You are on page 1of 12

Curr. Med. Chem.

- Anti-Cancer Agents, 2003, 3, 439-450 439

The Quinolone Family: From Antibacterial to Anticancer Agents

Claudia Sissi* and Manlio Palumbo

Dept. of Pharmaceutical Sciences, V. Marzolo 5, 35100 Padova, Italy

Abstract: The present review focuses on the structural modifications responsible for the transformation of an antibacterial
into an anticancer agent. Indeed, a distinctive feature of drugs based on the quinolone structure is their remarkable ability
to target different type II topoisomerase enzymes. In particular, some congeners of this drug family display high activity
not only against bacterial topoisomerases, but also against eukaryotic topoisomerases and are toxic to cultured mammalian
cells and in vivo tumor models. Hence, these cytotoxic quinolones represent an exploitable source of new anticancer
agents, which might also help addressing side-toxicity and resistance phenomena. Their ability to bind metal ion co-
factors represents an additional means of modulating their pharmacological response(s). Moreover, quinolones link
antibacterial and anticancer chemotherapy together and provide an opportunity to clarify drug mechanism across divergent
species.

THE QUINOLONE FAMILY: FROM ANTIBAC- At the moment, the main interest for these compounds is
TERIAL TO ANTICANCER AGENTS related to the treatment of respiratory diseases [3].
The history of quinolones begins in the early 1960’s with
the synthesis of the first member of the family, nalidixic acid O O
(Fig. 1) [1]. Its antibacterial properties appeared immediately
O
to be very interesting. Unfortunately, its applications in
humans revealed only a moderate response and it was soon
H3 C N N
dropped from clinical use. However, the generally simple
and flexible synthetic route to quinolone derivatives, allowed
to producing a large number of analogs including the most
NALIDIXIC ACID
active and broad spectrum oral antibacterials currently in use
[2]. O O O O
A most successful achievement was the preparation of F F
fluoroquinolones. In fact, the introduction of a fluorine atom O O
at the C-6 position increased several folds the activity of the
N N N N
drug and led to compounds active against a broader spectrum
of bacteria. The first fluoroquinolone clinically used was HN HN
norfloxacin (Fig. 1). Its poor tissue distribution limited its
applications to the treatment of urinary tract infections. NORFLOXACIN CIPROFLOXACIN
Modification of the substituent at N-1 position produced
ciprofloxacin (Fig. 1). This drug presents an excellent Fig. (1). Chemical structure of antibacterial quinolones.
systemic activity upon oral administration and has become
indeed one of the most frequently prescribed antibiotics. Unfortunately, the high efficiency and broad spectrum of
More recently, fluoroquinolones have been further optimised activity shown by quinolones are now seriously challenged
to improve both pharmacokinetic and pharmacodynamic by the selection of resistant strains, even for naturally highly
properties like favorable bioavailability allowing oral susceptible species like Escherichia coli [4]. Thus, the
administration, good tolerability, high tissue concentrations present research in the field is specially devoted to overcome
as well as superior bactericidal activity against a broad resistance phenomena that are emerging as the most relevant
spectrum of clinically relevant pathogens including complication in antibacterial therapies.
enterobacteria, Pseudomonas aeruginosa, Staphylococcus
aureus and Streptococcus pneumoniae. Thus, new Biological Targets of Antibacterial Quinolones
generations of fluoroquinolones are now available [2]. They
Forty years of research in the quinolone field have
preserve excellent potency against Gram-negative bacteria allowed to elucidating some key aspects of their mechanism
and, at the same time, they show an increased activity of action. Their main biological targets are the DNA gyrase
towards Gram-positive bacteria compared to ciprofloxacin.
(Gyr) and topoisomerase IV (Top4), two enzymes belonging
to the type II topoisomerase family [5, 6]. These enzymes are
*Address correspondence to this author at the Dept. of Pharmaceutical essential in all bacteria and are expressed in prokaryotic
Sciences, V. Marzolo 5, 35100 Padova, Italy; Tel: +39-049-827-5717; organisms only, features that make them ideal antibacterial
Fax: +39-049-827-5711; E-mail: claudia.sissi@unipd.it drug targets. Indeed, the prominent selectivity of quinolones

1568-0118/03 $41.00+.00 © 2003 Bentham Science Publishers Ltd.


440 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

in poisoning the bacterial type II topoisomerases vs. the In vitro, the two enzymes discriminate selectively
corresponding mammalian enzymes represents the rational between intra and inter-molecular topological changes: while
basis for their safe use in antibacterial chemotherapy. Top4 catalyses, preferentially intermolecular reactions like
decatenation, Gyr favors an intramolecular reaction such as
Gyr and Top4 exhibit close structural and functional
supercoiling [16].
similarities. This emerges from their high level of primary
sequence homology and from their common catalytic cycle. These differences are particularly significant in vivo. In
In fact, both cleave two DNA strands in a concerted fact, although both enzymes are essential for the proper
sequence of events and transport one duplex DNA segment occurrence of chromosome duplication and segregation
through the break. In so doing, they can catenate or processes, they probably work at different stages of
decatenate duplex DNA rings and alter DNA topology. The replication. In particular, experimental evidence suggests
major difference between the two enzymes rests on the fact that Top4 prefers to unlink precatenane, whereas DNA
that Gyr is able to introduce negative supercoils into DNA, gyrase works to remove positive supercoils [17-19].
whereas Top4 is not.
Gyr is formed by two different subunits, GyrA and GyrB, Mechanism of Action of Antibacterial Quinolones
that are arranged in a A2B2 tetramer to constitute the active Quinolones act in a similar fashion against the two
protein [7, 8]. prokaryotic type II enzymes. They essentially block the
GyrA is a 97 kDa protein (875 aa) encoded by gyrA. topoisomerase catalytic cycle when the protein is covalently
Tryptic cleavage of this subunit generates two fragments that linked to the cleaved DNA (cleavage complex). Stabilization
correspond to two main functional domains. The 64kDa N- of the enzyme-DNA complex has been confirmed to block
terminal domain (residues 7 to 571) contains the catalytic the progression of the replicative machinery and to create
tyrosine (Tyr122), whereas the 33kDa C-terminal domain DNA lesions that induce bacterial SOS response [20-22].
(residues 572 to 875) is involved in DNA binding and At the molecular level, the interactions of quinolones
wrapping. occur mainly with the A subunit of Gyr and with ParC of
GyrB is an 89.9 kDa (804 aa) protein encoded by the Top4. In fact, a region spatially close to the active tyrosine
gyrB gene. Also this protein contains two functional and called Quinolone Resistance Determining Region
domains. The 43kDa N-terminal fragment (aa 2-393) is the (QRDR) has been identified. Aminoacid mutations in this
ATP binding and hydrolysis domain; the 47 kDa C-terminal region greatly reduce the affinity of quinolones for the
domain (aa 394-804) is involved in interactions with GyrA enzyme-DNA complex. The most relevant positions in E.
and DNA. Coli are Ser83 and Asp87 in GyrA, and the corresponding
Ser79 and Asp83 in ParC [23, 24]. Mutations in GyrB and
More recently, the identification of the genes encoding ParE, although implicated in fluoroquinolone resistance, are
Top4 subunits, parC and parE, confirmed a similar less frequent. The current hypothesis is that they occur at the
quaternary structure for the two prokaryotic enzymes [9,10]. interface between the two subunits inducing subtle structural
In fact parC encoded an 83.7 kDa protein of 752 aa changes in the overall protein architecture.
called ParC, whereas parE encoded a 70.2 kDa protein of The structural features of the quinolone-gyrase-DNA
630 aa called ParE [10, 11]. These two proteins share an complex have been only partially elucidated [25].
extended sequence homology with GyrA and GyrB Experimental evidence suggests that quinolones, in the
respectively and a similar distribution of functional domains presence of proper metal ions, bind single stranded DNA and
[11,12]. In addition, the Top4 active form is constituted by a GyrA with moderate affinity [26, 27]. However, they bind
C2E2 tetramer as in the case of Gyr. efficiently to the gyrase-DNA complex [28].
The catalytic cycle of the two enzymes is essentially the At the DNA level, cleavage is not required to stimulate
same. They initially bind a dsDNA segment (G segment). drug binding: the structural distortion of the double helix
Complex formation with ATP induces the closure of an bound to the protein and stabilized by selective contacts with
ATP-operated clamp formed by the N-terminal domains of protein residues, appears to be sufficient to provide a
the GyrB or ParE dimer that lead to the capture of a second favorable interaction site for quinolones [29]. Additionally,
dsDNA fragment, the T segment [13]. Thus, a concerted the presence of quinolones alters not only the structural
mechanism occurs: trapping of the T segment induces features of GyrA (binding site) but also those of the overall
cleavage of the G segment on both strands. In the cleavage A2B2-DNA complex [27, 30] inducing modifications in the
complex a tyrosine is covalently bound to the 5’-end of the kinetic rates of different catalytic steps.
DNA cleavage site. On the opposite strand, the cut is 4-bp
staggered [14]. The combination of these events triggers Role of Quinolones in Eukaryotic Cells
nucleotide hydrolysis. ADP rapidly dissociates from the
proteins, thus opening the clamp and allowing the release of In eukaryotes, DNA gyrase and topoisomerase IV are
the T-segment after its crossing through the G-gate. The re- functionally replaced by two isoenzymes: topoisomerase II α
sealing and release of the G-segment allows the enzyme to and β, a 170 and a 180 kDa protein respectively [5]. These
start a new catalytic cycle [15]. proteins share a similar catalytic cycle, the main difference
resting in the fact that the active form of eukaryotic
Despite the extensive similarity in sequence, structure topoisomerase II (Top2) is constituted by a homodimer.
and functional properties, the two topoisomerases have However, as supported by the extended sequence homology,
distinct identities and roles. Top2 can be seen as the fusion of the GyrA-GyrB or ParC-
The Quinolone Family Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 441

ParE subunits. Additionally, although no crystallographic investigations on quinolone selectivity between bacterial and
data are available at present for any of the above mentioned mammalian type II topoisomerases have been carried out
full length enzymes, the structures of the crystallized [39-41]. It emerged that, generally, the inhibitory activities
fragments of GyrB and GyrA have been found to be of quinolones against prokaryotic or eukaryotic type II
complementary to the corresponding fragments of yeast topoisomerases significantly correlated with their
Top2 [31-33]. antimicrobial or cytotoxic activities respectively. However,
Like bacterial Gyr and Top4, also eukaryotic Top2 is a no relationship has been established between the two
validated pharmacological target. In fact, many clinically mechanisms as large differences in selectivity were observed
useful anticancer drugs exert their cytotoxic action by on varying the nature of the test quinolones.
interfering with this enzyme. Such drugs can be divided into In the late eighties, quinolones having a 4-pyridinyl
two main classes: inhibitors and poisons. group at C-7 were synthesized. Among them rosoxacin
Topoisomerase inhibitors, by binding to the enzyme, gained interest as it was particularly effective in treating
prevent it from performing its catalytic cycle. This impairs gonococcal infections (Fig. 2) [42]. With the aim to increase
DNA processing and causes cytostatic effects. As it is highly the quinolone activity toward gram-positive bacteria various
7-(4-pyridinyl) derivatives were examined [43]. Remarkably,
preferable to kill a cancer cell than to stop its growth, Top2
some of these new compounds produced an unacceptable
inhibitors are not particularly appealing as drug candidates.
toxicity profile in vitro inducing micronuclei, a genetic
On the other side, Top2 poisons act in the same way as toxicity endpoint.
quinolones: by increasing the DNA cleavage rate or by redu-
At the same time, a new 6,8-difluoro-7-pyridyl-4-
cing the DNA religation rate, the so-called cleavage complex
quinolone, CP-67, 015, with potent antibacterial activity, was
spans its life and DNA breaks persist longer. Stabilization of
shown to strongly enhance topoisomerase II-mediated DNA
the cleavage complex converts the topoisomerase into an
cleavage, which was related to its reported clastogenic effect
endogenous toxin that “freezes” DNA damage in the
genome. This triggers a sequence of biochemical events that on DNA in cell culture and to its positive mutagenic
finally leads to a programmed cell death, called apoptosis. response in mouse lymphoma cells [44].
Since then, different directions were followed to eluci-
Common features in the mechanism of action of antibac-
terial quinolones and antitumor drugs have suggested that date the structure activity relationships responsible for the
both compounds have a similar mode of interaction with the interference of quinolones with eukaryotic topoisomerase II.
type II DNA topoisomerase-DNA complexes. In particular,
Cytotoxic Quinolones: Structure Activity Relationships
comparison between quinolones and antitumor drugs of the
epipodophyllotoxin family (etoposide and teniposide) has Initial approaches to define proper SAR for cytotoxic
been considered especially relevant, since neither type of quinolones were based on systematic structural modifica-
drug is a DNA intercalator [34]. Further indications came tions of canonical antibacterial fluoroquinolones. These lead
from studies carried out within archaebacteria. These to the identification of characteristic structural features
organisms occupy an intermediate phylogenetic position linked to cytotoxic activity against eukaryotic cells. In parti-
between eukaryotes and eubacteria [35]. In these systems, cular, starting from norfloxacin or ciprofloxacin, it emerged
comparable sensitivity to poisons of each type II topoiso- that no single position has a controlling effect on the
merase was observed and the DNA cleavage patterns induced observed cytotoxicity but, more likely, a combination of the
by ciprofloxacin and etoposide were found to be very similar various substituents contributes to directing the poisoning
[36, 37]. These findings strongly supported a common mode activity. A schematic evolution of cytotoxic quinolones is
of interaction with the DNA-DNA topoisomerase II com- reported in Fig. (2).
plexes for ciprofloxacin and etoposide.
The presence of a fluorine atom at position C-6 of the
How can a small molecule discriminate efficiently quinolone ring is a general requirement to grant potent
between remarkably similar enzymes? Due to the lack of antibacterial activity in “classical” quinolones [45]. It has
information concerning the cleavage complex in the been proved that, in order to stabilize the cleavage complex
presence/absence of drugs, no final answer is available at the with mammalian Top2, the presence of two halogens at C-6
moment. However, this open question gives a new input in and C-8 of quinolone skeleton is preferred. In fact, a direct
the field: use quinolones as leading compounds to be comparison of 6-fluoro (CP-115, 955) and 6,8-difluoro (CP-
optimized not against their “traditional” targets (Gyr/Top4) 115, 953) derivatives showed that removal of the C-8
but towards mammalian Top2, thus transforming antibac- fluorine decreased the ability of the quinolone to stimulate
terial into anticancer drugs. enzyme-mediated DNA cleavage by approximately 2.5-fold.
One of the first reports in this direction came in the Activity on the mammalian enzyme parallels the cell killing
middle 80’s, when the influence of some antibacterial ability observed towards Chinese hamster ovary cells, confir-
quinolones on elements of the eukaryotic DNA replication ming that the C-8 fluorine increases the potency of quinolone
machinery was tested in vitro [38]. It was found that derivatives against Top2 and mammalian cells [46].
quinolones such as ciprofloxacin, norfloxacin and ofloxacin In the presence of fluorine atoms at 6 and 8 positions, the
interfered only slightly with the function of calf thymus change of N-1 substituent from ethyl to cyclopropyl was also
Top2, the prokaryotic enzyme Gyr being approximately 100- critical for antiproliferative activity. In fact, in most
fold more sensitive to inhibition than its eukaryotic homogeneous series of derivatives, the presence of a
counterpart. Starting from this pioneering study, systematic cyclopropyl group increased the activity compared to an
442 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

O O

N
N
ROSOXACIN

O O O O
F F
O O
H3 C
N
N F N F

CP-67,015 CH3
WIN 57294

O O O

F F H
O
H3 C
N R

F N F
HO
CH3
CP-67,804 R = H, CH 3, C6H5

O O O OH
F F
O
H3 C
HO
F N F
HO

CP-115,953 CH3
WIN 64593

Fig. (2). Historical development of some cytotoxic quinolones.

ethyl group [47, 48]. Thus, it seems that at N-1, the SAR [51]. These results suggested a direct interaction of this
profile obtained for antibacterial activity is conserved for cell portion of the quinolone with the enzyme. Conceivably, the
cytotoxicity. Combining the above findings, it is suggested C-7 substituent should affect the prokaryotic/eukaryotic
that the concerted presence of the halogen at C-8 and the enzyme selectively. Indeed, methyl substituents at the C-7
cyclopropyl at N-1 may produce better interference of piperazine group influenced potency against the mammalian
quinolones with Top2 than with Gyr [49, 50]. enzyme, stereochemistry being a discriminating factor. In
A variety of substitutions were also introduced at fact 3,5-dimethylpiperazinyl derivatives were active in
position C-7. In fact, the presence of a basic amino group in stimulating enzyme-mediated DNA cleavage only in the
the aliphatic cyclic substituent at position C-7 is a strict trans configuration. This was particularly interesting because
cis- or trans-methyl substitution on the piperazine had little
requirement for antibacterial activity. Additionally,
modifications at this position where shown to play a key role effect on the activity against Gyr, suggesting that only in the
in directing the drug preferentially towards Gyr or Top4 mammalian enzyme an asymmetric barrier exists which
The Quinolone Family Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 443

influences productive quinolone interaction and favors the involved in hydrogen bonding with the target macromole-
less bulky trans-3,5-dimethylpiperazine substituent at C-7 cules. In the new derivatives, the profound modifications
[52]. introduced suggest the onset of new interactions involving
the drug aromatic and/or hydroxyl-groups in the cleavage
Parallel studies evidenced that compounds with C-7
complex.
pyrrolidine substituents were more cytotoxic than those with
piperazine substituents [50]. This is actually in line with In the 7-(2,6-dimethyl-4-pyridinyl) family, the substitu-
observations made on derivatives related to the earlier tion of the nitrogen at position 1 with a sulphur atom leads to
cytotoxic quinolones [44]. Looking both at the stimulation of inactive derivatives [54]. However, as reported in Fig. (3),
DNA cleavage and at the cytotoxicity against cultured other positions can support the introduction of this heteroa-
mammalian cells, it was observed that the presence of an tom without the loss of activity. An example is represented
aromatic group contributed greatly to drug activity. In by the isothiazoloquinolones A-65281 and A-65282 [57].
particular, in this series of compounds, a 4'-hydroxyphenyl These derivatives exhibit a heterocyclic moiety added to the
substituent at the C-7 position was critical for potency quinolone system, which can be considered as a modification
towards the mammalian Top2 [48]. The compendium of of the carboxylic group. It is worth noting that, in spite of the
these studies on structure-activity relationships is represented fact that these derivatives do not exhibit an aromatic
by CP-115, 953, [6,8-difluoro-7-(4'-hydroxyphenyl)-1- substituent at position C-7, they are active against both
cyclopropyl-4- quinolone-3-carboxylic acid], one of the most bacterial and eukaryotic type II topoisomerases.
cytotoxic quinolone thus far synthesized (Fig. 2) [53].
More recently, hetero substitution produced thieno[3'
More recently, novel substitutions were introduced at 2':4.5]thieno[2,3-c]quinolones [58]. In this series, derivatives
position C-7, which can provide good models for further bearing a 3-dimethylaminopropyl substituent on the quinolone
design of potent antitumor quinolones. A remarkable example nitrogen and a methoxycarbonyl group at position 9 exhi-
is the introduction of an unsaturated aminoazabicyclo group bited prominent antitumor activity. Cytotoxicity was reduced
[47]. Compounds of this type are peculiar because they when an anilido substituent was present at position 9.
associate maximal activity with the presence of an atypical
amino group at C-5 of the quinoline carboxylic acid. The A similar approach leads to the screening of 1,8-
possibility to substitute the 5-H with NH2 or F, maintaining naphthyridine derivatives. Indeed, a series of 1,7-
the activity against mammalian Top2 was further confirmed disubstituted-6-fluoro-1,4-dihydro-4-oxo-1,8-naphthyridine-
in other series of derivatives [52, 54]. 3-carboxylic acids with cytotoxic activity comparable to that
of etoposide on cancer cell lines in vitro and on murine P388
In the absence of an aromatic moiety at position C-7, a 7- leukemia in vivo has been very recently disclosed, the
[4-(2-hydroxyiminoethyl)piperazin-1-yl] group has been leading compound being AT 3639-1 (Fig. 3) [59]. SAR
introduced. Among various combinations, 1-ethyl-6-fluoro-7- studies performed by changing N-1 and C-7 positions and
{4-[2-(4-chlorophenyl)-2-hydroxyiminoethyl]-1-piperazinyl}- the core ring structure led to the conclusion that a
4-oxo-1,4-dihydro-3-quinoline carboxylic acid demonstrated heterocyclic 2-thiazolyl group at the N-1 position of the
the most significant activity against renal cancer cell lines naphthyridine structure is the best substituent for antitumor
[55]. activity. Regarding core ring structure, the naphthyridine
Other intriguing results concern position C-3. It is well derivative, reminding nalidixic acid, is the most active
established that, in antibacterial quinolones, the 3-COOH or followed by pyridopyrimidine analogs. Finally, aminopyrro-
its isosteric replacement is a fundamental requirement for lidine substituents at the C-7 position were more effective
activity (Fig. 2). However, this functional group can be than other amines or thioethers.
replaced even with an hydrogen still granting effective Other cytotoxic derivatives are based upon the ofloxacin
poisoning of Top2. It seems that, to inhibit the eukaryotic or pyrimido[1,6-a]benzimidazole skeleton and thus they
enzyme, the basic requirement is the coplanarity of the C-3 present an additional ring with reference to classical
substituent with the quinoline ring. Thus, although in 3-H quinolones. Examples are WIN 58161 and Ro 47-3359
derivatives small substituents can be beneficial at C-2, no respectively, reported in Fig. (4). The WIN derivative was
such residue can be introduced if the carboxyl group is identified as a topoisomerase II poison during the screening
present, as it will destabilize the coplanar orientation of the of several enantiomerically pure (2,6-dimethyl-4-pyridinyl)
acidic moiety [54]. quinolones, previously shown to be potent inhibitors of
Removal of the carboxylic group opened up new bacterial DNA gyrase. Among these and other analogs,
synthetic opportunities. In fact, introduction of a phenyl topoisomerase II inhibitory potency was found to depend on
group or a related heteroaromatic ring at C-2 enhanced Top2 the size and substitution of the bridge spanning the 1- and 8-
poisoning activity [56]. The distance between the two positions of the quinoline ring [60]. The use of the
aromatic moieties is apparently crucial, as only a methylene enantiomerically pure WIN 58161 confirms that Gyr and
linker allowed to maintain biological activity. Further Top2 inhibitory activities are enantioselective processes. In
increase in cytotoxic activity was obtained upon introduction fact only the (S) enantiomers show biological activity [61].
of hydroxyl- substituents into the C-2 phenyl group. In
The pyrimido[1,6-a]benzimidazole family is another
particular, the 2,6-dihydroxy benzyl derivative (Win 64593) class of drugs modelled on quinolones and generally active
is the most active compound available to date. The role of
against DNA gyrase [62]. To introduce cytotoxic activity,
hydroxyl substitution is not clear yet. In quinolones carrying the aliphatic group (4-N-methylpiperazine) at the ring
the carboxyl group, the acidic moiety can participate with the
position equivalent to C-7 in quinolones was again replaced
ketone at position 4 in chelating metal ions or can be
444 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

O O
F
NH
N N S

N F
O O
F A 65281

NH O O
R X N S F
R NH
isothiazoloquinolones N N S
H2N
F

A 65282
O O
F
O

N N N
H2N

N S

AT 3639-1

Fig. (3). Chemical structure of cytotoxic quinolones containing a sulphur atom.

O O O O
F F
O O
H3 C
N N N
N O N S CH3
H3 C
H
OFLOXACIN CH3
WIN 58161

OH O OH
O
N N
O F N O
F N
H3 C
N
N N
N N
H3 C
Ro 46-2825 CH3
(CH2) 2N(CH3 )2 Ro 47-3359

N N
F O

H3 C
N
N F

CH3
WIN 63320

Fig. (4). Chemical structure of cytotoxic quinolones containing an extended planar ring system.

with an aromatic basic substituent (2,6-dimethylpyridine) An extended planar ring system is present also in WIN
yielding RO 47-3359 (Fig. 4) [63]. 63320, a pyrazoloquinoline derivative (Fig. 4) [64]. The
The Quinolone Family Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 445

pyrazole-ring fusion blocks the carbonyl group in the SAR studies, in particular connecting DNA-binding and
conformation previously reported as the active one, which biological activity, were performed for a number of A-62176
results in an increment of the in vitro and in vivo cytotoxicity analogs [68]. The results indicate that DNA-binding affinity
[54]. is predictive for topo II inhibition response. In addition, data
showed the requirement for the carbonyl group at position 4,
Starting from the fact that DNA-intercalating drugs often
exhibit cytotoxic properties, the quinolone structure has been for the free amine at position 1 and the possibility of
manipulated in order to yield a more extended planar surface increasing DNA-binding/topo II activity by introducing
possibly granting efficient insertion into a double-helical aliphatic (methyl) substitutions at position 9. On the
nucleic acid. A screening program to search for novel contrary, fluorine substitution at the latter position resulted in
strong reduction of drug potency.
quinolone-related antitumor agents lead to the identification
of the quinobenzoxazine family, characterized by a fused Finally, considering that the phenyl ring portion of the
hetero-tetracycle [65-67]. Several compounds were quinobenzoxazines might be inserted between DNA base
synthesized and investigated, some of which exhibited pairs in the cleavage complex, the drug intercalation ability
excellent in vitro cytotoxic activity using cancer cell lines, was increased by extending the phenyl ring to a naphthyl
including the multidrug resistant P388/ADR line, and also ring. A series of new pyridobenzophenoxazine analogs
possessed good activity in vivo against both systemic tumors (compounds 1-3 in Fig. 6) were thus designed and
and subcutaneously implanted murine solid tumors as well as synthesized [69]. Because of the steric hindrance between
human tumor xenografts. The drugs’ potency was typically the hydrogens on the pyrido- and benzo- rings, the naphthyl
in the sub-micromolar range and produced a significantly and quinolone portions of compound 1 are not coplanar. This
increased life span. may have a negative effect on its DNA intercalation ability.
Among the tested compounds, two analogs emerged as Therefore, compound 1 was less potent than compounds 2
and 3 and A-62176. Some of the new analogs had a greater
particularly interesting, A-62176 (1-(3-aminopyrrolidin-1-
DNA unwinding ability than A-62176, which was apparently
yl)-2-fluoro-4-oxo-4H-quino[2, 3, 4-ij][1, 4]benzoxazine-5-
related to higher potency in topoisomerase II inhibition and
carboxylic acid) and its amino-analog A-85226, reported in
increased cytotoxicity against tumor cells.
Fig (5). In both derivatives, the R substituent at position 1 is
an amino-pyrrolidine. Activity was retained and even The tested analogs showed strong cytotoxic effects on
increased when introducing an amino group at the 3 position several human and murine cell lines with IC50 values in the
of the condensed ring system (A-85226). range of 4-2,000 nM. They also showed potent inhibitory
effects against human topoisomerase II, with IC50 values in
R5 O the micromolar range.
F COOH
Mechanism of Action of Mammalian Topoisomerase II-
Directed Quinolones
N H
The discovery of the poisoning activity of quinolones
O
against eukaryotic topoisomerase II prompted a relevant
H2N number of studies devoted to clarify the molecular details of
this process. In fact, the drug interactions in the type II
R5 = H A-62176 topoisomerase-DNA complex are still ill defined, whereas
R5 = NH2 A-85226 the need for such a kind of information is multiple. These
new quinolone derivatives can yield a deep insight of what
Fig. (5). Chemical structure of quinobenzoxazines. occurs in the cleavage complex in the presence of type II

O O O

F COOH F COOH F COOH

R N R N R N

O O O

1 2 3

N
N or
R=
HN
H2N

Fig. (6). Chemical structure of pyridobenzophenoxazines.


446 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

topoisomerases of divergent species. A more precise model covalent complex as compared to the wild type enzyme and
for the ternary complex would be useful in designing new a reduced cleavage in the presence of CP-115, 953.
antibacterial derivatives safer for human use on one hand and
Although, it is not apparent whether tryptophan
new more potent congeners with anticancer activity on the
intercalation into the DNA double helix or a structural
other hand.
rearrangement of the enzyme conformation occurs in the
In this connection, CP-115,953 was the most widely mutant, it is suggested that the Ser-Thr mutation affects
investigated drug. In fact, as an antibacterial, it has the broa- mainly the DNA-protein interactions and that changes at the
dest activity spectrum of any other quinolone, being very enzyme/DNA interface might be critical also for the drug
effective toward both Gyr and Top4. In addition, it showed recognition process. Moreover, as it affects the sensitivity of
an activity higher than etoposide against any eukaryotic quinolone as well as of etoposide, the serine residue is likely
topoisomerases II examined to date [48, 53, 70-73]. (directly or indirectly) involved in a common (or at least
As previously mentioned, the starting point of this overlapping) site.
research was a possible similarity between etoposide and This serine residue is located in a conserved CAP homo-
quinolone topoisomerase poison mechanisms suggesting logy domain (a helix-turn-helix motif), a region involved in
parallel interactions in the cleavage complex [54]. This DNA interaction. The importance of this region was further
hypothesis was sustained by comparable DNA cleavage assessed producing more mutants in Saccharomyces
patterns produced by quinolone and etoposide in the cerevisiae type II topoisomerases. In particular, studies using
presence of different type II topoisomerases [37, 71]. Ser740Trp, Gln743Pro, and Thr744Pro mutants showed that
the α4-helix of this domain is critical for selecting the sites
As expected for topo II poisons, both drugs generate the
of DNA cleavage prior to cleavage of the G strand. They
typical 4 bases staggered double strand breaks on DNA.
also demonstrated that the solvent-exposed residues in the
Interestingly, sequence analysis of the drug-stimulated DNA
α4-helix are important in determining the binding as well as
cleavage showed that all test cytotoxic quinolones display a
preference for C at position –1 associated with a weaker the activity of quinolones/etoposide against topoisomerase
G preference at +5, this profile being shared by etoposide II-DNA complexes [78].
[74, 75]. Studies focused on the single catalytic steps of topoiso-
Cross-relations of these consensus sequences and drug merase II in the presence of etoposide/CP-115,953 have also
potency performed with several quinolones modified at been carried out.
positions 3 and 8 suggest possible interactions of specific Typical poisons of human Top2 like etoposide or m-
substituents on the quinolone derivatives with DNA and/or AMSA, stabilize the DNA-protein cleaved complex
topoisomerase. In particular, it is proposed that quinolones primarily by reducing the rate of topoisomerase II–mediated
may have at least one DNA-interactive region (involving DNA religation [79]. However, the behavior of quinolones
position 1 and/or 8) and at least one topoisomerase- with reference to eukaryotic Top2 is distinct, as they do not
interactive region (at position 3) [75]. impair the religation rate of cleaved DNA. In fact, the
quinolone-mediated stabilization of the cleavage complex is
Additionally, by in vitro mutagenesis followed by in vivo
due to increased levels of intermediates formed by an
selection, it was found that a single point mutation
increment of the enzyme’s forward rate of DNA cleavage
(His1012Tyr) can confer resistance both to CP-115, 953 and
[53]. Nonetheless, etoposide and CP-115,953 were found
to etoposide [48, 72]. As determined by DNA cleavage
assays, this mutant was resistant both prior to and following mutually exclusive in stimulating cleavage reaction as well
the DNA strand-passage event. In marked contrast, the as DNA strand passage [80].
mutant enzyme displayed sensitivity to amsacrine (a DNA Considering the enzyme-catalysed ATP hydrolysis step,
intercalating agent) comparable to the wild type and was it emerged that, although neither CP-115,953 nor etoposide
several folds hypersensitive to another intercalator, share an interaction domain with novobiocin, etoposide can
ellipticine. At first glance, it might appear that type II moderately inhibit ATPase activity [81]. This latter finding
topoisomerases can distinguish only between non- undercuts the common assumption that DNA cleavage-
intercalative and intercalative agents. enhancing drugs are clustered to the cleavage/religation steps
However, several lines of evidences indicate a non- of topoisomerase II.
complete identity of binding sites for quinolones and Furthermore, data on cell lines are available. In particular,
etoposide. Clear indications came from investigations the VpmR-5 line (an epipodophyllotoxin-resistant Chinese
performed in the presence of “designed” mutant enzymes. hamster ovary line) showed cross-resistance to CP-67,804
In E. Coli, high levels of quinolones resistance occur (approximately 3.7-fold) and CP-115,953 (approximately
1.3-fold). Although quinolone cross-resistance was less
when Ser83 in GyrA is mutated to Trp or Leu. Introduction
pronounced when compared to etoposide (approximately 12-
of the corresponding mutation in the human or yeast
topoisomerase II, leads to enzymes that acquire the ability to fold), this further confirms that topoisomerase II is a
physiological target for CP-67,804 and CP-115,953 in mam-
discriminate between quinolone and etoposide. In fact, a
Ser740Trp yeast or Ser763Trp human enzyme mutant malian cells [53].
become resistant to quinolones but hypersensitive to Altogether, these experimental observations led to a
etoposide (VP-16) [74, 76-77]. In particular, these purified model according to which quinolones and etoposide share
mutants show enhanced levels of etoposide-stabilized overlapping but not identical site on topoisomerase II [82].
The Quinolone Family Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 447

The role of direct DNA-drug interactions in modulating not apparent at present. Using a photocleavage assay,
potency and specificity is another interesting issue. As far as mismatched sequences, and competition experiments
quinolones, is well establish that they bind efficiently only between psorospermin and A-62176, the drug-binding site
single stranded DNA with the assistance of Mg2+ ions. It is to where enzyme poisoning occurs has been suggested to be
note that this metal ion is also required by topoisomerase to located on the DNA base pairs between positions +1 and +2
perform its catalytic cycle and to bind quinolone. Thus, it relative to the cleaved phosphodiester bonds. Based upon
appears that the metal cofactor participates in stabilizing the these results, the authors suggest that topo II induces
drug-cleavage complex interactions. Interestingly, etoposide transient structural distortion, possibly unwinding, of the
shows a similar behavior, since it also binds preferentially DNA that is captured or stabilized by the binding of A-
single stranded DNA in the presence of Mg2+ [83]. 62176 at the topo II cleavage gate.
Since quinobenzoxazines are quinolone derivatives In conclusion, the quinobenzoxazine represents an
designed to increment DNA affinity, a number of studies interesting novel pharmacophore with biological properties
were focused on their DNA-binding properties [84, 85]. Also distinct from those of antibacterial/cytotoxic quinolones.
in this case and similarly to their antibacterial congeners, A further line of research is based on the intriguing idea
DNA interaction was found to be dependent upon Mg2+.[26].
of combining the information obtained using mammalian and
It is proposed that the quinobenzoxazine binds duplex DNA
bacterial enzymes.
through intercalation in the presence of Mg2+, which bridges
the phosphate backbone of the DNA and the β-ketoacid unit Competition experiments clearly showed that, indeed, the
of the intercalated molecule. A 2:2 drug:Mg2+ complex drug interaction domain on type II topoisomerases has been
appears to form a "heterodimer complex" in which one drug conserved during the evolution from Gyr to Top4 and finally
molecule is intercalated into DNA and the second drug to Top2.
molecule is externally bound, held to the first molecule by Indication that similar interactions occur in the two types
two Mg2+ bridges, which themselves are chelated to of cleavage complex came also from the observation that
phosphates on DNA. A cooperativity in binding of the ciprofloxacin is actually able to displace both etoposide and
quinobenzoxazines to DNA, and a 4:4 drug:Mg2+ complex is CP-115,953 from their DNA-Top2 cleavage complex, thus
proposed, in which the two externally bound molecules from reducing DNA damage. This competitive character is
two different 2:2 dimers via π−π interactions. Further insight conserved in vivo as ciprofloxacin inhibited the cytotoxicity
into the metal ion mediated drug-DNA interaction has been of CP-115,953 and etoposide in mammalian cells to an
obtained by photochemical studies on A-62176 (Fig. 5). The extent that paralleled its in vitro attenuation of enzyme-
quinobenzoxazine undergoes photodecomposition to highly mediated cleavage [89].
fluorescent products. DNA accelerates the photochemical
decomposition of A-62176 up to 80-fold. This process In DNA gyrase, specific residues required to grant
requires Mg2+, duplex DNA, molecular oxygen, and efficient interaction with quinolones have been identified. A
intercalation of the drug into the DNA duplex. Furthermore, triple mutant of human Top2α in, which three amino acids
it was suggested that the precise positioning of the have been mutated to the corresponding residues present in
intercalated molecule, which is modulated by the structure gyrase, Met762Ser, Ser763Ala and Met766Asp showed a
and stereochemistry of the externally bound molecule, plays three-fold increase in sensitivity to ciprofloxacin in vitro and
an important role in determining the rate of photoreaction on similar sensitivities to a range of other quinolones. On the
DNA. Apparently, the photocleavage reaction occurs by a other hand, the activity of CP-115,953 was unaffected by any
free radical mechanism initiated by abstraction of the 4'- and of these mutations, whereas they conferred resistance to the
1'-hydrogens from the DNA minor groove, indicating that non-intercalating agent etoposide [90]. As reported above
the quinobenzoxazine-drug-Mg2+ complex is located on the and in agreement with these results, also a Thr(744)Pro
minor groove of the nucleic acid. Photochemical cleavage of substitution in the yeast type II topoisomerase was found to
DNA can occur in the presence of antibacterial render the enzyme sensitive to antibacterial quinolones [78].
fluoroquinolones but not with the same efficiency [86]. The above mutations occur in a relatively small region of
topoisomerase. Hence, this region is likely to be involved in
An investigation on the ability of the quinobenzoxazines determining the extent of interference of quinolones with the
to interfere with mammalian Top2 showed that they are topoisomerase II-DNA complexes. Thus, the presence of
potent inhibitors of this enzyme, both in vitro and in vivo defined amino acids at positions 762 to 767 appears to
[87]. According to this study, the quinobenzoxazines, contribute directly (with their side chains) or indirectly
surprisingly, appear to belong to the class of topo II (affecting the enzyme conformation) to create a binding site
suppressors that interfere with the catalytic activity of the for quinolone recognition.
enzyme at a step prior to the formation of the cleavage
complex. However, a subsequent study on A-62176, showed However, it is interesting to point out that subtle changes
that, at low drug concentrations and pH 6-7, the are actually occurring from one enzyme to the other in this
quinobenzoxazine enhances the formation of the cleavage overall conserved drug interaction domain. In fact, in
complex at certain sites but inhibits it at high drug prokaryotic type II topoisomerases (with maximal evidence
concentrations [88]. Conversely, at another topo II cleavage in gram positive Top4), the stabilization of the cleavage
site, A-62176 inhibits the cleaved complex formation at both complex by quinolones is insensitive to the presence of an
low and high drug concentrations. Hence, different aromatic moiety at position C-7 and is produced by an
mechanisms impairing topo II activity seem to be operating inhibition of the religation rate. On the opposite, quinolones
at different sites. The reason for this uncommon behavior is act against Top2 by accelerating the rate of DNA cleavage
448 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

and require aromatic substituents at C-7 [71]. Nonetheless, group is generally preserved in potent antibacterial
they operate against the sensitive Thr(744)Pro yeast Top2 quinolones.
closely resembling their action against prokaryotic enzymes,
2) Increase in the extent of aromatic/condensed rings is
supporting a conserved mechanisms of drug action along the
likely to increase the affinity of the quinolone for double
phylogenetic evolution.
stranded DNA, favoring stacking processes with the planar
structure produced by base pairing. Interestingly, classical
CONCLUSIONS quinolones interact better with single-stranded DNA and are
Quinolones can be considered as double-edged drugs: poorly effective on the B structure of the nucleic acid. They
depending upon fine details in their structure, they can are, therefore better suited to interfere with plasmids, in
switch their recognition of the cleavage complex from which distortion of base pairing occurs as a result of
bacteria to humans. Of course, this may raise concerns and supercoiling.
hopes. In fact, while cytotoxicity represents a serious In addition, structural changes that are likely to impair
drawback when pursuing antibacterial therapy, it becomes a bactericidal activity do not necessarily grant removal of the
desired property when developing anticancer agents. adverse effects known for antibacterial quinolones. Particular
Although exploitation of topoisomerase II is not new in the concern can be raised by the reported phototoxic properties
cancer field, the use of novel chemical entities is desirable to of the classical quinolones, which might represent an
increase the effectiveness of a treatment, in particular by additional drawback in anticancer therapy. However, this
reducing the onset of drug resistance. Indeed, pleiotropic unwanted side effect can be turned into an appreciable
mechanisms related to MDR are not likely to be overcome therapeutic tool when considering its potential applications
by antineoplastic quinolones, but these drugs might be useful in photodynamic therapy, as already proposed for
in the treatment of cancers with mutated top2s that have quinobenzoxazines [86].
become resistant to the traditionally used enzyme poisons.
When introducing structural changes into quinolones,
The knowledge thus far available on the mechanisms other cell-affecting mechanisms can be activated. This is
governing drug selectivity towards eukaryotic vs. indeed the case for the benzoxazines reported in Fig. (7)
prokaryotic species is as yet far from being fully understood. [91]. In fact, the extended planar system introduced with the
It appears that fine tuning processes occur by slightly aim of further stabilizing stacking interactions with DNA,
changing quinolone structure, that render quinolones more renders the new compounds available for efficient binding to
effective against the bacterial or the mammalian enzyme. G-quartet structures [92]. This, on turn, raised the possibility
The numerous modifications thus far examined give some that the enzyme telomerase, activated in the vast majority of
hints of the SAR for selectivity. tumor cells, might represent an additional target for
For anticancer quinolones, there is no need to preserve quinolone action and, hence, led to an improved
antibacterial efficacy. Hence, chemical functions that are not chemotherapeutic application of the new compounds.
suitable to modification in antibacterial chemotherapy can be In addition to practical applications, the possibility of
proficiently substituted to improve antineoplastic response. dual targeting for quinolones represents an effective
In fact, the SAR referring to antibacterial properties does not biochemical tool to dissect the molecular mechanism(s) of
simply hold for antitumor potency. In general, two major topoisomerase poisoning and to pinpoint similarities and
types of modification appear to drive the quinolone action differences produced by evolution in enzymes having a
towards human Top2: unique role in cell survival and proliferation.
1) Breakage of the zwitterionic properties of the In conclusion, quinolones represent a very valuable
compounds by modifying either the C-7 basic substituent or family of multi-faceted drugs, the chemical synthesis of,
the C-3 carboxyl group, or both will change the electron which is flexible and can be easily adapted to prepare new
density distribution of the quinolone and its protonation congeners with rationally devised structures. This is
equilibria. It is interesting to note that the keto-carboxyl witnessed by the description of many thousands of

O O O O
F F
O O

N N N N
O O
H2N H2N
O

QQ58 FQA-CR

Fig. (7). Chemical structure of fluoroquinophenoxazine (QQ58) and fluoroquinoanthoxazine (FQA-CR).


The Quinolone Family Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 449

derivatives in the literature, and has allowed, and still allows [41] Takei, M.; Fukuda, H.; Yasue, T.; Hosaka, M.; Oomori, Y.
to-day, to accurately describe their QSAR, which is essential Antimicrob. Agents Chemother., 1998, 42, 2678.
[42] Dobson, R.A.; O'Connor, J.R.; Poulin, S.A.; Kundsin, R.B.; Smith,
for effective drug design and optimization, and to discover T.F.; Came, P.E. Antimicrob. Agents Chemother., 1980, 18, 738.
new and unprecedented activities, which makes quinolones [43] Carabateas, P.M.; Brundage, R.P.; Gellote, M.D.; Lorenz, R.R.;
as yet endless source of (pleasant) surprises and, hopefully, Opalka, C.J.; Thielking, W.H., Williams, G.L.; Lesher, G.Y. J.
of further clinically useful drugs. Heterocycl. Chem., 1984, 21, 1875.
[44] Barrett, J.F.; Gootz, T.D.; McGuirk, P.R.; Farrell, C.A.;
Sokolowski, S.A. Antimicrob. Agents Chemother., 1989, 33, 1697.
REFERENCES [45] Peterson, L.R. Clin. Infect. Dis., 2001, 33 Suppl 3, S180.
[46] Robinson, M.J.; Martin, B.A.; Gootz, T.D.; McGuirk, P.R.;
[1] Lesher, G.Y. J. Med. Pharm. Chem., 1962, 5, 1063.
Osheroff, N. Antimicrob. Agents Chemother., 1992, 36, 751.
[2] Kim, O.K.; Ohemeng, K.; Barrett, J.F. Expert Opin. Investig.
[47] Arakawa, H.; Mano, E.; Hakoda, N.; Yoshinari, T.; Nakagawa, S.;
Drugs, 2001, 10, 199.
Okura, A. Anticancer Drug Des., 1996, 11, 221.
[3] Zhanel, G.G.; Ennis, K.; Vercaigne, L.; Walkty, A.; Gin, A.S.;
Embil, J.; Smith, H.; Hoban, D.J. Drugs, 2002, 62, 13. [48] Elsea, S.H.; McGuirk, P.R.; Gootz, T.D.; Moynihan, M.; Osheroff,
N. Antimicrob. Agents Chemother., 1993, 37, 2179.
[4] Heisig, P. Planta Med., 2001, 67, 3.
[49] Yamashita, Y.; Ashizawa, T.; Morimoto, M.; Hosomi, J.; Nakano,
[5] Wang, J.C. Annu. Rev. Biochem., 1985, 54, 665.
H. Cancer Res., 1992, 52, 2818.
[6] Higgins, P.G.; Fluit, A.C.; Schmitz, F.J. Curr. Drug Targets, 2003,
4, 181. [50] Suto, M.J.; Domagala, J.M.; Roland, G.E.; Mailloux, G.B.; Cohen,
[7] Gellert, M.; Mizuuchi, K.; O'Dea, M.H.; Nash, H.A. Proc. Natl. M.A. J. Med. Chem., 1992, 35, 4745.
[51] Alovero, F.L.; Pan, X.S.; Morris, J.E.; Manzo, R.H.; Fisher, L.M.
Acad. Sci. U. S. A., 1976, 73, 3872.
Antimicrob. Agents Chemother., 2000, 44, 320.
[8] Reece, R.J.; Maxwell, A. Crit. Rev. Biochem. Mol. Biol., 1991, 26,
[52] Gootz, T.D.; McGuirk, P.R.; Moynihan, M.S.; Haskell, S.L.
335.
[9] Kato, J.; Nishimura, Y.; Yamada, M.; Suzuki, H.; Hirota, Y. J Antimicrob. Agents Chemother., 1994, 38, 130.
[53] Robinson, M.J.; Martin, B.A.; Gootz, T.D.; McGuirk, P.R.;
Bacteriol, 1988, 170, 3967.
Moynihan, M.; Sutcliffe, J.A.; Osheroff, N. J. Biol. Chem., 1991,
[10] Kato, J.; Nishimura, Y.; Imamura, R.; Niki, H.; Hiraga, S.; Suzuki,
266, 14585.
H. Cell, 1990, 63, 393.
[11] Peng, H.; Marians, K.J. J. Biol. Chem., 1993, 268, 24481. [54] Wentland, M.P.; Lesher, G.Y.; Reuman, M.; Gruett, M.D.; Singh,
B.; Aldous, S.C.; Dorff, P.H.; Rake, J.B.; Coughlin, S.A. J. Med.
[12] Marians, K.J.; Hiasa, H. J. Biol. Chem., 1997, 272, 9401.
[13] Williams, N.L.; Howells, A.J.; Maxwell, A. J. Mol. Biol., 2001, Chem., 1993, 36, 2801.
[55] Fang, K.C.; Chen, Y.L.; Sheu, J.Y.; Wang, T.C.; Tzeng, C.C. J.
306, 969.
[14] Morrison, A.; Cozzarelli, N.R. Cell, 1979, 17, 175. Med. Chem., 2000, 43, 3809.
[15] Kampranis, S.C.; Bates, A.D.; Maxwell, A. Proc. Natl. Acad. Sci. [56] Eissenstat, M.A.; Kuo, G.H.; Weaver, I.J.; Wentland, M.P.;
U. S. A., 1999, 96, 8414. Robinson, R.G.; Klingbeil, K.M.; Danz, D.W.; Corbett, T.H.;
Coughlin, S.A. Bioorg. Med. Chem. Lett., 1995, 5, 1021.
[16] Ullsperger, C., Cozzarelli, N.R. J. Biol. Chem., 1996, 271, 31549.
[57] Kohlbrenner, W.E.; Wideburg, N.; Weigl, D.; Saldivar, A.; Chu,
[17] Levine, C.; Hiasa, H.; Marians, K.J. Biochim. Biophys. Acta., 1998,
1400, 29. D.T. Antimicrob. Agents Chemother., 1992, 36, 81.
[18] Khodursky, A.B.; Peter, B.J.; Schmid, M.B.; DeRisi, J.; Botstein, [58] DoganKoruznjak, J.; Slade, N.; Zamola, B.; Pavelic, K.;
Karminski-Zamola, G. Chem. Pharm. Bull. (Tokyo), 2002, 50, 656.
D.; Brown, P.O.; Cozzarelli, N.R. Proc. Natl. Acad. Sci. U. S. A.,
[59] Tomita, K.; Tsuzuki, Y.; Shibamori, K.; Tashima, M.; Kajikawa,
2000, 97, 9419.
[19] Deibler, R.W.; Rahmati, S.; Zechiedrich, E.L. Genes. Dev., 2001, F.; Sato, Y.; Kashimoto, S.; Chiba, K.; Hino, K. J. Med. Chem.,
2002, 45, 5564.
15, 748.
[20] Willmott, C.J.; Critchlow, S.E.; Eperon, I.C.; Maxwell, A. J. Mol. [60] Wentland, M.P.; Lesher, G.Y.; Reuman, M.; Pilling, G.M.;
Saindane, T.; Perni, R.B.; Eissenstat, M.A.; Weaver, I.J.; Singh, B.;
Biol., 1994, 242, 351.
[21] Wentzell, L.M.; Maxwell, A. J. Mol. Biol., 2000, 304, 779. Rake, J.; Coughlin, S.A. Bioorg. Med. Chem. Lett., 1993, 3, 1711.
[61] Coughlin, S.A.; Danz, D.W.; Robinson, R.G.; Klingbeil, K.M.;
[22] Shea, M.E.; Hiasa, H. J. Biol. Chem., 2000, 275, 14649.
Wentland, M.P.; Corbett, T.H.; Waud, W.R.; Zwelling, L.A.;
[23] Yoshida, H.; Bogaki, M.; Nakamura, M.; Nakamura, S.
Antimicrob. Agents Chemother., 1990, 34, 1271. Altschuler, E.; Bales, E.; Rake, J.B. Biochem. Pharmacol., 1995,
50, 111.
[24] Piddock, L.J. Drugs, 1999, 58 Suppl 2, 11.
[25] Heddle, J.G.; Barnard, F.M.; Wentzell, L.M.; Maxwell, A. [62] Gmunder, H.; Kuratli, K.; Keck, W. Nucleic Acids Res., 1997, 25,
604.
Nucleosides Nucleotides Nucleic Acids, 2000, 19, 1249.
[26] Palu, G.; Valisena, S.; Ciarrocchi, G.; Gatto, B.; Palumbo, M. Proc. [63] Corbett, A.H.; Hong, D.; Osheroff, N. J. Biol. Chem., 1993, 268,
14394.
Natl. Acad. Sci. U. S. A., 1992, 89, 9671.
[27] Sissi, C.; Perdona, E.; Domenici, E.; Feriani, A.; Howells, A.J.; [64] Wentland, M.P.; Aldous, S.C.; Guett, M.D.; Perni, R.B.; Poeles,
R.G.; Danz, D.W.; Klingbeil, K.M.; Peverly, A.D.; Robinson, R.G.;
Maxwell, A.; Palumbo, M. J. Mol. Biol., 2001, 311, 195.
[28] Shen, L.L.; Pernet, A.G. Proc. Natl. Acad. Sci. U. S. A., 1985, 82, Corbett, T.H.; Rake, J.; Coughlin, S.A. Bioorg. Med. Chem. Lett.,
1995, 5, 405.
307.
[65] Chu, D.T.; Hallas, R.; Clement, J.J.; Alder, J.; McDonald, E.;
[29] Critchlow, S.E.; Maxwell, A. Biochemistry, 1996, 35, 7387.
[30] Kampranis, S.C.; Maxwell, A. J. Biol. Chem., 1998, 273, 22606. Plattner, J.J. Drugs Exp. Clin. Res., 1992, 18, 275.
[66] Chu, D.T.; Hallas, R.; Tanaka, S.K.; Alder, J.; Balli, D.; Plattner,
[31] Maxwell, A. Nat. Struct. Biol., 1996, 3, 109.
[32] Berger, J.M.; Gamblin, S.J.; Harrison, S.C.; Wang, J.C. Nature, J.J. Drugs Exp. Clin. Res., 1994, 20, 177.
[67] Clement, J.J.; Burres, N.; Jarvis, K.; Chu, D.T.; Swiniarski, J.;
1996, 379, 225.
Alder, J. Cancer Res., 1995, 55, 830.
[33] Berger, J.M.; Fass, D.; Wang, J.C.; Harrison, S.C. Proc. Natl.
Acad. Sci. U. S. A., 1998, 95, 7876. [68] Kwok, Y.; Sun, D.; Clement, J.J.; Hurley, L.H. Anticancer Drug
Des., 1999, 14, 443.
[34] Chen, G.L.; Yang, L.; Rowe, T.C.; Halligan, B.D.; Tewey, K.M.;
Liu, L.F. J. Biol. Chem., 1984, 259, 13560. [69] Zeng, Q.; Kwok, Y.; Kerwin, S.M.; Mangold, G.; Hurley, L.H. J.
Med. Chem., 1998, 41, 4273.
[35] Woese, C.R. Microbiol. Rev., 1987, 51, 221.
[70] Anderson, V.E.; Gootz, T.D.; Osheroff, N. J. Biol. Chem., 1998,
[36] Forterre, P.; Nadal, M.; Elie, C. Syst. Appl. Microbiol., 1986, 7, 67.
273, 17879.
[37] Sioud, M.; Forterre, P. Biochemistry, 1989, 28, 3638.
[71] Anderson, V.E.; Zaniewski, R.P.; Kaczmarek, F.S.; Gootz, T.D.;
[38] Hussy, P.; Maass, G.; Tummler, B.; Grosse, F.; Schomburg, U.
Osheroff, N. J. Biol. Chem., 1999, 274, 35927.
Antimicrob. Agents Chemother., 1986, 29, 1073.
[72] Elsea, S.H.; Hsiung, Y.; Nitiss, J.L.; Osheroff, N. J. Biol. Chem.,
[39] Hoshino, K.; Sato, K.; Une, T.; Osada, Y. Antimicrob. Agents
Chemother., 1989, 33, 1816. 1995, 270, 1913.
[73] Spitzner, J.R.; Chung, I.K.; Gootz, T.D.; McGuirk, P.R.; Muller,
[40] Akasaka, T.; Kurosaka, S.; Uchida, Y.; Tanaka, M.; Sato, K.;
M.T. Mol. Pharmacol., 1995, 48, 238.
Hayakawa, I. Antimicrob. Agents Chemother., 1998, 42, 1284.
450 Curr. Med. Chem. – Anti-Cancer Agents, 2003, Vol. 3, No. 6 Sissi and Palumbo

[74] Strumberg, D.; Nitiss, J.L.; Dong, J.; Kohn, K.W.; Pommier, Y. J. [83] Chow, K.C.; Macdonald, T.L.; Ross, W.E. Mol. Pharm., 1988, 34,
Biol. Chem., 1999, 274, 28246. 467.
[75] Huff, A.C.; Robinson, R.G.; Evans, A.C.; Selander, K.N.; [84] Fan, J.Y.; Sun, D.; Yu, H.; Kerwin, S.M.; Hurley, L.H. J. Med.
Wentland, M.P.; Rake, J.B.; Coughlin, S.A. Anticancer Drug Des., Chem., 1995, 38, 408.
1995, 10, 251. [85] Yu, H.; Hurley, L.H.; Kerwin, S.M. J. Am. Chem. Soc., 1996, 118,
[76] Strumberg, D.; Nitiss, J.L.; Rose, A.; Nicklaus, M.C.; Pommier, Y. 7040.
J. Biol. Chem., 1999, 274, 7292. [86] Yu, H.; Kwok, Y.; Hurley, L.H.; Kerwin, S.M. Biochemistry, 2000,
[77] Hsiung, Y.; Elsea, S.H.; Osheroff, N.; Nitiss, J.L. J. Biol. Chem., 39, 10236.
1995, 270, 20359. [87] Permana, P.A.; Snapka, R.M.; Shen, L.L.; Chu, D.T.; Clement, J.J.;
[78] Strumberg, D.; Nitiss, J.L.; Dong, J.; Walker, J.; Nicklaus, M.C.; Plattner, J.J. Biochemistry, 1994, 33, 11333.
Kohn, K.W.; Heddle, J.G.; Maxwell, A.; Seeber, S.; Pommier, Y. [88] Kwok, Y.; Zeng, Q.; Hurley, L.H. J. Biol. Chem., 1999, 274,
Antimicrob. Agents. Chemother., 2002, 46, 2735. 17226.
[79] Robinson, M.J.; Osheroff, N. Biochemistry, 1991, 30, 1807. [89] Elsea, S.H.; Westergaard, M.; Burden, D.A.; Lomenick, J.P.;
[80] Corbett, A.H.; Guerry, P.; Pflieger, P.; Osheroff, N. Antimicrob. Osheroff, N. Biochemistry, 1997, 36, 2919.
Agents Chemother., 1993, 37, 2599. [90] Hammonds, T.R.; Foster, S.R.; Maxwell, A. J. Mol. Biol., 2000,
[81] Robinson, M.J.; Corbett, A.H.; Osheroff, N. Biochemistry, 1993, 300, 481.
32, 3638. [91] Kim, M.Y.; Duan, W.; Gleason-Guzman, M.; Hurley, L.H. J. Med.
[82] Osheroff, N.; Corbett, A.H.; Elsea, S.H.; Westergaard, M. Cancer Chem., 2003, 46, 571.
Chemother. Pharmacol., 1994, 34 Suppl, S19. [92] Neidle, S.; Parkinson, G. Nature Rev. Drug Discovery, 2002, 1,
383.

You might also like