations produced by doxorubicin. tion of adriamycin, melphalan, and cisplatin cytotoxicity
J. clin. Invest. 65: 128-135. in drug-resistant and -sensitive human ovarian carcinoma
DOROSHOW, J. H., LOCKER, G. Y., IFRIM,I. and MYERS, C. E. cell lines by buthionine sulfoximine mediated glutathione
(1981) Prevention of doxorubicin cardiac toxicity in the depletion. Biochem. Pharmac. 34: 2583-2586.
mouse by N-acetylcysteine. J. din. Invest. 68: 1053-1064. ILIAKIS, G. and LAZAR,W. (1987) Effect of sodium chloride
ELIOT, H., GIANNI, L. and MYERS, C. (1984) Oxidative concentration on adriamycin and N-trifluoroacetyladri-
destruction of DNA by the adriamycin-iron complex. amycin-14-valerate(AD32)-induced cell killing and DNA
Biochemistry 23: 928-936. damage in Chinese hamster V79 cells. Cancer Res. 47:
ERTTMANN, R., ERB, N., STEINHOFF, A. and LANDBECK, G. 1853-1858.
(1988) Pharmacokinetics of doxorubicin in man: dose and JOHNSON, B. A., CHEANG,M. S. and GOLDENBERG, G. J.
schedule dependence. J. Cancer Res. clin. Oncol. 114: (1986) Comparison of adriamycin uptake in chick embryo
509-513. heart and liver cells and murine L5178Y lymphoblasts in
FREEMAN, R. W., MACDONALD, J. S., OLSON, R. D., BOERTH, vitro: role of drug uptake in cardiotoxicity. Cancer Res.
R. C., OATES, J. A. and HARBISON, R. D. (1980) 46: 218-223.
Effect of sulfhydryl-containing compounds on the anti- JULICHER, R. H. M., VAN DER LAARSE,A., STERRENBERG,L.,
tumor effects of adriamycin. Toxic. appl. Pharmac. 54: BLOYSVANTRESLONG,C. H. F., BAST,A. and NOORDHOEK,
168-175. J. (1985) The involvement of an oxidative mechanism in
FUJITA, K., SHINPO, K., YAMADA, K., SATO, T., NIIMI, H., the adriamycin induced toxicity in neonatal rat heart cell
SHAMOTO,M., NAGATSU,T., TAKEUCHI,T. and UMEZAWA, cultures. Res. Commun. Chem. Path. Pharmac. 47: 35-47.
H. (1982) Reduction of adriamycin toxicity by ascorbate KALYANARAMAN, B., PEREZ-REYES, E. and MASON, R. P.
in mice and Guinea pigs. Cancer Res. 42: 309-316. (1980) Spin-trapping and direct electron spin resonance
GELVAN, D. and SAMUNI, A. (1988) Reappraisal of the investigations of the redox metabolism of quinone anti-
association between adriamycin and iron. Cancer Res. 48: cancer drugs. Biochem. biophys. Acta 630: 119-130.
5645-5649. KEIZER, H. G., VAN RIJN, J., PINEDO, H. M. and JOENJE, H.
GIANNI, L., ZWEIER,J. L., LEVY, A. and MYERS, C. E. (1985) (1988) Effect of endogenous glutathione, superoxide
Characterization of the cycle of iron-mediated electron dismutases, catalase, and glutathione peroxidase on adri-
transfer from adriamycin to molecular oxygen. J. biol. amycin tolerance of Chinese hamster ovary cells. Cancer
Chem. 260: 6820~i826. Res. 48: 4493-4497.
230 H.G. KEIZER et al.

KEIZER, H. G., DE LEEUW, S. J., VAN RIJN, J., PINEDO, H. M. MIMNAUGH, E. G., TRUSH, M. A. and GRAM, T. E. (1986) A
and JOENJE, H. (1989a) Effect of artificial electron accep- possible role for membrane lipid peroxidation in anthra-
tors on the cytotoxicity of mitomycin C and doxorubicin cycline nephrotoxicity. Biochem. Pharmac. 35: 4327-4335.
in human lung tumor cells. Eur. J. Cancer clin. Oncol. 25: MOORE, H. W. (1977) Bioactivation as a model for drug
1113-1118. design bioreductive alkylation. Science 197:527 532.
KEIZER, H. G., SCnUURHUIS, G. J., BROXTERMAN, H. J., MROSS, K., MAESSEN,P., VAN DER VIJGH, W. J. F., GALL, H.,
LANKELMA, J., SCHOONEN, W. G. E. J., VAN RIJN, J., BOVEN, E. and PINEDO, H. M. (1988) Pharmacokinetics
P1NEDO, H. M. and JOENJE, H. (1989b) Correlation of and metabolism of epidoxorubicin and doxorubicin in
multidrug resistance with decreased drug accumulation, humans. J. din. Oncol. 6: 517-526.
altered subcellular drug distribution and increased P- MUINDI, J. R. F., SINHA, B. K., GIANII, L. and MYERS, C. E.
glycoprotein expression in cultured SW-1573 human lung (1984) Hydroxyl radical production and DNA damage
tumor cells. Cancer Res. 49: 2988-2993. induced by anthracycline-iron complex. FEBS Lett. 172:
KENNEDY, K. A., SIEGFRIED, J. M., SARTORELLI, A. C. and 226-230.
TRITTON, T. R. (1983) Effects of anthracyclines on oxy- MUINDI, J., S1NHA, B. K., GIANNI, L. and MYERS, C. (1985)
genated and hypoxic tumor cells. Cancer Res. 43: 54-59. Thiol-dependent DNA damage produced by anthracy-
KRAMER, R, A., ZAKHER,J. and KIM, G. (1988) Role of the cline-iron complexes; the structure-activity relationships
glutathione redox cycle in acquired and de novo multi- and molecular mechanisms. Molec. Pharmac. 27:
drug resistance. Science 241: 694-697. 356-365.
LAMPIDIS,T. J., JOHNSON, L. V. and ISRAEL,M. (1981) Effects MULIAWAN, H., SCHEULEN,M. E. and KAPPUS, H. (1980)
of adriamycin on rat heart cells in culture: increased Acute adriamycin treatment of rats does not increase
accumulation and nucleoli fragmentation in cardiac ethane expiration. Res. Commun. Chem. Path. Pharmac.
muscle v. non-muscle cells. J. molec, cell. Cardiol. 13: 30:509-519.
913-924. MYERS, C. E., LISS, R. H., IFRIM, 1., GROTZINGER, K. and
LANE, P., VICHI, P., BAIN, D. L. and TRITTON, T. R. (1987) YOUNG, R. C. (1977) Adriamycin: the role of lipid perox-
Temperature dependence studies of adriamycin uptake idation in cardiac toxicity and tumor response. Science
and cytotoxicity. Cancer Res. 47: 4038-4042. 197: 165-167.
LAWRENCE, T. S. (1988) Reduction of doxorubicin cytotox- MYERS, C. E., GIANN1, L., S1MONE,C. B., KLECKER, R. and
icity by ouabain: correlation with topoisomerase-induced GREENE, R. (1982) Oxidative destruction of erythrocyte
DNA strand breakage in human and hamster cells. ghost membranes catalyzed by the doxorubine iron
Cancer Res. 48: 725-730. complex. Biochemistry 21:1707 1713.
LAZZAR1NO,G., VIOLA, A. R., MULLER1,L., ROTILIO, G. and NEWMAN, R. A., HACKER, M. P. and KRAKOEE, L H. (1981)
MAVELLI, I. (1987) Prevention by fructose-l,6-biphos- Amelioration of adriamycin and daunorubicin myo-
phate of cardiac oxidative damage induced in mice by cardial toxicity by adenosine. Cancer Res. 41: 3483-3488.
subchronic doxorubicin treatment. Cancer Res. 47: NOHL, H. and JORDAN, W. (1983) OH'-generation by adri-
651 lq5516. amycin semiquinone and H202: an explanation for the
LIU, L. F., ROWE, T. C., YANG, L., TEWEY, K. M. and CHEN, cardiotoxicity of anthracycline antibiotics. Biochem. bio-
G. L. (1983) Cleavage of DNA by mammalian DNA phys. Res. Commun. 114: 197-205.
topoisomerase II. J. biol. Chem. 258: 15365-15370. NORONHA-DUTRA, A. A. and STEEN, E. M. (1982) Lipid
MEIJER, C., MULDER, N. H., TIMMER-BOSSCHA,H., ZIJLSTRA, peroxidation as a mechanism of injury in cardiac my-
J. G. and DE VR1ES, E. G. E. (1987) Role of free radicals ocytes. Lab. Invest. 47:346 353.
in an adriamycin-resistant human small cell lung cancer OLSON, H. M. and CAPEN, C. C. (1977) Subacute cardio-
cell line. Cancer Res. 47: 4613-4617. toxicity of adriamycin in the rat. Biochemical and ultra-
MEREDITH, M. J. and REED, D. J. (1983) Depletion in vitro structural investigations. Lab. Invest. 37: 386-394.
of mitochondrial glutathione in rat hepatocytes and OLSON, R. D., MACDONALD, J. S., VAN BOXTEL, C. J.,
enhancement of lipid peroxidation by adriamycin and BOERTH,R. C., HARB1SON,R. D., SLON1M,A. E., FREEMAN,
1,3-bis(2-chloroethyl)-l-nitrosourea (BCNU). Biochem. R. W. and OATES, J. A. (1980) Regulatory role of gluta-
Pharmac. 32: 1383-1388. thione and soluble sulfhydryl groups in the toxicity of
MIMNAUGH, E. G. (1986) Potentiation by reduced gluta- adriamycin. 3. Pharmac. exp. TheE. 215: 450-454.
thione of Adriamycin-stimulated lipid peroxidation in PAN, S.-S. and BACHUR, N. R. (1980) Xanthine oxidase
kidney microsomes. Biochem. Pharmac. 35: 4337-4339. catalyzed reductive cleavage of anthracycline antibiotics
MIMNAUGH, E. G., TRUS8, M. A. and GRAM, T. E. (1981) and free radical formation. Molec. Pharmae. 17: 95--99.
Stimulation by adriamycin of rat heart and liver micro- PAN, S.-S., PEDERSEN, L. and BACHUR,N. R. (1981) Compar-
somal NADPH-dependent lipid peroxidation. Biochem. ative flavoprotein catalysis of anthracycline antibiotic:
Pharmac. 30: 2797-2804. reductive cleavage and oxygen consumption. Molec.
MIMNAUG8, E. G., TRUS8, M. A., GINSaURG, E. and GRAM, Pharmac. 19:184 186.
T. E. (1982) Differential effects of anthracycline drugs on PANNEERSELVAM, M., I~REDEI-IORST, R. and VOGEL, C. W.
rat heart and liver microsomal reduced nicotinamide (1987) Cell surface cytotoxic effect of immobilized
adenine dinucleotide phosphate-dependent lipid peroxi- doxorubicin is target cell-dependent and involves oxygen
dation. Cancer Res. 42: 3574-3582. radical formation. Proc. Am. Ass. Cancer Res. 28: 267.
MIMNAUGH, E. G., GRAM, T. E. and TRUSH, M. A. (1983) PORTA, E. A., JOUN, N. S., MATSUMURA,L., NAKASONE, B.
Stimulation of mouse heart and liver microsomat lipid and SABLAN,H. (1983) Acute adriamycin cardiotoxicity in
peroxidation by anthracycline anticaneer drugs: charac- rats. Res. Commun. Chem. Path. Pharmac. 41:125 137.
terization and effects of reactive oxygen scavengers. POTMESIL, M., KIRSCHENBAUM, S., ISRAEL, M., LEVIN, M.,
J. Pharmac. e.xp. Ther. 226: 806-816. KrmTARPAL, V. K. and SILBER, R. (1983) Relationship of
MIMNAUGH, E. G., KENNEDY, K. A., TRUSH, M. A. and Adriamycin concentrations to the DNA lesions induced
SINHA, B. K. (1985a) Adriamycin-enhanced membrane in hypoxic and euoxic LI210 cells. Cancer Res. 43:
lipid peroxidation in isolated rat nuclei. Cancer Res. 45: 3528-3533.
3296-3304. POTMESIL, M., ISRAEL, M. and SILVER, R. (1984) Two
MIMNAUGH, E. G., TRUSH, M. A., BHATNAGAR,M. and mechanisms of Adriamycin-DNA interaction in L1210
GRAM, T. E. (1985b) Enhancement of reactive oxygen- cells. Biochem. Pharmac. 33:3137 3142.
dependent mitochondrial membrane lipid peroxidation by POTMESIL, M., HSIANG, Y, H,, LIU, L. F,, BANK, B.,
the anticancer drug adriamycin. Biochem. Pharmac. 34: GROSSBERG, H., KIRSCHENBAUM, S., F'ORLENZAR, T. J.,
847-856. PENZINER, A., KANGANIS,D., KNOWLES, D., TRAGANOS,F'.
Doxorubicin (Adriamycin) 231

and SILVER,R. (1988) Resistance of human leukemic and with 8 structurally related anthracylines. Free Rad. Res.
normal lymphocytes to drug-induced DNA cleavage and Commun. 1: 41-54.
low levels of DNA topoisomerase II. Cancer Res. 48: SUGIMOTO,Y., NISHIMURA,T., SUZUKI, H. and TANAKA,N.
3537-3543. (1981) Alteration of membrane-associated enzymes in
RAJAGOPALAN,S., POLITI, P. M., SINHA, B. K. and MYERS, C. drug-resistant sublines of mouse lymphoblastoma
E. (1988) Adriamycin-induced free radical formation in L5178Y cells. J. Antibiotics 34: 1200-1205.
the perfused rat heart: implications for cardiotoxicity. SUG1OKA, K. and NAKANO, M. (1982) Mechanism of
Cancer Res. 48: 4766-4769. phospholipid peroxidation induced by ferric ion-ADP-
RAMU, A., COHEN, L. and GLAUBIGER,D. (1984) Oxygen adriamycin-co-ordination complex. Biochim. biophys.
radical detoxification enzymes in doxorubicin-sensitive Acta 713: 333-343.
and -resistant P388 murine leukemia cells. Cancer Res. 44: SVINGEN, B. A. and PowIs, G. (1981) Pulse radiolysis studies
1976-1980. of antitumor quinones: radical lifetimes, reactivity with
ROGERS, K. E. and T6K~S, Z. A. (1984) Novel mode of oxygen, and one-electron reduction potentials. Archs
cytotoxicity obtained by coupling inactive Anthracycline Biochem. Biophys. 2119:119-126.
to a polymer. Biochem. Pharmac. 33: 605~08. TEWEY, K. M., CHEN, G. L., NELSON, E. M. and LIU, L. F.
ROGERS, K. E., CARR, B. I. and TOKES, Z. A. (1983) (1984a) Intercalative antitumor drugs interfere with the
Cell surface-mediated cytotoxicity of polymer-bound breakage-reunion reaction of mammalian DNA topoiso-
Adramycin against drug-resistant hepatocytes. Cancer merase II. J. biol. Chem. 259: 9182-9187.
Res. 43:2741 2748. TEWEY, K. M., ROWE, T. C., YANG, L., HALLIGAN,B. D. and
Ross, K. L., AKMAN, S. A., SIK1C, B. I. and DOROSHOW,J. H. LIu, L. F. (1984b) Adriamycin-induced DNA damage
(1988) Free radical detoxifying enzymes in adriamycin- mediated by mammalian DNA topoisomerase II. Science
resistant cell lines. Proc. Am. Ass. Cancer Res. 29: 503. 226: 466-468.
ROSS, W. E. and SMITH,M. C. (1982) Repair of deoxyribonu- THAYER, W. S. (1984) Serum lipid peroxides in rats treated
cleic acid lesions caused by Adriamycin and ellipticine. chronically with adriamycin. Biochem. Pharmac. 33:
Biochem. Pharmac. 31:1931 1935. 2259-2263.
Ross, W., ROWE, T., GLISSON, B., YALOWICH, J. and LID, L. THORNALLEY, P. J. and DODD, N. J. F. (1985) Free radical
(1984) Role of topoisomerase II in mediating epipodo- production from normal and adriamycin-treated rat
phyllotoxin-induced DNA cleavage. Cancer Res. 44: cardiac sarcosomes. Biochem. Pharmac. 34: 669~574.
5857 5860. TRITTON, T. R. and YEE, G. (1982) The anticancer agent
ROWLEY, D. A. and HALLIWELL, B. (1983) DNA damage Adriamycin can be actively cytotoxic without entering
by superoxide-generating systems in relation to the mech- cells. Science 217: 248-250.
anism of action of the anti-tumor antibiotic Adriamycin. WADLER, S., GREEN, M. D. and MUGGIA, F. M. (1986)
Biochim. biophys. Acta 761: 86-93. Synergistic activity of doxorubicin and the bisdioxopi-
SATO, S., IWAIZUMI,M., HANDRA, K. and TAMURA,Y. (1977) perazine (+)-l,2-Bis(3,5-dioxopiperazinyl-l-yl)propane
Electron spin resonance study on the mode of generation (ICRF 187) against the routine sarcoma Sl80 cell line.
of free radicals of daunomycin, adriamycin, and carbo- Cancer Res. 46:1176-I 181.
quone in NAD(P)H-microsome system. Gann 68: WINTERBOURN,C. C., GUTTERIDGE,J. M. C. and HALLIWELL,
603~08. B. (1985) Doxorubicin-dependent lipid peroxidation at
SINHA, B. K. and CHIGNELL, C. F. (1979) Binding mode of low partial pressures of 02. J. Free Rad. Biol. Med. 1:
chemically activated semiquinone free radicals from 43-49.
quinone anticancer agents to DNA. Chem. Biol. Interact. YALOWICH,J. C., ROBERTS,D., BENTON,S. and PARGANAS,E.
28: 301-308. (1987) Resistance to etoposide (VP-16) in human
SINHA, B. K. and LEWIS GREGORY, J. (1981) Role of one- leukemia K562 cells is associated with altered DNA
electron and two-electron reduction products of topoisomerase II (TOPO II) activity and rapid reversal of
adriamycin and daunomycin in deoxyribonucleic acid drug-induced DNA damage. Proc. Am. Ass. Cancer Res.
binding. Biochem. Pharmac. 30: 2626-2629. 28: 277.
SINHA, B. K., TRUSH, M. A., KENNEDY, K. A. and YODA, Y., NAKAZAWA, M., ABE, T. and KAWAKAMI, Z.
MIMNAUGH, E. G. (1984) Enzymatic activation and (1986) Prevention of doxorubicin myocardial toxicity
binding of adriamycin to nuclear DNA. Cancer Res. 44: in mice by reduced glutathione. Cancer Res. 46:
2892 2896. 2551-2556.
SINHA,B. K., KATKI,A. G., BATIST, G., COWAN,K. H. and YOUNG, R. C., OZOLS, R. F. and MYERS, C. E. (1981) The
MYERS, C. E. (1987a) Adriamycin-stimulated hydroxyl anthracycline antineoplastic drugs. New Engl. J. Med.
radical formation in human breast tumor cells. Biochem. 305: 139-153.
Pharmac. 36:793 796. ZWEIER,J. L. (1983) Reduction of 02 by iron-adriamycin.
SINHA, B. K., KATKI, A. G., BATIST, G., COWAN, K. H. and J. biol. Chem. 259: 6056-6058.
MYERS, C. E. (1987b) Differential formation of hydroxyl ZWEIER,J. L. (1985) Iron-mediated formation of an oxidized
radicals by Adriamycin in sensitive and resistant MCF-7 adriamycin free radical. Biochim. biophys. Acta 839:
human breast tumor cells: implications for the mechanism 209-213.
of action. Biochemistry 26:3776 3781. ZWE1ER, J. L., GIANNI, L., MUINDI, J. and MYERS, C. E.
STERRENBERG,t., JULICHER,R. H. M., GOOSSENS, P. A. L., (1986) Differences in O 2 reduction by the iron complexes
BAST, A. and NOORDHOEK, J. (1985) Anthracycline- of adriamycin and daunomycin- the importance of the
induced oxygen consumption and oxidative damage in rat sidechain hydroxyl group. Biochim. biophys. Acta 884:
liver microsomes are not necessarily coupled: a study 326-336.

View publication stats

Vous aimerez peut-être aussi