You are on page 1of 19

molecules

Review
The Current Case of Quinolones: Synthetic
Approaches and Antibacterial Activity
Abdul Naeem 1 , Syed Lal Badshah 1,2, *, Mairman Muska 1 , Nasir Ahmad 2 and Khalid Khan 2

1 National Center of Excellence in Physical Chemistry, University of Peshawar, Peshawar,


Khyber Pukhtoonkhwa 25120, Pakistan; naeeem64@yahoo.com (A.N.); mmuskachem@yahoo.com (M.M.)
2 Department of Chemistry, Islamia College University Peshawar, Peshawar, Khyber Pukhtoonkhwa 25120,
Pakistan; nasirshah121@yahoo.com (N.A.); drkhalidchem@yahoo.com (K.K.)
* Correspondence: shahbiochemist@gmail.com; Tel.: +92-331-931-6672

Academic Editor: Peter J. Rutledge


Received: 23 December 2015 ; Accepted: 15 February 2016 ; Published: 28 March 2016

Abstract: Quinolones are broad-spectrum synthetic antibacterial drugs first obtained during the
synthesis of chloroquine. Nalidixic acid, the prototype of quinolones, first became available for clinical
consumption in 1962 and was used mainly for urinary tract infections caused by Escherichia coli and
other pathogenic Gram-negative bacteria. Recently, significant work has been carried out to synthesize
novel quinolone analogues with enhanced activity and potential usage for the treatment of different
bacterial diseases. These novel analogues are made by substitution at different sites—the variation
at the C-6 and C-8 positions gives more effective drugs. Substitution of a fluorine atom at the C-6
position produces fluroquinolones, which account for a large proportion of the quinolones in clinical
use. Among others, substitution of piperazine or methylpiperazine, pyrrolidinyl and piperidinyl rings
also yields effective analogues. A total of twenty six analogues are reported in this review. The targets
of quinolones are two bacterial enzymes of the class II topoisomerase family, namely gyrase and
topoisomerase IV. Quinolones increase the concentration of drug-enzyme-DNA cleavage complexes
and convert them into cellular toxins; as a result they are bactericidal. High bioavailability, relative
low toxicity and favorable pharmacokinetics have resulted in the clinical success of fluoroquinolones
and quinolones. Due to these superior properties, quinolones have been extensively utilized and this
increased usage has resulted in some quinolone-resistant bacterial strains. Bacteria become resistant
to quinolones by three mechanisms: (1) mutation in the target site (gyrase and/or topoisomerase IV)
of quinolones; (2) plasmid-mediated resistance; and (3) chromosome-mediated quinolone resistance.
In plasmid-mediated resistance, the efflux of quinolones is increased along with a decrease in
the interaction of the drug with gyrase (topoisomerase IV). In the case of chromosome-mediated
quinolone resistance, there is a decrease in the influx of the drug into the cell.

Keywords: quinolones; analogues; gyrase; topoisomerase IV; resistance

1. Introduction
In the early 20th century, infectious diseases were the most common cause of human illness
leading to death, out of which bacterial infections accounted for about one-third of these infections [1].
In addition to bacterial infections, malaria also has an enormous impact on global human health, with
over 200 million cases of malaria and over 600,000 deaths each year [2]. While different therapies were
used for the treatment of these diseases, the introduction of antibiotic agents opened new avenues for
the treatment of bacterial infections. The invention of antibiotics such as sulfonamide, and afterward
penicillin by Alexander Fleming in 1928 and synthetic antibiotics such as quinolones were some of the

Molecules 2016, 21, 268; doi:10.3390/molecules21040268 www.mdpi.com/journal/molecules


Molecules 2016, 21, 268 2 of 19

Molecules 2016, 21, 268 2 of 18


great milestones of science [3,4]. However, with progress in the development of antibiotics has come
increasingly
increasingly widespread
widespread antibiotic-resistant
antibiotic-resistant pathogens,
pathogens, which which represent
represent an an unprecedented
unprecedented health health
issue [5].
issue [5].
Here,
Here, we we focus
focus onon an
an important
important class
class of
of synthetic
synthetic antibiotics
antibiotics known
known as as quinolones,
quinolones, thatthat constitute
constitute
an
an important class of biologically-active broad-spectrum antibacterial drugs [6]. The spectacular
important class of biologically-active broad-spectrum antibacterial drugs [6]. The spectacular
discovery
discovery of of nalidixic
nalidixicacid,
acid,which
whichisischemically
chemicallya naphthyridone
a naphthyridone and and
thethe prototype
prototype compound
compound of
of the
the quinolones as a byproduct in chloroquine synthesis (Figure 1) by
quinolones as a byproduct in chloroquine synthesis (Figure 1) by Lesher and coworkers in 1962Lesher and coworkers in 1962
initiated
initiated the
the improvement
improvement of of quinolones
quinolones [7,8].
[7,8]. It
It is
is present
present inin aa number
number of of biologically-active
biologically-active natural
natural
products. Because of its valuable medicinal activities, quinolones
products. Because of its valuable medicinal activities, quinolones have attracted a lot have attracted a lot
of of attention
attention in
in
thethe field
field of medicinal
of medicinal andand synthetic
synthetic chemistry
chemistry [9].
[9]. In theIn the1960s,
later later 1960s,
nalidixicnalidixic
acid cameacidinto
came into
clinical
clinical
use for theusecure
for the cure oftract
of urinary urinary tract caused
infections infections causedbacteria
by enteric by enteric[10].bacteria
Around[10]. Around
the 1970s, the
several
1970s, several new generations of quinolones, with oxolinic acid being
new generations of quinolones, with oxolinic acid being the most prominent, had been synthesized the most prominent, had
been synthesized
and became and became
available available
for clinical usefor clinical Until
[10–12]. use [10–12]. Until 1980, quinolones
1980, quinolones were a less-used
were a less-used class of
class of antibacterial agents and later a second generation of quinolones
antibacterial agents and later a second generation of quinolones was developed. In the quest was developed. In the
for
quest
potent quinolones, it was discovered that alteration with different groups at positions C-6 and C-6
for potent quinolones, it was discovered that alteration with different groups at positions C-8
and
givesC-8 gives
more more effective
effective antibacterial
antibacterial analogues analogues
[13]. The[13].mostThe most significant
significant changes in changes
the basicin the basic
skeleton
skeleton
of quinolones are the substitution of fluorine at C-6 and a key ring substituent piperazine or
of quinolones are the substitution of fluorine at C-6 and a key ring substituent piperazine or
methylpiperazine,
methylpiperazine, pyrrolidinyl
pyrrolidinyland andpiperidinyl.
piperidinyl. DueDueto thetoinsertion of fluorine
the insertion at C-6, most
of fluorine clinically
at C-6, most
used quinolones
clinically are fluoroquinolones
used quinolones (FQ) [14–16].
are fluoroquinolones (FQ) [14–16].

O O

OH

N N

Nalidixic acid

Figure 1.
Figure 1. Chemical
Chemical structure
structure of
of nalidixic
nalidixic acid.
acid.

Fluoroquinolones (formally called simply quinolones) are characterized as broad-spectrum


Fluoroquinolones (formally called simply quinolones) are characterized as broad-spectrum
antibacterial drugs active against both Gram positive and Gram negative bacteria. Besides
antibacterial drugs active against both Gram positive and Gram negative bacteria.
broad-spectrum activity, the success of fluoroquinolones can be attributed to their properties such as
Besides broad-spectrum
good bioavailability activity,
after oral the success
administration, of fluoroquinolones
relatively low toxicity andcan be attributed
favorable to their
pharmacokinetics
properties such adverse
[17]. Although as good bioavailability
events still happen after oral
while administration, relatively low these
utilizing fluoroquinolones, toxicityareand
stillfavorable
some of
pharmacokinetics [17]. Although adverse events still happen while
the major antimicrobial agents and a lot of work has gone into the structural evolution in the utilizing fluoroquinolones,
these are still
framework ofsome of the major to
fluoroquinolones antimicrobial
make novel agents analoguesandwitha lotimproved
of work has gone into
potency. the structural
Fluoroquinolones
evolution in the framework of fluoroquinolones to make novel analogues
have been primarily utilized for the treatment of urinary tract infections (UTI), respiratory with improved potency.
tract
Fluoroquinolones have been primarily utilized for the treatment of urinary
infections, sexually transmitted diseases, gastrointestinal and abdominal infections, skin and soft tract infections (UTI),
respiratory tract infections,
tissue infections sexually
and infections of bonetransmitted
and joints diseases,
[11,18]. gastrointestinal and abdominal
Besides these functions, infections,
fluoroquinolones
skin
with broad spectrum activity have been widely utilized in veterinary medicine to treatfunctions,
and soft tissue infections and infections of bone and joints [11,18]. Besides these bacterial
fluoroquinolones with broad spectrum activity have been widely utilized
infections in food-producing animals, aquaculture and pets. Many fluoroquinolones are available on in veterinary medicine to
treat bacterial infections in food-producing animals, aquaculture and pets.
the market; however the use of these antibacterial agents highly depends on the animal species andMany fluoroquinolones are
available on the market;
their geographical however[18].
distribution the They
use ofhave
theseaantibacterial
wide rangeagents highlyapplication
of medical depends oninthe animal
livestock,
species
poultry,andfishtheir
andgeographical
domestic animal distribution
in the [18]. They have
treatment a wide range
and prevention of ofrespiratory,
medical application
enteric and in
livestock, poultry, fish and domestic animal in the treatment and prevention
complex urinary tract infections [19]. Some quinolones such as 6-chloro-7-methoxy-4(1H)-quinolones of respiratory, enteric and
complex
have good urinary tract infections
antimalarial activities,[19].
with Some
goodquinolones such asmultiple
activity against 6-chloro-7-methoxy-4(1H)-quinolones
stages of Plasmodium infection
have good antimalarial
[2]. Antimalarial activityactivities,
was alsowithshown good activity against multiple
in (1H)-quinolone, stages of Plasmodium
and its tetrahydroacridine infectionwith
analogue, [2].
Antimalarial activity was also shown in (1H)-quinolone,
substitution of various benzenoid ring features and aryl moieties [20]. and its tetrahydroacridine analogue, with
substitution of various benzenoid ring features and aryl moieties [20].
2. Recent Developments in the Synthesis of Quinolones
Quinolones are very important in medicinal chemistry as they have broad spectrum antibacterial
activity, and they acts as anticancer and antimalarial agents. A lot of work has been carried out on
the synthesis of quinolones to develop more effective and economical analogues. A number of
different methods are reported for the synthesis of quinolones. In general these, procedures involve
Molecules 2016, 21, 268 3 of 19

2. Recent Developments in the Synthesis of Quinolones


Quinolones are very important in medicinal chemistry as they have broad spectrum antibacterial
activity, and they acts as anticancer and antimalarial agents. A lot of work has been carried out
on the synthesis of quinolones to develop more effective and economical analogues. A number of
Molecules 2016, 21, 268 3 of 18
different methods
Molecules 2016, 21, 268 are reported for the synthesis of quinolones. In general these, procedures involve 3 of 18
the utilization
the utilization of of aa preactivated
preactivated substrate,
substrate, aa multistep
multistep strategy
strategy and
and harsh
harsh reaction
reaction conditions,
conditions, which
which
limit the compatibility
compatibility
the utilization
limit the and range
of a preactivated
and rangesubstrate,
of the
of the functional
functional groups
a multistep involved
strategy
groups in these
and harsh
involved in these reactions
reaction [9]. The
The use
conditions,
reactions [9]. use of
which of
transition
limit the metal catalysts
compatibility and has also
range ofbeen
the utilized
functionalfor the
groupssynthesis
involved of quinolones
in these
transition metal catalysts has also been utilized for the synthesis of quinolones and their analogues. and
reactions their
[9]. analogues.
The use of
The
The catalytic
transition
catalytic activation
metal catalysts
activation of the
of has C-H
the also
C-H beenbond allows
bondutilized the synthesis
for synthesis
allows the the synthesisof a
of a ofremarkable
quinolones
remarkable variety of
and their
variety heterocyclic
analogues.
of heterocyclic
compounds,
The catalytic including
activation quinolones
of the C-H [21].
bond The
allowsfollowing
the are
synthesis some
of a
compounds, including quinolones [21]. The following are some of the latest reported data on of the
remarkablelatest reported
variety of data on the
heterocyclicthe
development of
compounds, including
development new methods
quinolones
of new methods for the
for the synthesis
[21]. of quinolones
The following
synthesis and
are some
of quinolones their derivatives.
of thederivatives.
and their latest reported data on the
Hu et
development
Hu et al.
al. developed
developed
of new methodsaa novel
novel method
formethod in which
the synthesis
in which aa broad
broad range
of quinolones range
andof 2-arylquinoline-4(1H)-ones
oftheir derivatives.
2-arylquinoline-4(1H)-ones were
were
synthesized
Hu et by
al. a metal-free
developed a oxidative
novel method intermolecular
in which a Mannich
broad reaction
range of
synthesized by a metal-free oxidative intermolecular Mannich reaction between secondary amines between secondary
2-arylquinoline-4(1H)-ones amines and
were
unmodified
synthesized
and unmodified ketones (Scheme
by aketones
metal-free 1). The
oxidative
(Scheme advantage of
1). Theintermolecular this
advantage of this method
Mannich is
method that it
reaction does not
is that itbetweenrequire the
does notsecondary use of
useany
require theamines of
transition
any metal
and transition
unmodified catalyst;
ketones
metal andand
(Scheme
catalyst; it occurs under
it1).occurs
The mild
advantage
under conditions.
mild This
of conditions.
this method ismethod
This that cancan
it does
method produce
notproduce
requirea number
athe use of
number
analogues
any
of analogues in in
transition very
metal
veryhigh yield
catalyst;
high [6].[6].
and
yield it occurs under mild conditions. This method can produce a number
of analogues in very high yield [6].
O O Transition metal free
TEMPO (2equiv)
O R' O R' Direct sp3 C-H
Transition metal
/ sp 3 C-H coupling
free
KOtBu (2equiv)
TEMPO (2equiv) 3
R R' R R' Direct substrate
Broad sp3 C-H coupling
sp C-H / scope
KOtBu (0.05M)
DMSO (2equiv)
R N Ar 800C (0.05M)
, 6hr R N Ar Broad substrate scope
H DMSO H Simple and mild condition
N Ar 800C , 6hr N Ar Simple and mild condition
H H Excellent yields
20 examples
Excellent yields
20 examples
upto 98% yield
upto 98% yield
Scheme 1. Metal-free oxidative intermolecular Mannich reaction for the synthesis of 2-arylquinone-
Scheme 1. Metal-free oxidative intermolecular Mannich reaction for the synthesis of
Scheme 1. Metal-free
4(1H)-ones oxidative
from secondary intermolecular
amines and ketones Mannich
[6]. reaction for the synthesis of 2-arylquinone-
2-arylquinone-4(1H)-ones from secondary amines and ketones [6].
4(1H)-ones from secondary amines and ketones [6].
Kang and Hong performed alkynylation of 4-quinolones at three different positions for making
Kang
Kang and
quinolones and Hong
Hong performed
analogues. In the firstalkynylation
performed alkynylation
method, theyof
of 4-quinolones
4-quinolones at
at three
utilized TIPS-EBX (andifferent
three different
alkylatingpositions
agent) for
positions for making
making
to develop
quinolones
quinolones
an analogues.
analogues.
efficient and In the
In the
site-selective first
C5firstmethod, they
method, they
alkynylation utilized TIPS-EBX
utilized TIPS-EBX
of 4-quinolones (an
(Scheme (an alkylating
alkylating
2). In agent)
the second to
agent) develop an
to develop
strategy a Ru(II)
efficient and site-selective
an efficientC2and
catalyzed site-selective
selective C5 alkynylation
C5 alkynylation
alkynylation of 4-quinolones
of 4-quinolones
was achieved (Scheme 2). In the second
(Scheme 2).group-directed
through N-pyrimidyl strategy
In the second strategy a Ru(II)
a Ru(II)
cross-couplings
catalyzed
catalyzed C2
C2 selective
selective alkynylation
alkynylation was
was achieved
achieved through
through N-pyrimidyl
N-pyrimidyl group-directed
group-directed
to obtained useful C2-alkynylated 4-quinolones, while isoquinolones were developed through C3 cross-couplings
cross-couplings
to
to obtained
obtained useful
alkynylation. These C2-alkynylated
useful C2-alkynylated
three 4-quinolones,
4-quinolones,
different routes of producingwhile
while isoquinolones
isoquinolones
quinolones analogueswereare
were developed
developed through
through
site selective C3
C3
methods
alkynylation.
alkynylation. These
These three
three different
different routes
routes of
of
and useful for obtaining effective derivatives [22]. producing
producing quinolones
quinolones analogues
analogues are
are site
site selective
selective methods
methods
and
and useful
useful for
for obtaining
obtaining effective
effective derivatives
derivatives [22].
[22].
SiiPr3
SiiPr3
O H O TIPS-EBX
TIPS-EBX
O H5 O TIPS-EBX
[Ru] or [Rh]
[Rh]
TIPS-EBX 5
[Ru] or [Rh]
N R=[Rh]
alkyl N 2 H
R= Pyrimidyl
N
R= alkyl R= Pyrimidyl
N
R RN 2 H RN SiiPr3
R R R SiiPr3
Scheme 2. Site selective C-H alkynylation of Quinolones [22].
Scheme 2. Site selective C-H alkynylation of Quinolones [22].
Scheme 2. Site selective C-H alkynylation of Quinolones [22].
Satio et al. synthesized chiral substituted 2,3-dihydro-4-quinolones in high yields using an
Satio et addition
aza-Michael al. synthesized
reactionchiral substituted
(Scheme 3). The 2,3-dihydro-4-quinolones
advantage of this methodinishigh yields using an
the enantioselective
Satio
production et al.
aza-Michaelofadditionsynthesized
compounds reactionchiral substituted
and (Scheme 3). The
some of the 2,3-dihydro-4-quinolones
advantage
analogues can beof purified
this method in high yields using an
is the recrystallization
in a single enantioselective
aza-Michael
production
step. ofaddition
The chiral compoundsreaction
and (Scheme
some of 3).
2,3-dihydro-4-quinolones canThe
the advantage
analogues
be used can
for of
thebe this method
purified
preparation is theuseful
inofa other
single enantioselective
recrystallization
quinolone
production of
step. The chiral
analogues. compounds and some
2,3-dihydro-4-quinolones
The 2-substituted of the analogues
can be used
2,3-dihydro-4-quinolones can be
formitosis
halt purified
the preparation in a single
of other
and are useful recrystallization
usefulagents
antitumor quinolone
[23].
step. The chiral
analogues. 2,3-dihydro-4-quinolones
The 2-substituted can be used
2,3-dihydro-4-quinolones formitosis
halt the preparation of other
and are useful usefulagents
antitumor quinolone
[23].
analogues. The 2-substituted 2,3-dihydro-4-quinolones halt mitosis and X are useful antitumor
O O
agents [23]. X
O
O O O
Catalyst P O
benzene / C6H12 O
O
NH2 R
Catalyst
v/v = 1/1 N R P OH
benzene / C6H12 H O OH
NH2 R v/v 0 1/1 N R X
PG free amine 70=C H
upto quantitative yield
PG free amine 700C uptoup to 94% eeyield
quantitative X= C6F5 X
up to 94% ee X= C6F5
Catalyst
Scheme 2. Site selective C-H alkynylation of Quinolones [22].

Satio et al. synthesized chiral substituted 2,3-dihydro-4-quinolones in high yields using an


aza-Michael addition reaction (Scheme 3). The advantage of this method is the enantioselective
production of compounds and some of the analogues can be purified in a single recrystallization
Molecules
step. The
2016, 21,chiral
268 2,3-dihydro-4-quinolones can be used for the preparation of other useful quinolone4 of 19
analogues. The 2-substituted 2,3-dihydro-4-quinolones halt mitosis and are useful antitumor agents [23].

X
O O
O O
Catalyst P
benzene / C6H12 O OH
NH2 R v/v = 1/1 N R
H
0 X
PG free amine 70 C upto quantitative yield
up to 94% ee X= C6F5

Catalyst

Scheme 3. Preparation of chiral 2-substituted 2,3-dihydro-4-quinolones through chiral phosphoric


Scheme 3. Preparation of chiral 2-substituted 2,3-dihydro-4-quinolones through chiral phosphoric acid
acid catalyzed intra-molecular aza-Michael addition reaction.
catalyzed intra-molecular aza-Michael addition reaction.
Molecules 2016, 21, 268 4 of 18
Kuppusamy and Kasi synthesized enantiomerically enriched 2-aryl-2,3-dihydroquinolin-4(1H)-
Kuppusamy
Molecules 2016, 21, 268 and Kasi synthesized enantiomerically enriched 2-aryl-2,3-dihydroquinolin-4(1H)-
ones (in up to 99% yield) in a one pot synthesis using per-6-ABCD (Scheme 4). This synthesis 4method of 18
is
ones (in up to 99% yield) in a one pot synthesis using per-6-ABCD (Scheme 4). This synthesis
very useful for obtaining enantiomer analogues of quinolones. The yield is very high and it is one-pot
method Kuppusamy and Kasi
is very useful synthesized
for obtaining enantiomerically
enantiomer analoguesenriched 2-aryl-2,3-dihydroquinolin-4(1H)-
of quinolones. The yield is very high and
synthesis
ones which
it is one-pot occurs
to 99% in
(in up synthesis a single
yield)
which in a step reaction
oneinpot
occurs with
synthesis
a single step reuseper-6-ABCD
using
reaction of the
with expensive
reuse of(Schemecatalyst
4). This
the expensive after separation
synthesis
catalyst after
from method
the product.
separation is very The
fromusefuldisadvantage
for obtaining
the product. of this method
enantiomer of
The disadvantage is that
analogues the low temperature
of quinolones.
this method is that the The and
low yield long reaction
is very high
temperature and time
and long
reaction
it is conditions
one-pot
reaction are a which
timesynthesis
reaction bit cumbersome
occurs
conditions [24].
areinaabit
single step reaction
cumbersome [24].with reuse of the expensive catalyst after
separation from the product. The disadvantage of this method is that the low temperature and long
O
reaction time reaction conditions are a bit cumbersome [24]. O

O Per-6-ABCD
O + O
Ethanol , Water
NH2 R N R
O 15hr , -50C
Per-6-ABCD H
+ 20 examples
Ethanol , Water
NH2 R N R
15hr , -50C H 99%
ee up to
20 examples
R= C6H5- , p-NO2C6H4-, p-CH3OC6H4-, cyclohexyl-, heterocycles-, etc
ee up to 99%
Scheme 4. Per-6-ABCD catalyzed asymmetric one-pot synthesis of 2-aryl-2,3-dihydro-4-quinolones
Scheme 4. Per-6-ABCD 6H5- , p-NOasymmetric
R= Ccatalyzed 2C6H4-, p-CHone-pot
3OC6H4-,synthesis
cyclohexyl-,
ofheterocycles-, etc
2-aryl-2,3-dihydro-4-quinolones
using substituted aldehydes [24].
using substituted aldehydes [24].
Scheme 4. Per-6-ABCD catalyzed asymmetric one-pot synthesis of 2-aryl-2,3-dihydro-4-quinolones
Ǻkerbladh
using et al. aldehydes
substituted synthesized functionalized 4-quinolones from 2-iodoanilines and alkynes using
[24].
Åkerbladh
two different et protocols
al. synthesized functionalized
for which molybdenum4-quinolones
hexacarbonylfrom
was 2-iodoanilines
used as a solid and alkynes
source of COusing
Ǻkerbladh
(Scheme
two different 5). et al.
In one
protocols ofsynthesized
the which functionalized
for methods, 4-quinolones
the reaction conditions
molybdenum fromwas
include
hexacarbonyl 2-iodoanilines
microwave
used as aand
heating alkynes
of 120
solid °C
sourceusing
and
of CO
two
the different
cyclized protocols
product was for which
obtained molybdenum
in 20 min in hexacarbonyl
high yield. The was used
second as a solid
procedure
(Scheme 5). In one of the methods, the reaction conditions include microwave heating of 120 C and source
involves of
the CO
use
˝
(Scheme
the cyclized 5). Insubstituents
of sensitive
product onewas
of the methods,inthe
like nitro-
obtained reaction
20and conditions
minbromo-groups,
in high yield.include
but
Thethe microwave
reaction
second isheating of 120at°Croom
performed
procedure involves anduse of
the
the cyclized product
temperature. was method
This second obtainedisinalso
20 one-pot
min in high yield.that
synthesis Theoccurs
secondinprocedure involves
two reaction steps. the use
In both
sensitive substituents like nitro- and bromo-groups, but the reaction is performed at room temperature.
of the
sensitive
synthesis substituents like nitro-isand
methods palladium usedbromo-groups, but the reaction is performed at room
as a catalyst [25].
This second method is also one-pot synthesis that occurs in two reaction steps. In both of the synthesis
temperature. This second method is also one-pot synthesis that occurs in two reaction steps. In both
methods
of thepalladium is used as
synthesis methods a catalyst
palladium [25].as Method
is used
A
a catalyst [25]. O
I
Pd2(dba)3Method
, dppf , A
Mo(CO)6 , Et2NH
R1 + R2 O R2
H2 N MW , 1200C , 20min R1 N
I H
Pd2(dba)3 , dppf , Mo(CO)6 , Et2NH
R1 + R2 13+20 entriesR
2

H2 N MW , 1200C , 20min R1 upNto 85% yield


H
13+20 entries
Pd(OAc)2
up to 85% yield
{HP(tBu)3}BF4 O
Pd(OAc), 2Et N
Mo(CO) Et2NH
6 3
{HP(tBu) 3}BF4
rt, 16hr R2 rt, 5hr
R1 O
H2 N Et2NH
Mo(CO)6 , Et3N
rt, 16hr R2 rt, 5hr
R1
HMethod
2N B

Method B

Scheme 5. Synthesis of 4-quinolones via palladium catalyzed carbonylative Sonogashira cross-coupling


Scheme 5. Synthesis of 4-quinolones via palladium catalyzed carbonylative Sonogashira cross-coupling
reaction [25].
reaction [25].
Scheme 5. Synthesis of 4-quinolones via palladium catalyzed carbonylative Sonogashira cross-coupling
Monastyrskyi
reaction [25]. et al. developed a protocol for the synthesis of 3-aryl-4(1H)-quinolones from ethyl
acetoacetate (EAA) using hypervalent diaryliodonium salts under mild and metal-free conditions
Monastyrskyi
(Scheme et al. developed
6). This method has alsoa been
protocol for the
applied forsynthesis of 3-aryl-4(1H)-quinolones
the synthesis from ethyl
of the antimalarial compound
acetoacetate
ELQ-300. This (EAA) using hypervalent
compound diaryliodonium
is active against salts under
Plasmodium falciparum andmild and metal-free
Plasmodium vivax atconditions
all stages
(Scheme 6). This
of its lifecycle andmethod
is in thehas also been
preclinical applied
testing forThe
stage. thetoxicity
synthesis of the
of this antimalarial
product is not yetcompound
reported,
ELQ-300. This compound
but the preclinical is activeimportant
data provides against Plasmodium
information falciparum
concerningandefficacy.
Plasmodium
The vivax at all of
usefulness stages
this
Molecules 2016, 21, 268 5 of 19

Monastyrskyi et al. developed a protocol for the synthesis of 3-aryl-4(1H)-quinolones from ethyl
acetoacetate (EAA) using hypervalent diaryliodonium salts under mild and metal-free conditions
(Scheme 6). This method has also been applied for the synthesis of the antimalarial compound ELQ-300.
This compound is active against Plasmodium falciparum and Plasmodium vivax at all stages of its lifecycle
and is in the preclinical testing stage. The toxicity of this product is not yet reported, but the preclinical
data provides important information concerning efficacy. The usefulness of this synthetic method is its
product that is highly desirable, as malaria is one of the deadliest diseases, affecting millions of people
in Asia 2016,
Molecules and Africa
21, 268 [26]. 5 of 18
Molecules 2016, 21, 268 5 of 18
O
O 1
O O COOEt R
Ar2IX O
O COOEt R1
Ar2IXDMF
Base, Conrad-Limphac
COOEt R1 N
Base, DMF 1 Conrad-Limphac R2 H
COOEt R N
EAA 2 H
R3-aryl-4(1H)-quinolones
EAA 3-aryl-4(1H)-quinolones
Scheme 6. Metal free arylation of ethylacetoacetate with hypervalent diaryliodonium for the
Scheme 6. Metal free arylation of ethylacetoacetate with hypervalent diaryliodonium for the synthesis
Scheme 6.
synthesis Metal free arylation of[26].
of 3-aryl-4(1H)-quinolones ethylacetoacetate with hypervalent diaryliodonium for the
of 3-aryl-4(1H)-quinolones [26].
synthesis of 3-aryl-4(1H)-quinolones [26].
Wang et al. synthesized a wide variety of quinolones in a high yield in one step process using a
N-H et
mild Wang
Wang et al. synthesizedreaction
al. synthesized
activation/Heck aa wide
widevariety
variety
method ofquinolones
of quinolones
(Scheme 7). ininaThis
ahigh
high yieldinin
yield
process onestep
one stepprocess
involves process
the using
using
palladiuma
a mild N-H activation/Heck
mild N-H activation/Heck
catalyzed-oxidative reaction
annulationreaction method
method
of acrylamide (Scheme
(Scheme
with 7).
strained7). This process
This This
arynes. process involves
involves
technique the
was the
madepalladium
palladium
possible
catalyzed-oxidative
catalyzed-oxidative
for annulation
annulation
the first time as previous of acrylamide
of acrylamide
attempts with
with
by other strained arynes.
strained resulted
researchers This
arynes. This technique
technique
in lower was
yieldswas made possible
made
[27]. possible
for
for the
the first
first time
time asas previous
previous attempts
attempts byby other
other researchers
researchers resulted
resulted in in lower
lower yields
yields[27].
[27].
TfO
TfO R3
TMS R3
TMS

CsF
O CsF O
Pd(OAc)2 (5 mol%)
R1 O OMe R1 O OMe
1
N Cu(OAc)22 (5
Pd(OAc) mol%)
(2equiv) N
R OMe+ 3 1
R R OMe
H
N Cu(OAc)2 (2equiv) N
R2 H + R3 DMSO/Dioxane = 1/9 R2
H
R2 H DMSO/Dioxane = 1/9 R2
R3
R 3
Scheme 7. Palladium catalyzed-oxidative annulation of acrylamide with strained arynes for the
Scheme 7.
synthesis of Palladium
quinolonescatalyzed-oxidative
[27]. annulation of acrylamide with strained arynes for the
Scheme 7. Palladium catalyzed-oxidative annulation of acrylamide with strained arynes for the
synthesis of quinolones [27].
synthesis of quinolones [27].
Manikandan and Jeganmohan prepared 2-quinolones by Ru-catalyzed cyclization of anilides with
Manikandan
propiolates and Jeganmohan
or acrylates with further prepared 2-quinolones
conversion by Ru-catalyzed cyclization
into 3-halo-2-quinolones of anilides with
and 2-chloro-quinolones
(Scheme 8). The 2-quinolones produced through this mechanism are very useful for theanilides
Manikandan
propiolates or and
acrylatesJeganmohan
with prepared
further 2-quinolones
conversion into by Ru-catalyzed
3-halo-2-quinolones cyclization
and of
2-chloro-quinolones
synthesis with
of
propiolates
(Scheme
other 8). or
Theacrylates
quinolone withand
2-quinolones
derivatives, further
produced conversion
through
are prepared into
in this 3-halo-2-quinolones
mechanism
excellent are very
yield. The and 2-chloro-quinolones
useful
mechanisms forproposed
the synthesis of
for the
(Scheme
above 8). Thewere
other quinolone
reactions 2-quinolones
derivatives, produced
stronglyand through
are prepared
supported inthis mechanism
excellent
by experimental yield. are
evidenceThe very useful forproposed
mechanisms
and deuterium the synthesis
labeling of
for the
studies,
other
abovequinolone
reactions
suggesting derivatives,
easy were
isolation of and
strongly are prepared
supported
reaction in excellent
by experimental
intermediates during yield.
evidenceThe mechanisms
and
synthesis. deuterium proposed for the
labelingproducts
The halogenated studies,
above
made reactions
suggesting
through easy were
such strongly
isolation
synthesis supported
of methods
reaction still by experimental
intermediates
need to be during evidence and
assayedsynthesis. deuterium labeling
The halogenated
for their toxicity in mouse models studies,
products
for
suggesting
madefuture
their througheasy isolation
such
medicinal of reaction
synthesis
value methods
[28]. intermediates
still need toduring synthesis.
be assayed Thetoxicity
for their halogenated
in mouseproducts
models made
for
through such synthesis methods
their future medicinal value [28]. still need to be assayed for their toxicity in mouse models for their
future medicinal value [28]. 1- Ru(II)AgSbF6
H Cu(OAc)2, H2O H H
N O 1- Ru(II)AgSbF6 N Me N O
H 11020,C,
Cu(OAc)
DCE, H212hr
O H Ru(II)AgSbF6 H
N O N O Me N O
DCE, 110 0C, 012hr Ru(II)AgSbF
RCOOH, i-PrOH
6 H
R1
2- 30% HCl, 130 C, 10hr R1 O R1
1300C,
24hr
RCOOH, i-PrOH R2 H
OR0C,
2- 30% HCl, 130 4 10hr
R1 R1
R1
4 R21300C, COOR
24hr
3
R2
O OR
R2 COOR3
O H
N O
H NBS or NCS
N O AIBN
X NBS or NCS
R1 X=AIBN
Br, Cl
R2 X
R1 X= Br, Cl
R2
Manikandan and Jeganmohan prepared 2-quinolones by Ru-catalyzed cyclization of anilides with
propiolates or acrylates with further conversion into 3-halo-2-quinolones and 2-chloro-quinolones
(Scheme 8). The 2-quinolones produced through this mechanism are very useful for the synthesis of
other quinolone derivatives, and are prepared in excellent yield. The mechanisms proposed for the
above reactions were strongly supported by experimental evidence and deuterium labeling studies,
suggesting easy isolation of reaction intermediates during synthesis. The halogenated products
Molecules 2016, 21, 268 6 of 19
made through such synthesis methods still need to be assayed for their toxicity in mouse models for
their future medicinal value [28].

1- Ru(II)AgSbF6
H Cu(OAc)2, H2O H H
N O N Me N O
DCE, 1100C, 12hr Ru(II)AgSbF6
O
2- 30% HCl, 1300C, 10hr R1 RCOOH, i-PrOH H
R1 R1
1300C, 24hr R2
OR4
R2 COOR3
O
H
N O
NBS or NCS
AIBN
X
R1 X= Br, Cl
R2

N Cl
POCl3

R1
R2

Scheme 8. Synthesis of 2-quinolones through ruthenium catalyzed cyclization of anilides with


Scheme Synthesis
8.substituted of 2-quinolones
propiolates through ruthenium catalyzed cyclization of anilides
or acrylates [28]. with
substituted propiolates or acrylates [28].

Suet et al. synthesized and evaluated bisaryl quinolones for biological activities against
P. falciparum [29]. Pateland his coworkers synthesized a series of novel pyridoquinolone analogues
by introducing sulfonamide, urea, thiourea, amine, amide, piperazine and β-lactam moieties in the
pyridoquinolone molecule. These quinolones on the one hand inhibit gyrase and topoisomerase
IV of bacteria while the other attached groups are also inhibitors of specific metabolic pathways.
These medicinal molecules have antibacterial and antifungal activities and some of them are even
active against drug resistant Mycobacterium tuberculosis strains. However there is a need to test these
novel quinolones in mammals for their potential toxicity [30–36]. Komarnicka et al. made phosphine
derivatives of sparfloxacin in high yield by treating sparfloxacin with methoxy(diphenyl)phosphine.
The oxide derivative was made by treating the aminomethyl(diphenyl)phosphine analog of
sparfloxacin with 35% hydrogen peroxide in chloroform at room temperature with continuous stirring
for 3 h. The aminomethyl(diphenyl)phosphine analog of sparfloxacin showed antibacterial activity of
similar nature to sparfloxacin, while the oxide form had high antitumor activity against cancer cell
lines [37]. One of the beautiful modifications in the existing quinolone is the production of a series
of 1,2,4-triazole-ciprofloxacin hybrid compounds that have potential antibacterial potency against
both Gram-positive as well as Gram-negative bacteria. These 1,2,4-triazole-ciprofloxacin hybrid
compounds were more active than ciprofloxacin against both vulnerable and quinolone resistant
bacteria. Such promising hybrid triazole analogues of other quinolone compounds should also be
prepared and tested against both resistant and non-resistant bacterial strains that cause different
diseases. In addition to determine their action against pathogenic bacteria, they should also be tested
for viral infections, especially HIV [38]. A number of new natural antibiotics are being discovered that
can target fluoroquinolone resistant bacteria (FQR). For example deoxynymbomycins can target the
mutant bacterial DNA gyrase and can be used against FQR bacteria but the chances of resistance to
this new drug are still there [39]. Similarly, the FQR Neisseria gonorrhoeae type II topoisomerase can be
inhibited by a novel compound called spiropyrimidinetrione AZD0914 [40] so on one hand we have
the continuously evolving resistant strains to quinolones and other antibacterial agents, but on the
other hand scientific community is working hard to design and synthesize new drugs for the resistant
bacterial strains.

3. Analogues of Quinolones
The first member of the quinolone family was nalidixic acid, which was synthesized as a
byproduct in the synthesis of chloroquine. Later, it was modified in order to enhance its activities.
One modification was the substitution of fluorine at the C-6 position, which was found to provide good
antibacterial activities. Moreover, substitution of piperazine or methyl piperazine, pyrrolidinyl and
piperidinyl moieties was also determined to result in increased antibacterial activities. Following are
the analogues of quinolones available in the common literature (Figure 2) [15,16,41–48].
3. Analogues of Quinolones
The first member of the quinolone family was nalidixic acid, which was synthesized as a
byproduct in the synthesis of chloroquine. Later, it was modified in order to enhance its activities.
One modification was the substitution of fluorine at the C-6 position, which was found to provide
good antibacterial activities. Moreover, substitution of piperazine or methyl piperazine, pyrrolidinyl
Molecules 2016, 21, 268
and piperidinyl moieties was also determined to result in increased antibacterial activities. Following 7 of 19
are the analogues of quinolones available in the common literature (Figure 2) [15,16,41–48].

O O O O O O
F
OH O OH OH

N N O N N N
HN

Nalidixic acid Oxolinic acid Norfloxacin


O O
O O
F OH F
OH
N N
HN N N
N O
Molecules 2016, 21, 268Ciprofloxacin Ofloxacin 7 of 18

O O O O
F OH F OH
H
N N N N
N
O O
N H
H
Levofloxacin Moxifloxacin
NH2 O O O O
F
F OH
OH
H2 N
N N
N N
O O
HN F N

Sparfloxacin 8-methoxyfloroquinolone
O O O O
F F
OH OH

HN N N N
HN N N N
O
S N
O
6-Fluoro-7-(3-methoxy-4-methylamino-pyrrolidin-1-yl)
-4-oxo-1-phenyl-1,4-dihydro-[1,8]naphthyridine-3-
Voreloxin carboxylic acid
O O
F OH
HN F O O
N N F
O OH
O
N N
5-Fluoro-6-(3-methoxy-4-methylamino- HN F
pyrrolidin-1-yl)-3-oxo-3H-7-oxa-11b-aza-
benzo[de]anthracene-2-carboxylic acid Orbifloxacin
O O NH2 O O
F F
OH OH

N N N N
HN F HN O

Lomefloxacin Gatifloxacin
O O
O O
F
OH
OH O
N N N N
N
HN
O
H2 N
CHF2
Garenofloxacin Gemifloxacin

Figure 2. Cont.
Molecules 2016, 21, 268 8 of 19
Molecules 2016, 21, 268 8 of 18

O O
F O O
OH
F
H N N N OH
F
H2N N N
H
HN
F
Trovafloxacin Enoxacin
O O O O
F F
OH OH
H2N
N N N
N

Enrofloxacin Pazufloxacin
O O
O O F
F OH
OH
O N N
O N N Cl
O S
N
H2N H
Prulifloxacin Besifloxacin
NH2 O O O O
O O F
F OH OH
F
OH
HN N N N N
N N N
O Cl
O
H2 N F
Balofloxacin Antofloxacin Sitafloxacin
O O
O O O O
F F F OH
OH OH
N N N N N
N N

Enrofloxacin Danofloxacin Flumequine


O O O O
F OH F OH
NH2 O O
N N N N
F OH HN N

N N
N
O N
F F
Marbofloxacin Sarafloxacin Difloxacin

Figure 2. Common analogues of quinolones used for the treatment of different diseases.
Figure 2. Common analogues of quinolones used for the treatment of different diseases.
4. Clinical Use
4. Clinical Use
There are a number of quinolone analogues used in clinical practice all over the world due to
their
There increased antibacterial
are a number range and favorable
of quinolone analoguespharmacokinetics,
used in clinical while the search
practice for new
all over the hybrid
world due
next generation quinolones is ongoing. Among the most suitable quinolones candidates that
to their increased antibacterial range and favorable pharmacokinetics, while the search for new are highly
prescribed are gatifloxacin, levofloxacin, ciprofloxacin. There are several quinolones that have more
hybrid next generation quinolones is ongoing. Among the most suitable quinolones candidates that
toxicity for the patients than usefulness, like halogenated quinolones, but such toxic analogues
are highly prescribed are gatifloxacin, levofloxacin, ciprofloxacin. There are several quinolones
that have more toxicity for the patients than usefulness, like halogenated quinolones, but such
toxic analogues should be used when no other option is available and the toxic damage is less
Molecules 2016, 21, 268 9 of 19

as compared to the pathogen harm [49]. The quinolones are also suitable choice for lung infections like
cystic fibrosis. Clinafloxacin is cytotoxic, but it can be used against multi-drug resistant Burkholderia
cepacia [49]. Pseudomonas aeruginosa-related respiratory diseases can also be treated best with anionic
fluroquinolones. In P. aeruginosa based diseases like cystic fibrosis where they form a thick biofilm of
anionic molecules such as rhamnolipids and alginate, it is difficult for most antibiotics to penetrate
and inhibit bacterial growth. However the negatively charged fluoroquinolones have the ability to
cross the mucus and thus have better pharmacodynamics due to their negatively charged nature [50].
Some quinolones like ciprofloxacin, levofloxacin, moxifloxacin, ofloxacin, and gatifloxacin are
also approved by Federal Drug Administration (FDA) for pediatric use for different diseases like
conjunctivitis, otitis, sinusitis, respiratory tract infections, pneumonia, UTI and gastrointestinal
diseases [51]. Fluoroquinolones can be used to cure tuberculosis (TB) as other active agents used for
treatment take six months or more, while FQ can shorter this time span [52]. The development of
multi-drug resistance in Mycobacterium tuberculosis strains has already alarmed the medical community
as these strains are highly resistant to antibiotics. Quinolones also suffer the same fate as antibiotics as
several strains of M. tuberculosis generate mutations in their DNA gyrase enzyme encoded by GyrA
and GyrB genes [53–55]. Mutations in the amino acids Ala90 and Asp94 of GyrA of several strains
of M. tuberculosis resulted in high levels of resistance to levofloxacin [56]. The level of resistance to
various analogues of quinolones is different in different countries [54,57–61].
The quinolones can also be used to treat diseases that occur as a result of excessive production
of aldosterone. Some quinolone analogues can inhibit a group of mitochondrial cytochrome P450
(CYP) enzymes that are involved in aldosterone biosynthesis. Thus they can be used for the treatment
of heart related diseases like hypertension, primary aldosteronism, congestive heart failure, cardiac
fibrosis, ventricular remodelling, and diabetic nephropathy that occurs due to high level of aldosterone
production [62].
Beside the antibacterial role of quinolone, there are certain quinolonyl diketo acid analogues that
are active against human immunodeficiency virus (HIV). They act against multiple viral enzymes
that include reverse transcriptase, integrase and ribonuclease. These agents bind in the active site of
integrase, ribonuclease and reverse transcriptase with the help of magnesium ion with acidic residues
in their catalytic site and in that way they halt the function of these enzymes [63–67]. These quinolonyl
diketo acid analogues are synthesized by introduction of various alkylating group at the nitrogen atom
of the quinolinone ring [63]. Similarly the mono and bifunctional quinolinonyl diketo acids are also
potent inhibitors of strand transfer function of HIV integrase, which are synthesized by treating aniline
with ethyl orthoformate and ethyl acetoacetate [65–67]. The antitumor activities of quinolones were
also investigated by different research groups. A Mannich base can be prepared from ciprofloxacin and
kojic acid that possesses antitumor activities. This Mannich base and its complex with copper are toxic
to cancerous cells in different ways that include inhibition of replication, disruption of polarization of
mitochondrial membrane resulting into loss of ATP activity of cancerous cell [68]. It is also reported
that quinolones acts against other types of cancer where its mode of action is suggested to be inhibition
of topoisomerase II.
In veterinary medicine the quinolones are also prescribed due to their favorable
pharmacodynamics and pharmacokinetics, especially for various types of diseases and wounds in
dogs and cats. The commonly prescribed fluoroquinolones for different dog and cat diseases includes
ciprofloxacin, difloxacin, enrofloxacin, marbofloxacin, orbifloxacin and pradofloxacin [69,70].

5. Targets of Quinolones
The main targets of quinolones are DNA topoisomerases. These enzymes are essential for DNA
replication, transcription, recombination and condensed DNA remodeling, and function by carrying
out transient single- and double-strand breaks. The topoisomerases modulate the supercoiling of DNA
to enable proper function and interaction with proteins. These enzymes also have a fundamental role
in most nucleic acid processes, like helping to control levels of DNA under- and over-winding, as
Molecules 2016, 21, 268 10 of 19

well as removing knots and tangles from bacterial chromosomes [71–73]. The enzymes that cleave
just one strand of the DNA are called type I topoisomerases, and they are further divided into
type IA, based on the protein covalent linkage to 5’-phosphate, or type IB if the linkage is to 3’
phosphate of the DNA strand. The type II topoisomerases cleave both strands of the double stranded
DNA concurrently. The binding and subsequent hydrolysis of ATP with type II topoisomerase
results in a conformational changes in DNA. In most of the bacteria, two different but structurally
similar type II topoisomerases are present, namely: gyrase and topoisomerase-IV. Both gyrase and
topoisomerase-IV are heterotetrameric subunits. The gyrase has two each GyrB and GyrA subunits
while the topoisomerase-IV contains two subunits each of ParE and ParC subunits in Gram-positive
bacteria. On the other hand GrlA and GrlB are the subunits of topoisomerase-IV in the case of
Gram-positive species [71,74–77]. Schoeffler and Berger have reviewed the structure and function
of the type II topoisomerase mechanism in great detail, as well as the role of their ATP-dependent
domains [76,78,79]. The processes of ligation and DNA cleavage, which make up the central part of
enzyme function, use a non-canonical two-metal ion mechanism [80,81]. Gyrase and topoisomerase-IV
make staggered cuts in DNA that are four base pairs apart and on opposite strands, leaving a 5’
overhang. In this process, a covalent bond is formed between active site tyrosine residues and the
newly generated 5’ DNA termini, in order to maintain a genomic integrity [16,71,74,75,77]. It is
noteworthy that humans also have type II enzymes; topoisomerase IIα and topoisomerase IIβ which
have significant amino acid sequence similarity to bacterial enzymes, but in humans, the genes that
code for subunits A and B are fused, forming a single polypeptide chain. Thus, the human type
II enzymes function as homodimers. Due to this difference, the clinically-relevant quinolones can
discriminate between the human and bacterial type II topoisomerases [16,71,82].

6. Mode of Action of Quinolones


It was first recognized in 1977 that the bacterial enzyme gyrase is the primary target of quinolones.
Later, with the discovery of topoisomerases, it was suggested that this enzyme could also be a target of
quinolones. Analysis of Escherichia coli strains carrying drug-resistance mutations in these enzymes
showed that quinolones inhibit gyrase and topoisomerase-IV as primary and secondary targets,
respectively [8]. There are several high-resolution structures of topoisomerase in different catalytic
modes with DNA, which have shown how the topoisomerases modify the topology of DNA [83–85].
In a recent investigation, the interaction of quinolones and type II topoisomerase of human and bacteria
are mediated by different types of interactions. It was shown that the quinolones target the gyrase
and topoisomerase-IV enzymes and inhibit bacterial growth [86]. These enzymes have the potential to
fragment the cell genome, which is essential for cell survival and reproduction. Quinolones inhibit
cell growth and division by increasing the concentration of enzyme-DNA cleavage complexes [16,73].
Thus quinolones are referred to as topoisomerase poisons as they convert gyrase and topoisomerase
IV into cellular toxins [87].

7. Interaction of Quinolones with Topoisomerases


The interaction of drugs with proteins plays an important role in stabilizing the complexes. In the
case of Gram-negative bacteria, quinolones bind to gyrase, while in Gram-positive bacteria, the drugs
preferentially bind to topoisomerase-IV [88,89]. The amino acids that are usually associated with
quinolone-resistance are GyrA-Serine 83 and GyrA-aspartate 87 in E. coli [90–94]. Mutation of these
two amino acids to alanine results in resistance to quinolones, which showed the importance of these
two amino acids in drug binding [95]. Consequently, it is has been assumed that these two amino acids
play a crucial role in mediating quinolone-enzyme interactions. A large number of crystallographic
studies have been performed to explain the quinolone-enzyme interactions. These structures showed
how the drugs interact with both the enzyme and DNA and how these drugs halt the normal process
of DNA cleavage and religation. There are differences in the interaction of drugs with the enzyme
among the resolved structures as they vary with the species and the drug used. However they are
Molecules 2016, 21, 268 11 of 19

helpful in designing more potent analogues that can bind more tightly with the enzyme and DNA.
In addition, molecular docking and simulation of such structures and different quinolone analogues
Molecules 2016, 21, 268 11 of 18
will be helpful in modelling the interactions to help design new drugs [83,84,96–100]. Osheroff and
coworkers
coworkers provided
providedmost most of of
the the
datadata
aboutabout
the interaction of quinolones
the interaction and topoisomerases
of quinolones [16,45,73,
and topoisomerases
77,81,82,86,101–109]. In a recent study, the quinolone structure was placed near
[16,45,73,77,81,82,86,101–109]. In a recent study, the quinolone structure was placed near the serine the serine and acidic
residues,
and acidicbut it was found
residues, that found
but it was the aminothat acids were acids
the amino not close
wereenough
not closeto mediate
enough to a direct
mediatebinding to
a direct
drugs. On the other hand, a structure of a quinolone complex was found having a non-catalytic Mg 2+
binding to drugs. On the other hand, a structure of a quinolone complex was found having a
chelated by theMg C3/C4 keto acid of the drugsketo
[86].acid
Theof Mg 2+ was in coordination with four molecules of
non-catalytic 2+ chelated by the C3/C4 the drugs [86]. The Mg2+ was in coordination
water, two of which were located close enough to the
with four molecules of water, two of which were located close enough serine and acidic residuestofavorable
the serine forand
hydrogen
acidic
bonding (Figure 3). The above studies suggested that the interaction of water
residues favorable for hydrogen bonding (Figure 3). The above studies suggested that the interaction to metal bridged the
quinolone
of water totometalthe enzyme
bridged[83,84,86,93,97,99].
the quinolone to the It was also observed
enzyme that a mutation
[83,84,86,93,97,99]. It was in theobserved
also place of serine
that a
or nearby another amino residue limits the variety of metal ions that
mutation in the place of serine or nearby another amino residue limits the variety of metal support the drug activity
ions[101].
that
Thus mutation resulted in decrease in affinity of the enzyme 2+
support the drug activity [101]. Thus mutation resulted in for quinolones
decrease at theof
in affinity noncatalytic
the enzyme Mgfor
site. Additionally,
quinolones these mutations
at the noncatalytic Mg2+decrease the affinity these
site. Additionally, of gyrase and topoisomerase-IV
mutations for quinolones
decrease the affinity of gyrase
and
and topoisomerase-IV for quinolones and in the case of mutation in both residues, there is atocomplete
in the case of mutation in both residues, there is a complete loss of relevant quinolones stabilize
cleavage complexes
loss of relevant [16,86,103].
quinolones to stabilize cleavage complexes [16,86,103].

Glu/Asp

O O

H Ser
O

O O
F
OH
N N
HN

Figure 3. Quinolone-enzyme interaction [86].


Figure 3. Quinolone-enzyme interaction [86].

Thus the existence of a metal ion-water complex verifies that serine and acidic residues act as
Thus
anchor pointsthe existence of a metal
that coordinate ion-water
the bridge to thecomplex
enzyme verifies thatthe
and that serine and acidic
water-metal residues
ion bridge act as
is the
anchor points that coordinate the bridge to the enzyme and that the water-metal
primary interaction between the drug and bacterial type II enzymes [104]. It is noteworthy that theion bridge is the
primary interaction between
type II topoisomerase of humansthe drug
lacks and
serinebacterial typeresidues
and acidic II enzymes [104].
to affix theItwater-metal
is noteworthyionthat the
bridge
type
and isII therefore
topoisomerase of humans
not capable lacks serine
of utilizing and acidic
this critical residuestotointeract
mechanism affix thewith
water-metal ion That
quinolones. bridge is
and is therefore not capable of utilizing this critical mechanism to interact with quinolones.
why the toxicity of quinolones is also minor for humans [104]. These differences in topoisomerases That is why
the toxicityprovide
structure of quinolones
a baseis also minor for
by which humans [104].
quinolones These differences
differentiate between in topoisomerases
the bacterial andstructure
human
provide a base by which quinolones differentiate between
topoisomerases and opens beneficial avenues for this class of drugs. the bacterial and human topoisomerases
and opens beneficial avenues for this class of drugs.
8. Bacterial Resistance to Quinolones
8. Bacterial Resistance to Quinolones
Like other antibiotics, quinolones face resistance from infectious bacteria due to their widespread
Like other antibiotics, quinolones face resistance from infectious bacteria due to their widespread
use and this resistance is a major clinical issue. Bacterial resistance to quinolones is increasing and it
use and this resistance is a major clinical issue. Bacterial resistance to quinolones is increasing and
is common reported throughout the world. Most vancomycin-resistant enterococcus (VRE) and
it is common reported throughout the world. Most vancomycin-resistant enterococcus (VRE) and
methicillin-resistant Staphylococcus aureus (MRSA) are also cross-resistant to fluoroquinolones [109,110].
methicillin-resistant Staphylococcus aureus (MRSA) are also cross-resistant to fluoroquinolones [109,110].
Bacterial resistance to quinolones can be classified into three categories [16].
Bacterial resistance to quinolones can be classified into three categories [16].
9. Target-Mediated Quinolones Resistance
9. Target-Mediated Quinolones Resistance
Mutation in the target site of quinolone: gyrase (GyrE subunit) and topoisomerase-IV (ParC
Mutation in the target site of quinolone: gyrase (GyrE subunit) and topoisomerase-IV (ParC
subunit) of bacteria are the most common and they result in resistance to quinolones. The serine and
subunit) of bacteria are the most common and they result in resistance to quinolones. The serine
acidic residue of amino acids that bind the water-metal on bridges undergo mutation frequently
and acidic residue of amino acids that bind the water-metal on bridges undergo mutation frequently
results in ten times less binding affinity for quinolones [95,104]. Most probably, the disturbance in
the water-metal ion bridge leads to the quinolone resistance. Normally, mutations in serine comprise
above 90% of the mutant pool in both clinical and laboratory isolates, while changes at the acidic
residues comprising the bulk of other mutations [92,93]. In most of the resistant strains of bacteria,
serine is the most common residue that is mutated: to leucine in MRSA and to isoleucine, arginine or
tyrosine in GyrA subunit in VRE. After the mutation in gyrase and topoisomerase-IV, wild type
Molecules 2016, 21, 268 12 of 19

results in ten times less binding affinity for quinolones [95,104]. Most probably, the disturbance in the
water-metal ion bridge leads to the quinolone resistance. Normally, mutations in serine comprise above
90% of the mutant pool in both clinical and laboratory isolates, while changes at the acidic residues
comprising the bulk of other mutations [92,93]. In most of the resistant strains of bacteria, serine is the
most common residue that is mutated: to leucine in MRSA and to isoleucine, arginine or tyrosine in
GyrA subunit in VRE. After the mutation in gyrase and topoisomerase-IV, wild type DNA cleavage
does not stop in the absence of drugs, however quinolones have little ability to enhance the cleavage of
enzyme mediated DNA [16,102,111–113]. In addition, the binding capability of quinolones is largely
reduced and it loses much of their ability to inhibit DNA ligation or to make enzyme-DNA-drug
complex [16,104].

10. Plasmid-Mediated Quinolone Resistance


The second method of bacterial resistance to quinolones are plasmids that carry quinolone
resistance genes that are also a rising clinical issue, commonly causing low level resistance (less than
10-fold) [114–116] though a 250-fold resistance has also been reported [117,118]. The transmittance
of plasmids mediated quinolone resistance from one bacterial generation to another can be done
horizontally via conjugation as well as vertically (target mediated resistance is transmitted only
vertically). The plasmids responsible for quinolones bear additional genes which offer resistance to
other drug classes [115,119,120]. Plasmid mediated quinolone resistance is due to three group of genes.
These genes are qnr genes, AAC (6)-Ib-cr and the third group comprised of efflux pumps. The qnr
genes offer resistance by decreasing the binding of DNA to gyrase and topoisomerase-IV; as a result
there is decrease in the target site of chromosomes. The efflux pump is also a plasmid encoded protein
that causes resistance to quinolones [121,122]. The second gene associated with quinolone resistance is
an alternative protein of an aminoglycoside acetyltransferase having a mutation at two specific points.
The enzymes decrease the activity of the quinolones by acylation of unsubstituted nitrogens of the C7
piperazine ring in norfloxacin and ciprofloxacin [123].

11. Chromosome-Mediated Quinolone Resistance


The concentration of quinolones inside the cell is regulated by the diffusion-mediated uptake
of drugs and pump-mediated efflux. For a drug to enter into cell of Gram-negative bacteria, it must
pass through extra barriers. Therefore the influx of drug into Gram-negative species is facilitated
by protein channels called porins. The down regulation of porins expression leads to low level
quinolone resistance [109,118,119]. Along with the plasmid encoded efflux pump, if the expression of
chromosomes encoded efflux pumps are enhanced, it will also offer low level resistance. Usually the
mutations in regulatory proteins cause the up-regulation of these efflux pumps [109,124]. The resistance
that arises due to lowering the concentration of quinolones within the cell may not be a big clinical
issue but it provides a favorite environment to other kind of resistance to antibacterial agents [124,125].

12. Conclusions
Quinolones are utilized worldwide as broad spectrum antibacterial agents, effective against both
Gram-positive as well as Gram-negative bacteria. There are several quinolones derivatives that also
work as antimalarial, antiviral and anticancer agents. After their initial synthesis as a byproduct of
chloroquine, quinolones evolved as a separate class of synthetic drugs. Currently different synthetic
methods are explored to synthesize more effective novel analogues of quinolones. Due to the overuse of
these drugs, certain bacteria produce resistance to them and these strains of bacteria are now a common
threat worldwide. Several mechanisms for resistance have been explained among which mutation
at the target site is common. There is a need to explore new synthetic routes for novel quinolone
analogues that are cost effective, less cytotoxic and have better pharmacokinetic and pharmacodynamic
properties. This latest information about quinolones can help scientists to develop such analogues
which can avoid resistance to quinolones and can extend their clinical utilization in the future.
Molecules 2016, 21, 268 13 of 19

Acknowledgments: We thank Kiera Reifschneider for her suggestions and support in writing of this manuscript.
A.N. acknowledges financial support from National Center of Excellence in Physical Chemistry, University of
Peshawar, Pakistan for covering the costs to publish in open access.
Author Contributions: A.N.; S.B. and M.M. provided the concept and designed the manuscript. N.A. and K.K.
participated in the discussion during preparation and writing of the manuscript. All authors read and approved
the final manuscript of this review.
Conflicts of Interest: All the authors declare that they have no competing interests.

References
1. Armstrong, G.L.; Conn, L.A.; Pinner, R.W. Trends in infectious disease mortality in the united states during
the 20th century. JAMA 1999, 281, 61–66. [CrossRef] [PubMed]
2. Cross, R.M.; Flanigan, D.L.; Monastyrskyi, A.; LaCrue, A.N.; Saenz, F.E.; Maignan, J.R.; Mutka, T.S.;
White, K.L.; Shackleford, D.M.; Bathurst, I. Orally bioavailable 6-chloro-7-methoxy-4(1H)-quinolones
efficacious against multiple stages of plasmodium. J. Med. Chem. 2014, 57, 8860–8879. [CrossRef] [PubMed]
3. Baharoglu, Z.; Garriss, G.; Mazel, D. Multiple pathways of genome plasticity leading to development of
antibiotic resistance. Antibiotics 2013, 2, 288–315. [CrossRef]
4. Walsh, C. Where will new antibiotics come from? Nat. Rev. Microbiol. 2003, 1, 65–70. [CrossRef] [PubMed]
5. Livermore, D.M. Has the era of untreatable infections arrived? J. Antimicrob. Chemother. 2009, 64, i29–i36.
[CrossRef] [PubMed]
6. Hu, W.; Lin, J.-P.; Song, L.-R.; Long, Y.-Q. Direct synthesis of 2-aryl-4-quinolones via transition-metal-free
intramolecular oxidative C (sp3 )-H/C(sp3 )-H coupling. Org. Lett. 2015, 17, 1268–1271. [CrossRef] [PubMed]
7. Oliphant, C.M.; Green, G.M. Quinolones: A comprehensive review. Am. Fam. Physician 2002, 65, 455–464.
[PubMed]
8. Lesher, G.Y.; Froelich, E.J.; Gruett, M.D.; Bailey, J.H.; Brundage, R.P. 1,8-naphthyridine derivatives. A new
class of chemotherapeutic agents. J. Med. Chem. 1962, 5, 1063–1065. [CrossRef]
9. Wu, J.; Xiang, S.; Zeng, J.; Leow, M.; Liu, X.-W. Practical route to 2-quinolinones via a pd-catalyzed C-H
bond activation/C-C bond formation/cyclization cascade reaction. Org. Lett. 2014, 17, 222–225. [CrossRef]
[PubMed]
10. Emmerson, A.M.; Jones, A.M. The quinolones: Decades of development and use. J. Antimicrob. Chemother.
2003, 51, 13–20. [CrossRef] [PubMed]
11. Andriole, V.T. The quinolones: Past, present, and future. Clin. Infect. Dis. 2005, 41, S113–S119. [CrossRef]
[PubMed]
12. Bisacchi, G.S. Origins of the quinolone class of antibacterials: An expanded “discovery story”. J. Med. Chem.
2015, 58, 4874–4882. [CrossRef] [PubMed]
13. Mitscher, L.A. Bacterial topoisomerase inhibitors: Quinolone and pyridone antibacterial agents. Chem. Rev.
2005, 105, 559–592. [CrossRef] [PubMed]
14. Domagala, J.M. Structure-activity and structure-side-effect relationships for the quinolone antibacterials.
J. Antimicrob. Chemother. 1994, 33, 685–706. [CrossRef] [PubMed]
15. Guo, X.; Liu, M.L.; Guo, H.Y.; Wang, Y.C.; Wang, J.X. Synthesis and in vitro antibacterial activity of
7-(3-amino-6,7-dihydro-2-methyl-2H-pyrazolo[4,3-c]pyridin-5(4H)-yl) fluoroquinolone derivatives. Molecules
2011, 16, 2626–2635. [CrossRef] [PubMed]
16. Aldred, K.J.; Kerns, R.J.; Osheroff, N. Mechanism of quinolone action and resistance. Biochemistry 2014, 53,
1565–1574. [CrossRef] [PubMed]
17. Sharma, P.C.; Jain, A.; Jain, S. Fluoroquinolone antibacterials: A review on chemistry, microbiology and
therapeutic prospects. Acta. Pol. Pharm. 2009, 66, 587–604. [PubMed]
18. Liu, M.L.; Guo, H.Y. Evolution of the quinolones. World Notes Antibiot. 2006, 27, 69–75.
19. Blasco, C.; PicoÌ, Y. Development of an improved method for trace analysis of quinolones in eggs of laying
hens and wildlife species using molecularly imprinted polymers. J. Agric. Food Chem. 2012, 60, 11005–11014.
[CrossRef] [PubMed]
20. Cross, R.M.; Maignan, J.R.; Mutka, T.S.; Luong, L.; Sargent, J.; Kyle, D.E.; Manetsch, R. Optimization of
1,2,3,4-tetrahydroacridin-9(10H)-ones as antimalarials utilizing structure-activity and structure-property
relationships. J. Med. Chem 2011, 54, 4399–4426. [CrossRef] [PubMed]
Molecules 2016, 21, 268 14 of 19

21. Ferguson, J.; Zeng, F.; Alwis, N.; Alper, H. Synthesis of 2 (1H)-quinolinones via Pd-catalyzed oxidative
cyclocarbonylation of 2-vinylanilines. Org. Lett. 2013, 15, 1998–2001. [CrossRef] [PubMed]
22. Kang, D.; Hong, S. Rh (III) and Ru (II)-catalyzed site-selective C-H alkynylation of quinolones. Org. Lett.
2015, 17, 1938–1941. [CrossRef] [PubMed]
23. Saito, K.; Moriya, Y.; Akiyama, T. Chiral phosphoric acid catalyzed asymmetric synthesis of 2-substituted
2, 3-dihydro-4-quinolones by a protecting-group-free approach. Org. Lett. 2015, 17, 3202–3205. [CrossRef]
[PubMed]
24. Kanagaraj, K.; Pitchumani, K. Per-6-amino-β-cyclodextrin as a chiral base catalyst promoting one-pot
asymmetric synthesis of 2-aryl-2,3-dihydro-4-quinolones. J. Org. Chem. 2013, 78, 744–751. [CrossRef]
[PubMed]
25. Åkerbladh, L.; Nordeman, P.; Wejdemar, M.; Odell, L.R.; Larhed, M. Synthesis of 4-quinolones via a
carbonylative Sonogashira cross-coupling using molybdenum hexacarbonyl as a CO source. J. Org. Chem.
2015, 80, 1464–1471. [CrossRef] [PubMed]
26. Monastyrskyi, A.; Namelikonda, N.K.; Manetsch, R. Metal-free arylation of ethyl acetoacetate with
hypervalent diaryliodonium salts: An immediate access to diverse 3-aryl-4 (1H)-quinolones. J. Org. Chem.
2015, 80, 2513–2520. [CrossRef] [PubMed]
27. Wang, W.; Peng, X.; Qin, X.; Zhao, X.; Ma, C.; Tung, C.-H.; Xu, Z. Synthesis of quinolinones with
palladium-catalyzed oxidative annulation between acrylamides and arynes. J. Org. Chem. 2015, 80, 2835–2841.
[CrossRef] [PubMed]
28. Manikandan, R.; Jeganmohan, M. Ruthenium-catalyzed cyclization of anilides with substituted propiolates
or acrylates: An efficient route to 2-quinolinones. Org. Lett. 2014, 16, 3568–3571. [CrossRef] [PubMed]
29. Leung, S.C.; Gibbons, P.; Amewu, R.; Nixon, G.L.; Pidathala, C.; Hong, W.D.; Pacorel, B.; Berry, N.G.;
Sharma, R.; Stocks, P.A.; et al. Identification, design and biological evaluation of heterocyclic quinolones
targeting Plasmodium falciparum type II NADH: Quinone oxidoreductase (PfNDH2). J. Med. Chem. 2012, 55,
1844–1857. [CrossRef] [PubMed]
30. Patel, N.B.; Pathak, K.K. Pyridoquinolones containing azetidinones: Synthesis and their biological evaluation.
Med. Chem. Res. 2011, 21, 2044–2055. [CrossRef]
31. Patel, N.B.; Chauhan, H.I. Novel pyridoquinolones of sulfonamides, thioureas and amines and their
antimicrobial activity. Indian J. Heterocycl. Chem. 2005, 15, 39–42.
32. Patel, N.B.; Modi, S.H. Synthesis and antimicrobial activity of novel 6-hydroxy-4-oxo-pyrido[2,3-H]
quinoline-3[(substituted aryl ureido/piperazinyl)carbonyl]. Indian J. Pharm. Educational Res. 2010, 44,
8–21.
33. Patel, N.B.; Patel, J.C. Synthesis and antimicrobial study of fluoroquinolone based 4-thiozolidinones.
Med. Chem. Res. 2010, 19, 757–770. [CrossRef]
34. Patel, N.B.; Patel, J.C. Synthesis and antimicrobial activities of 2-azetidinyl-4-quinazolinone derivatives of
diclofenac analogue. Med. Chem. Res. 2011, 20, 511–521. [CrossRef]
35. Patel, N.B.; Patel, J.C.; Modi, S.H. Synthesis and antimicrobial activity of carbonyl pyridoquinolones
containing urea and piperazine residue. J. Saudi Chem. Soc. 2010, 15, 167–176. [CrossRef]
36. Patel, N.B.; Patel, S.D.; Chauhan, H.I. Synthesis and in vitro microbial activities of amides of pyridoquinolone.
Med. Chem. Res. 2011, 20, 1054–1067. [CrossRef]
37. Komarnicka, U.K.; Starosta, R.; Guz-Regner, K.; Bugla-Płoskońska, G.; Kyzioł, A.; Jeżowska-Bojczuk, M.
Phosphine derivatives of sparfloxacin—Synthesis, structures and in vitro activity. J. Mol. Struct. 2015, 1096,
55–63. [CrossRef]
38. Plech, T.; Wujec, M.; Kosikowska, U.; Malm, A.; Rajtar, B.; Polz-Dacewicz, M. Synthesis and in vitro activity
of 1,2,4-triazole-ciprofloxacin hybrids against drug-susceptible and drug-resistant bacteria. Eur. J. Med. Chem.
2013, 60, 128–134. [CrossRef] [PubMed]
39. Parkinson, E.I.; Bair, J.S.; Nakamura, B.A.; Lee, H.Y.; Kuttab, H.I.; Southgate, E.H.; Lezmi, S.;
Lau, G.W.; Hergenrother, P.J. Deoxynybomycins inhibit mutant DNA gyrase and rescue mice infected
with fluoroquinolone-resistant bacteria. Nat. Commun. 2015, 6. [CrossRef] [PubMed]
40. Kern, G. Inhibition of Neisseria gonorrhoeae type II topoisomerases by the novel spiropyrimidinetrione
azd0914. J. Biol. Chem. 2015, 290, 20984–20994. [CrossRef] [PubMed]
41. Soni, K. Fluoroquinolones: Chemistry & action—A review. Indo Global J. Pharm. Sci. 2012, 2, 43–53.
Molecules 2016, 21, 268 15 of 19

42. Mohammadhosseini, N.; Alipanahi, Z.; Alipour, E.; Emami, S.; Faramarzi, M.; Samadi, N.; Khoshnevis, N.;
Shafiee, A.; Foroumadi, A. Synthesis and antibacterial activity of novel levofloxacin derivatives containing a
substituted thienylethyl moiety. DARU J. Pharm. Sci. 2012, 20. [CrossRef] [PubMed]
43. Vijan, L.E.; Giosanu, D. Binding of norfloxacin, enoxacin and enrofloxacin to calf thymus DNA.
Rev. Roum. Chim. 2012, 57, 823–827.
44. Mdluli, K.; Ma, Z. Mycobacterium tuberculosis DNA gyrase as a target for drug discovery. Infect. Disord.
Drug Targets 2007, 7, 159–168. [CrossRef] [PubMed]
45. Hawtin, R.E.; Stockett, D.E.; Byl, J.A.; McDowell, R.S.; Nguyen, T.; Arkin, M.R.; Conroy, A.; Yang, W.;
Osheroff, N.; Fox, J.A. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons
topoisomerase II. PLoS ONE 2010, 5. [CrossRef] [PubMed]
46. Cazedey, E.C.L.; Salgado, H.R. Development and validation of a microbiological agar assay for determination
of orbifloxacin in pharmaceutical preparations. Pharmaceutics 2011, 3, 572–581. [CrossRef] [PubMed]
47. Qin, W.; Panunzio, M.; Biondi, S. Beta-lactam antibiotics renaissance. Antibiotics 2014, 3, 193–215. [CrossRef]
48. Zhao, S.; Li, X.; Ra, Y.; Li, C.; Jiang, H.; Li, J.; Qu, Z.; Zhang, S.; He, F.; Wan, Y. Developing and optimizing an
immunoaffinity cleanup technique for determination of quinolones from chicken muscle. J. Agric. Food Chem.
2009, 57, 365–371. [CrossRef] [PubMed]
49. Owens, R.C.; Ambrose, P.G. Clinical use of the fluoroquinolones. Med. Clin. N. Am. 2000, 84, 1447–1469.
[CrossRef]
50. Long, T.E.; Keding, L.C.; Lewis, D.D.; Anstead, M.I.; Ryan Withers, T.; Yu, H.D. Anionic fluoroquinolones as
antibacterials against biofilm-producing Pseudomonas aeruginosa. Bioorg. Med. Chem. Lett. 2016, 26, 1305–1309.
[CrossRef] [PubMed]
51. Bradley, J.; Jackson, M. The use of systemic and topical fluoroquinolones. Pediatrics 2011, 128, e1034–e1045.
[CrossRef] [PubMed]
52. Takiff, H.; Guerrero, E. Current prospects for the fluoroquinolones as first-line tuberculosis therapy.
Antimicrob. Agents Chemother. 2011, 55, 5421–5429. [CrossRef] [PubMed]
53. Sun, Z.; Zhang, J.; Zhang, X.; Wang, S.; Zhang, Y.; Li, C. Comparison of gyrA gene mutations
between laboratory-selected ofloxacin-resistant Mycobacterium tuberculosis strains and clinical isolates. Int. J.
Antimicrob. Agents 2008, 31, 115–121. [CrossRef] [PubMed]
54. Avalos, E.; Catanzaro, D.; Catanzaro, A.; Ganiats, T.; Brodine, S.; Alcaraz, J.; Rodwell, T. Frequency and
geographic distribution of gyrA and gyrB mutations associated with fluoroquinolone resistance in clinical
Mycobacterium tuberculosis isolates: A systematic review. PLoS ONE 2015, 10, e0120470.
55. Nosova, E.Y.; Bukatina, A.A.; Isaeva, Y.D.; Makarova, M.V.; Galkina, K.Y.; Moroz, A.M. Analysis of mutations
in the gyrA and gyrB genes and their association with the resistance of Mycobacterium tuberculosis to
levofloxacin, moxifloxacin and gatifloxacin. J. Med. Microbiol. 2013, 62, 108–113. [CrossRef] [PubMed]
56. Lu, J.; Liu, M.; Wang, Y.; Pang, Y.; Zhao, Z. Mechanisms of fluoroquinolone monoresistance in mycobacterium
tuberculosis. FEMS Microbiol. Lett. 2014, 353, 40–48. [CrossRef] [PubMed]
57. Zhang, Z.; Lu, J.; Wang, Y.; Pang, Y.; Zhao, Y. Prevalence and molecular characterization of
fluoroquinolone-resistant Mycobacterium tuberculosis isolates in china. Antimicrob. Agents Chemother. 2014, 58,
364–369. [CrossRef] [PubMed]
58. Zhang, Y. Advances in the treatment of tuberculosis. Clin. Pharmacol. Ther. 2007, 82, 595–600. [CrossRef]
[PubMed]
59. Zhu, C.; Zhang, Y.; Shen, Y.; Siu, G.K.H.; Wu, W.; Qian, X.; Deng, G.; Xu, Y.; Lau, R.; Fan, X.; et al. Molecular
characterization of fluoroquinolone-resistant Mycobacterium tuberculosis clinical isolates from Shanghai,
China. Diagn. Microbiol. Infect. Dis. 2012, 73, 260–263. [CrossRef] [PubMed]
60. Shi, R.; Zhang, J.; Li, C.; Kazumi, Y.; Sugawara, I. Emergence of ofloxacin resistance in Mycobacterium
tuberculosis clinical isolates from china as determined by gyrA mutation analysis using denaturing
high-pressure liquid chromatography and DNA sequencing. J. Clin. Microbiol. 2006, 44, 4566–4568.
[CrossRef] [PubMed]
61. Umubyeyi, A.N. Limited fluoroquinolone resistance among Mycobacterium tuberculosis isolates from rwanda:
Results of a national survey. J. Antimicrob. Chemother. 2007, 59, 1031–1033. [CrossRef] [PubMed]
Molecules 2016, 21, 268 16 of 19

62. Hu, Q.; Yin, L.; Ali, A.; Cooke, A.J.; Bennett, J.; Ratcliffe, P.; Lo, M.M.-C.; Metzger, E.; Hoyt, S.; Hartmann, R.W.
Novel pyridyl substituted 4,5-dihydro-[1,2,4]triazolo[4,3-a]quinolines as potent and selective aldosterone
synthase inhibitors with improved in vitro metabolic stability. J. Med. Chem. 2015, 58, 2530–2537. [CrossRef]
[PubMed]
63. Pescatori, L.; Métifiot, M.; Chung, S.; Masoaka, T.; Cuzzucoli Crucitti, G.; Messore, A.; Pupo, G.; Madia, V.N.;
Saccoliti, F.; Scipione, L.; et al. N-substituted quinolinonyl diketo acid derivatives as HIV integrase strand
transfer inhibitors and their activity against RNase H function of reverse transcriptase. J. Med. Chem. 2015,
58, 4610–4623. [CrossRef] [PubMed]
64. Di Santo, R.C.R.; Roux, A.; Artico, M.; Lavecchia, A.; Marinelli, L.; Novellino, E.; Palmisano, L.; Andreotti, M.;
Amici, R.; Galluzzo, C.M.; et al. Novel bifunctional quinolonyl diketo acid derivatives as HIV-1 integrase
inhibitors: Design, synthesis, biological activities, and mechanism of action. J. Med. Chem. 2006, 49,
1939–1945. [CrossRef] [PubMed]
65. Di Santo, R. Diketo acids derivatives as dual inhibitors of human immunodeficiency virus type 1 integrase
and the reverse transcriptase RNase H domain. Curr. Med. Chem. 2011, 18, 3335–3342. [CrossRef] [PubMed]
66. Costi, R.M.M.; Chung, S.; Cuzzucoli Crucitti, G.; Maddali, K.; Pescatori, L.; Messore, A.; Madia, V.N.;
Pupo, G.; Scipione, L.; Tortorella, S.; et al. Basic quinolinonyl diketo acid derivatives as inhibitors of hiv
integrase and their activity against RNase H function of reverse transcriptase. J. Med. Chem. 2014, 57,
3223–3234. [CrossRef] [PubMed]
67. Di Santo, R.C.R.; Roux, A.; Miele, G.; Cuzzucoli Crucitti, G.; Iacovo, A.; Rosi, F.; Lavecchia, A.; Marinelli, L.;
Di Giovanni, C.; Novellino, E.; et al. Novel quinolinonyl diketo acid derivatives as HIV-1 integrase inhibitors:
Design, synthesis, and biological activities. J. Med. Chem. 2008, 51, 4744–4750. [CrossRef] [PubMed]
68. Fu, Y.; Yang, Y.; Zhou, S.; Liu, Y.; Yuan, Y.; Li, S.; Li, C. Ciprofloxacin containing Mannich base and its copper
complex induce antitumor activity via different mechanism of action. Int. J. Oncol. 2014, 45, 2092–2100.
[CrossRef] [PubMed]
69. Boothe, D.; Boeckh, A.; Simpson, B.; Dubose, K. Comparison of pharmacodynamics and pharmacokinetic
indices of efficacy for 5 fluoroquinolones toward pathogens of dogs and cats. J. Vet. Inter. Med. 2006, 20,
1297–1306. [CrossRef]
70. Mueller, R.S.; Stephan, B. Pradofloxacin in the treatment of canine deep pyoderma: A multicentred, blinded,
randomized parallel trial. Vet. Dermatol. 2007, 18, 144–151. [CrossRef] [PubMed]
71. Champoux, J.J. DNA topoisomerases: Structure, function, and mechanism. Annu. Rev. Biochem. 2001, 70,
369–413. [CrossRef] [PubMed]
72. Dale, A.G.; Hinds, J.; Mann, J.; Taylor, P.W.; Neidle, S. Symmetric bis-benzimidazoles are potent
anti-staphylococcal agents with dual inhibitory mechanisms against DNA gyrase. Biochemistry 2012, 51,
5860–5871. [CrossRef] [PubMed]
73. Deweese, J.E.; Osheroff, N. The DNA cleavage reaction of topoisomerase II: Wolf in sheep’s clothing.
Nucleic Acids Res. 2009, 37, 738–748. [CrossRef] [PubMed]
74. Corbett, K.D.; Shultzaberger, R.K.; Berger, J.M. The c-terminal domain of DNA gyrase A adopts a
DNA-bending beta-pinwheel fold. Proc. Natl. Acad. Sci. USA 2004, 101, 7293–7298. [CrossRef] [PubMed]
75. Levine, C.; Hiasa, H.; Marians, K.J. DNA gyrase and topoisomerase IV: Biochemical activities, physiological
roles during chromosome replication, and drug sensitivities. Biochim. Biophys. Acta 1998, 1400, 29–43.
[CrossRef]
76. Schoeffler, A.J.; Berger, J.M. DNA topoisomerases: Harnessing and constraining energy to govern
chromosome topology. Q. Rev. Biophys. 2008, 41, 41–101. [CrossRef] [PubMed]
77. Anderson, V.E.; Osheroff, N. Type II topoisomerases as targets for quinolone antibacterials turning Dr. Jekyll
into Mr. Hyde. Curr. Pharm. Des. 2001, 7, 337–353. [CrossRef] [PubMed]
78. Schoeffler, A.J.; Berger, J.M. Recent advances in understanding structure—function relationships in the type
II topoisomerase mechanism. Biochem. Soc. Trans. 2005, 33, 1465–1470. [CrossRef] [PubMed]
79. Schoeffler, A.J.; May, A.P.; Berger, J.M. A domain insertion in Escherichia coli gyrB adopts a novel fold that
plays a critical role in gyrase function. Nucleic Acids Res. 2010, 38, 7830–7844. [CrossRef] [PubMed]
80. Schmidt, B.H.; Burgin, A.B.; Deweese, J.E.; Osheroff, N.; Berger, J.M. A novel and unified two-metal
mechanism for DNA cleavage by type II and Ia topoisomerases. Nature 2010, 465, 641–644. [CrossRef]
[PubMed]
Molecules 2016, 21, 268 17 of 19

81. Pitts, S.L.; Liou, G.F.; Mitchenall, L.A.; Burgin, A.B.; Maxwell, A.; Neuman, K.C.; Osheroff, N. Use of
divalent metal ions in the DNA cleavage reaction of topoisomerase IV. Nucleic Acids Res. 2011, 39, 4808–4817.
[CrossRef] [PubMed]
82. Deweese, J.E.; Osheroff, M.A.; Osheroff, N. DNA topology and topoisomerases. Biochem. Mol. Biol. Educ.
2009, 37, 2–10. [CrossRef] [PubMed]
83. Laponogov, I.; Pan, X.-S.; Veselkov, D.A.; McAuley, K.E.; Fisher, L.M.; Sanderson, M.R. Structural basis of
gate-DNA breakage and resealing by type II topoisomerases. PLoS ONE 2010, 5, e11338. [CrossRef]
84. Laponogov, I.; Sohi, M.K.; Veselkov, D.; Pan, X.; Sawhney, R.; Thompson, A.; McAuley, K.; Fisher, L.;
Sanderson, M. Structural insight into the quinolone–DNA cleavage complex of type IIa topoisomerases.
Nat. Struct. Mol. Biol. 2009, 16, 667–669. [CrossRef] [PubMed]
85. Baker, N.M.; Weigand, S.; Maar-Mathias, S.; Mondragon, A. Solution structures of DNA-bound gyrase.
Nucleic Acids Res. 2011, 39, 755–766. [CrossRef] [PubMed]
86. Aldred, K.J.; Schwanz, H.A.; Li, G.; Williamson, B.H.; McPherson, S.A.; Turnbough, C.L., Jr.; Kerns, R.J.;
Osheroff, N. Activity of quinolone cp-115,955 against bacterial and human type II topoisomerases is mediated
by different interactions. Biochemistry 2015, 54, 1278–1286. [CrossRef] [PubMed]
87. Kreuzer, K.N.; Cozzarelli, N.R. Escherichia coli mutants thermosensitive for deoxyribonucleic acid gyrase
subunit A: Effects on deoxyribonucleic acid replication, transcription, and bacteriophage growth. J. Bacteriol.
1979, 140, 424–435. [PubMed]
88. Hooper, D.C. Mechanisms of Quinolone Resistance. In Quinolone Antimicrobial Agents, 3rd ed.; ASM Press:
Washington, DC, USA, 2003; pp. 41–67.
89. Hooper, D.C.; Jacoby, G.A. Mechanisms of drug resistance: Quinolone resistance. Ann. N. Y. Acad. Sci. 2015,
1354, 12–31. [CrossRef] [PubMed]
90. Fournier, B.; Zhao, X.; Lu, T.; Drlica, K.; Hooper, D.C. Selective targeting of topoisomerase iv and DNA
gyrase in Staphylococcus aureus: Different patterns of quinolone-induced inhibition of DNA synthesis.
Antimicrob. Agents Chemother. 2000, 44, 2160–2165. [CrossRef] [PubMed]
91. Price, L.B.; Vogler, A.; Pearson, T.; Busch, J.D.; Schupp, J.M.; Keim, P. In vitro selection and characterization
of Bacillus anthracis mutants with high-level resistance to ciprofloxacin. Antimicrob. Agents Chemother. 2003,
47, 2362–2365. [CrossRef] [PubMed]
92. Morgan-Linnell, S.K.; Boyd, L.B.; Steffen, D.; Zechiedrich, L. Mechanisms accounting for fluoroquinolone
resistance in Escherichia coli clinical isolates. Antimicrob. Agents Chemother. 2009, 53, 235–241. [CrossRef]
[PubMed]
93. Sissi, C.; Perdona, E.; Domenici, E.; Feriani, A.; Howells, A.J.; Maxwell, A.; Palumbo, M. Ciprofloxacin affects
conformational equilibria of DNA gyrase A in the presence of magnesium ions. J. Mol. Biol. 2001, 311,
195–203. [CrossRef] [PubMed]
94. Sissi, C.; Palumbo, M. Effects of magnesium and related divalent metal ions in topoisomerase structure and
function. Nucleic Acids Res. 2009, 37, 702–711. [CrossRef] [PubMed]
95. Barnard, F.M.; Maxwell, A. Interaction between DNA gyrase and quinolones: Effects of alanine mutations
at gyrA subunit residues Ser83 and Asp87. Antimicrob. Agents Chemother. 2001, 45, 1994–2000. [CrossRef]
[PubMed]
96. Chan, P.F.; Srikannathasan, V.; Huang, J.; Cui, H.; Fosberry, A.P.; Gu, M.; Hann, M.M.; Hibbs, M.; Homes, P.;
Ingraham, K.; et al. Structural basis of DNA gyrase inhibition by antibacterial QPT-1, anticancer drug
etoposide and moxifloxacin. Nat. Commun. 2015, 6. [CrossRef] [PubMed]
97. Bax, B.C.P.; Eggleston, D.; Fosberry, A.; Gentry, D.; Gorrec, F.; Giordano, I.; Hann, M.; Hennessy, A.; Hibbs, M.;
Huang, J.; et al. Type IIa topoisomerase inhibition by a new class of antibacterial agents. Nature 2010, 466,
935–940. [CrossRef] [PubMed]
98. Srikannathasan, V.; Wohlkonig, A.; Shillings, A.; Singh, O.; Chan, P.F.; Huang, J.; Gwynn, M.N.; Fosberry, A.P.;
Homes, P.; Hibbs, M.; et al. Crystallization and initial crystallographic analysis of covalent DNA-cleavage
complexes of Staphyloccocus aureus DNA gyrase with QPT-1, moxifloxacin and etoposide. Acta Crystallogr. F
Struct. Biol. Commun. 2015, 71, 1242–1246. [CrossRef] [PubMed]
99. Wohlkonig, A. Structural basis of quinolone inhibition of type IIa topoisomerases and target-mediated
resistance. Nat. Struct. Mol. Biol. 2010, 17, 1152–1153. [CrossRef] [PubMed]
100. Collin, F.; Karkare, S.; Maxwell, A. Exploiting bacterial DNA gyrase as a drug target: Current state and
perspectives. Appl. Microbiol. Biotechnol. 2011, 92, 479–497. [CrossRef] [PubMed]
Molecules 2016, 21, 268 18 of 19

101. Aldred, K.J.; Breland, E.J.; Vlckova, V.; Strub, M.-P.; Neuman, K.C.; Kerns, R.J.; Osheroff, N. Role of the
water metal ion bridge in mediating interactions between quinolones and Escherichia coli topoisomerase IV.
Biochemistry 2014, 53, 5558–5567. [CrossRef] [PubMed]
102. Aldred, K.J.; McPherson, S.A.; Turnbough, C.L., Jr.; Kerns, R.J.; Osheroff, N. Topoisomerase IV-quinolone
interactions are mediated through a water-metal ion bridge: Mechanistic basis of quinolone resistance.
Nucleic Acids Res. 2013, 41, 4628–4639. [CrossRef] [PubMed]
103. Aldred, K.J.; McPherson, S.A.; Wang, P.; Kerns, R.J.; Graves, D.E.; Turnbough, C.L., Jr.; Osheroff, N. Drug
interactions with Bacillus anthracis topoisomerase IV: Biochemical basis for quinolone action and resistance.
Biochemistry 2012, 51, 370–381. [CrossRef] [PubMed]
104. Aldred, K.J.; Schwanz, H.A.; Li, G.; McPherson, S.A.; Turnbough, C.L., Jr.; Kerns, R.J.;
Osheroff, N. Overcoming target-mediated quinolone resistance in topoisomerase IV by introducing
metal-ion-independent drug-enzyme interactions. ACS Chem. Biol. 2013, 8, 2660–2668. [CrossRef] [PubMed]
105. Baldwin, E.L.; Osheroff, N. Etoposide, topoisomerase ii and cancer. Curr. Med. Chem. Anticancer Agents 2005,
5, 363–372. [CrossRef] [PubMed]
106. Hsiung, Y.; Elsea, S.H.; Osheroff, N.; Nitiss, J.L. A mutation in yeast top2 homologous to a quinolone-resistant
mutation in bacteria. Mutation of the amino acid homologous to Ser83 of Escherichia coli GyrA alters
sensitivity to eukaryotic topoisomerase inhibitors. J. Biol. Chem. 1995, 270, 20359–20364. [CrossRef]
[PubMed]
107. Lee, S.; Jung, S.-R.; Heo, K.; Byl, J.A.W.; Deweese, J.E.; Osheroff, N.; Hohng, S. DNA cleavage and opening
reactions of human topoisomerase iiα are regulated via Mg2+ -mediated dynamic bending of gate-DNA.
Proc. Natl. Acad. Sci. USA 2012, 109, 2925–2930. [CrossRef] [PubMed]
108. Schmidt, B.H.; Osheroff, N.; Berger, J.M. Structure of a topoisomerase ii-DNA-nucleotide complex reveals a
new control mechanism for ATPase activity. Nat. Struct. Mol. Biol. 2012, 19, 1147–1154. [CrossRef] [PubMed]
109. Jacoby, G.A. Mechanisms of resistance to quinolones. Clin. Infect. Dis. 2005, 41, 120–126. [CrossRef]
[PubMed]
110. Redgrave, L.S.; Sutton, S.B.; Webber, M.A.; Piddock, L.J. Fluoroquinolone resistance: Mechanisms, impact on
bacteria, and role in evolutionary success. Trends Micobiol. 2014, 22, 438–445. [CrossRef] [PubMed]
111. Pan, X.-S.; Gould, K.A.; Fisher, L.M. Probing the differential interactions of quinazolinedione pd 0305970 and
quinolones with gyrase and topoisomerase IV. Antimicrob. Agents Chemother. 2009, 53, 3822–3831. [CrossRef]
[PubMed]
112. Yague, G.; Morris, J.E.; Pan, X.-S.; Gould, K.A.; Fisher, L.M. Cleavable-complex formation by wild-type and
quinolone-resistant streptococcus pneumoniae type II topoisomerases mediated by gemifloxacin and other
fluoroquinolones. Antimicrob. Agents Chemother. 2002, 46, 413–419. [CrossRef] [PubMed]
113. Pfeiffer, E.S.; Hiasa, H. Determination of the primary target of a quinolone drug and the effect of
quinolone resistance-conferring mutations by measuring quinolone sensitivity based on its mode of action.
Antimicrob. Agents Chemother. 2007, 51, 3410–3412. [CrossRef] [PubMed]
114. Drlica, K.; Hiasa, H.; Kerns, R.; Malik, M.; Mustaev, A.; Zhao, X. Quinolones: Action and resistance updated.
Curr. Top. Med. Chem. 2009, 9, 981–988. [CrossRef] [PubMed]
115. Wang, M.; Tran, J.H.; Jacoby, G.A.; Zhang, Y.; Wang, F.; Hooper, D.C. Plasmid-mediated quinolone resistance
in clinical isolates of Escherichia coli from shanghai, china. Antimicrob. Agents Chemother. 2003, 47, 2242–2248.
[CrossRef] [PubMed]
116. Rodriguez-Martinez, J.M.; Cano, M.E.; Velasco, C.; Martinez-Martinez, L.; Pascual, A. Plasmid-mediated
quinolone resistance: An update. J. Infect. Chemother. 2011, 17, 149–182. [CrossRef] [PubMed]
117. Li, Z.; Deguchi, T.; Yasuda, M.; Kawamura, T.; Kanematsu, E.; Nishino, Y.; Ishihara, S.; Kawada, Y. Alteration
in the gyra subunit of DNA gyrase and the parc subunit of DNA topoisomerase IV in quinolone-resistant
clinical isolates of Staphylococcus epidermidis. Antimicrob. Agents Chemother. 1998, 42, 3293–3295. [PubMed]
118. Robicsek, A.; Jacoby, G.A.; Hooper, D.C. The worldwide emergence of plasmid-mediated quinolone
resistance. Lancet Infect. Dis. 2006, 6, 629–640. [CrossRef]
119. Strahilevitz, J.; Jacoby, G.A.; Hooper, D.C.; Robicsek, A. Plasmid-mediated quinolone resistance: A
multifaceted threat. Clin. Microbiol. Rev. 2009, 22, 664–689. [CrossRef] [PubMed]
120. Martinez-Martinez, L.; Pascual, A.; Jacoby, G.A. Quinolone resistance from a transferable plasmid. Lancet
1998, 351, 797–799. [CrossRef]
Molecules 2016, 21, 268 19 of 19

121. Tran, J.H.; Jacoby, G.A. Mechanism of plasmid-mediated quinolone resistance. Proc. Natl. Acad. Sci. USA
2002, 99, 5638–5642. [CrossRef] [PubMed]
122. Xiong, X.; Bromley, E.H.C.; Oelschlaeger, P.; Woolfson, D.N.; Spencer, J. Structural insights into quinolone
antibiotic resistance mediated by pentapeptide repeat proteins: Conserved surface loops direct the activity of
a qnr protein from a gram-negative bacterium. Nucleic Acids Res. 2011, 39, 3917–3927. [CrossRef] [PubMed]
123. Guillard, T.; Cambau, E.; Chau, F.; Massias, L.; de Champs, C.; Fantin, B. Ciprofloxacin treatment failure in
a murine model of pyelonephritis due to an AAC (6’)-Ib-cr-producing Escherichia coli strain susceptible to
ciprofloxacin in vitro. Antimicrob. Agents Chemother. 2013, 57, 5830–5835. [CrossRef] [PubMed]
124. Poole, K. Efflux pumps as antimicrobial resistance mechanisms. Ann. Med. 2007, 39, 162–176. [CrossRef]
[PubMed]
125. Goldman, J.D.; White, D.G.; Levy, S.B. Multiple antibiotic resistance (mar) locus protects Escherichia coli from
rapid cell killing by fluoroquinolones. Antimicrob. Agents Chemother. 1996, 40, 1266–1269. [PubMed]

© 2016 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons by Attribution
(CC-BY) license (http://creativecommons.org/licenses/by/4.0/).

You might also like