ArticlePDF AvailableLiterature Review

Abstract and Figures

Since their discovery in the early 1960s, the quinolone group of antibacterials has generated considerable clinical and scientific interest. Nalidixic acid, the first quinolone to be developed, was obtained as an impurity during the manufacture of quinine. Since this time, many derivatives have been synthesized and evaluated for their antibacterial potency. Two major groups of compounds have been developed from the basic molecule: quinolones and naphthyridones. Manipulations of the basic molecule, including replacing hydrogen with fluorine at position 6, substituting a diamine residue at position 7 and adding new residues at position 1 of the quinolone ring, have led to enhanced antibacterial efficacy. In general these compounds are well tolerated. However, some of these structural changes have been found to correlate with specific adverse events: the addition of fluorine or chlorine at position 8 being associated with photo-reactivity, e.g. Bay y 3118 and sparfloxacin; and the substitution of an amine or a methyl group at position 5 having a potential role in QTc prolongation, e.g. sparfloxacin and grepafloxacin. Progressive modifications in molecular configuration have resulted in improved breadth and potency of in vitro activity and pharmacokinetics. One of the most significant developments has been the improved anti-Gram-positive activity of the newer compounds, such as moxifloxacin and garenoxacin. In the current millennium, these new agents may play an important role in the treatment of respiratory infections.
Content may be subject to copyright.
Journal of Antimicrobial Chemotherapy (2003)
51
, Suppl. S1, 1–11
DOI: 10.1093/jac/dkg212
1
...................................................................................................................................................................................................................................................................
© 2003 The British Society for Antimicrobial Chemotherapy
Development of the quinolones
Monique I. Andersson and Alasdair P. MacGowan*
Bristol Centre for Antimicrobial Research and Evaluation, North Bristol NHS Trust and University of Bristol,
Department of Medical Microbiology, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
Since their discovery in the early 1960s, the quinolone group of antibacterials has generated
considerable clinical and scientific interest. Nalidixic acid, the first quinolone to be developed,
was obtained as an impurity during the manufacture of quinine. Since this time, many deriva-
tives have been synthesized and evaluated for their antibacterial potency. Two major groups of
compounds have been developed from the basic molecule: quinolones and naphthyridones.
Manipulations of the basic molecule, including replacing hydrogen with fluorine at position 6,
substituting a diamine residue at position 7 and adding new residues at position 1 of the quino-
lone ring, have led to enhanced antibacterial efficacy. In general these compounds are well toler-
ated. However, some of these structural changes have been found to correlate with specific
adverse events: the addition of fluorine or chlorine at position 8 being associated with photo-
reactivity, e.g. Bay y 3118 and sparfloxacin; and the substitution of an amine or a methyl group at
position 5 having a potential role in QTc prolongation, e.g. sparfloxacin and grepafloxacin. Pro-
gressive modifications in molecular configuration have resulted in improved breadth and
potency of
in vitro
activity and pharmacokinetics. One of the most significant developments has
been the improved anti-Gram-positive activity of the newer compounds, such as moxifloxacin
and garenoxacin. In the current millennium, these new agents may play an important role in the
treatment of respiratory infections.
Keywords: quinolones, pharmacokinetics, pharmacodynamics, drug development
Introduction
Quinolones have been the centre of considerable scientific
and clinical interest since their discovery in the early 1960s.
This is because they potentially offer many of the attributes of
an ideal antibiotic, combining high potency, a broad spectrum
of activity, good bioavailability, oral and intravenous formu-
lations, high serum levels, a large volume of distribution indi-
cating concentration in tissues and a potentially low incidence
of side-effects. Much research has attempted to make these
potential attributes real. Nalidixic acid was the first quinolone
to be developed (Table 1), but it took more than a decade
before additional compounds, such as flumequin, norfloxacin
and enoxacin became available for clinical use. The main use
for all these agents was the treatment of urinary tract infec-
tion. In the late 1980s more systemically active drugs, such as
ciprofloxacin and ofloxacin, were marketed. Recently, there
has been a considerable increase in the number of agents that
are in development, and to date over 10 000 molecules have
been patented. This review will trace the development of
quinolones from their sole use as treatment for urinary tract
infections, via systemic use, to those agents whose primary
use is for the treatment of respiratory tract infections. This
development may be described in terms of structures,
potency, pharmacokinetics and pharmacodynamics, and
finally indications and clinical use.
Structural developments
Quinolones were derived from quinine. Figure 1 shows the
basic fluoroquinolone molecule or pharmacore.
1
The addi-
tion of a fluorine molecule at position 6 was one of the earliest
changes to the structure. This single alteration provides a
more than 10-fold increase in gyrase inhibition and up to
100-fold improvement in MIC. Two major groups have been
developed from the basic structure: quinolones and naph-
thyridones.
15
The presence of a nitrogen at position 8 identi-
..................................................................................................................................................................................................................................................................
*Corresponding author. Tel: +44-117-959-5652; Fax: +44-117-959-3154; E-mail: alasdair.macgowan@north-bristol.swest.nhs.uk
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
M. I. Andersson and A. P. MacGowan
2
fies the naphthyridones, a carbon and associated group at
position 8 identifies the quinolones.
15
The quinolones and napththyridones were further enhanced
by the addition of groups to the N
1
, C-5 and C-7 positions of
their respective basic molecules. The addition of piperazine to
the C-7 position (e.g. norfloxacin) improves activity against
Gram-negative organisms. There are data to suggest that a
piperazine ring may play a role in inhibiting efflux mechan-
isms, thereby improving the potency of these drugs. The
structure of norfloxacin illustrates these developments and
subsequent to this all quinolones (except garenoxacin) have a
fluorine at position 6 and many have six-membered rings at
position C-7 (Figure 2). The presence of a pyrrolodinyl group
at position C-7 (e.g. clinafloxacin) improves activity against
Gram-positive organisms. In addition to piperazine at the C-7
position, a cyclopropyl group was introduced to the N
1
posi-
tion and is best exemplified by ciprofloxacin, which was first
synthesized in 1983 (Figure 3). This increases the potency of
the drug and many subsequent quinolones have a cyclopropyl
group (e.g. grepafloxacin, moxifloxacin, gatifloxacin and
garenoxacin; Figure 4). The addition of a 2,4-difluorophenyl
group at position 1 (e.g. trovafloxacin) also improves
potency, especially in improving activity against anaerobes.
A number of other structural manipulations have been tried
to improve the anti-Gram-positive activity of fluoroquino-
lones. One of the first additions was an NH
2
group at position
Table 1.
Chronology of quinolones that reached the UK
market
Date Quinolone
19601969 nalidixic acid
19701975 cinoxacin
19751985 norfloxacin
19851990 ciprofloxacin, ofloxacin
19901995 temafloxacin, sparfloxacin
19952000 grepafloxacin, levofloxacin, trovafloxacin
20002005 moxifloxacin, possibly gemifloxacin and
garenoxacin in 2003 or later
Figure 1.
Structure of the quinolone and napthyridone molecule. In
molecules where X is a carbon atom, the molecule is a quinolone
(cinoxacin, norfloxacin, ofloxacin, ciprofloxacin, temafloxacin, spar-
floxacin, grepafloxacin, levofloxacin, clinafloxacin, moxifloxacin, gati-
floxacin). Where X is a nitrogen atom the molecule is a naphthyridone
(nalidixic acid, enoxacin, tosufloxacin, trovafloxacin, gemifloxacin).
Adapted from Domagala (1994).
2
Figure 2.
Modifications of quinolone structures: norfloxacin. In all
subsequent molecules, except garenoxacin, fluorine was retained at
position 6. A six-membered ring at position 7 is found in ciprofloxacin,
temafloxacin, sparfloxacin, ofloxacin and levofloxacin.
Figure 3.
Modification of quinolone structures: ciprofloxacin. A cyclo-
propyl group at position 1 is found in sparfloxacin, grepafloxacin, clina-
floxacin, gatifloxacin, moxifloxacin and garenoxacin.
Figure 4.
Modification of quinolone structures: moxifloxacin. A five-
membered ring or azabiocyclo rings at position 7 are found in clina-
floxacin and trovafloxacin (five-membered ring) or moxifloxacin and
garenoxacin (azabicyclo rings). A methoxy group at position 8 is found
in moxifloxacin and gatifloxacin. A difluoromethyl ether (OHF
2
) group
is found at position 8 in garenoxacin.
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
Development of the quinolones
3
C-5, which resulted in a general increase in anti-Gram-positive
activity. This is seen with sparfloxacin which otherwise has a
very similar structure to ciprofloxacin. Sparfloxacin also has
a fluorine at position C-6, a piperazine at position C-7 and is
alkylated. Grepafloxacin is also substituted at position C-5
but by a CH
3
group and has improved anti-Gram-positive
potency compared with ciprofloxacin. The substituents at
position C-7 are associated with a number of key attributes,
such as antibacterial spectrum, bioavailability and side-effects.
The most common substituents are cyclic amino groups,
for example piperazine or pyrrolidine rings; other groups
have been less successful. Piperazine rings are particularly
common (e.g. norfloxacin, enoxacin or ciprofloxacin) and
confer potency against Gram-negative bacteria. The addition
of methyl groups can improve both oral absorption and
in vivo
activity. However, the improved activity against
Gram-positive bacteria can sometimes be at the expense of
activity against
Pseudomonas aeruginosa
.
Pyrrolidine rings (five-membered) are also common sub-
stituents at position 7, and are associated with enhanced
potency against Gram-positive bacteria. However, this group
is associated with low water solubility and low oral bioavail-
ability so
in vivo
activity may be compromised. Introduction
of methyl groups on the pyrrolidine ring helps to overcome
some of these physical properties. Gemifloxacin, a naphthyri-
done, is a good example of the advantages and disadvantages
associated with a pyrrolidine ring at position 7.
6
The addition
of azabicyclo groups onto position 7 has resulted in agents
(moxifloxacin and trovafloxacin) with significant anti-Gram-
positive activity, marked lipophilicity and half-lives of
>10 h.
7,8
Manipulation of the group at position 8 has also been
shown to play a role in altering oral pharmacokinetics, broad-
ening the spectrum of activity and reducing the selection of
mutants.
913
Whilst alkylation has been shown to increase
further anti-Gram-positive activity, it also improves tissue
penetration and extends the half-life by increasing lipophil-
icity, as with grepafloxacin, levofloxacin and sparfloxacin.
The structural changes to the quinolone molecule and
correlation with adverse events are now well documented.
3,14
Photo-reactivity is probably most influenced by position 8,
with fluorine or chlorine producing most phototoxic potential
(e.g. lomefloxacin, clinafloxacin, Bay y 3118 and spar-
floxacin) and methoxy groups the least (e.g. moxifloxacin or
gatifloxacin). Garenoxacin has fluorine incorporated through
a C-8 difluoromethyl ether linkage as there is no fluorine at
C-6. It has been suggested that substitution at position 5 may
have a role in QTc prolongation as those agents that have been
associated with significant problems, such as sparfloxacin
and grepafloxacin, have either a NH
2
or a CH
3
group in this
position.
15,16
Much speculation has also surrounded the likely structural
correlates of the haemolytic uraemic-like syndrome caused
by temafloxacin,
17
hepatic eosinophilia caused by trova-
floxacin,
18
pulmonary interstitial eosinophilia and other
immunological side-effects caused by tosufloxacin
19
and
gemifloxacin, and the hypoglycaemia seen with temafloxacin
and clinafloxacin.
18,20,21
Although a number of these agents
are naphthyridones (trovafloxacin, tosufloxacin and gemi-
floxacin) others are fluoroquinolones (temafloxacin), hence it
is unclear whether this difference is relevant. A more power-
ful association is that of the 2,4-difluorophenyl group at posi-
tion 1, as this is shared by trovafloxacin, temafloxacin and
tosufloxacin
1
but not by gemifloxacin.
6
A further hypothesis
is that metabolites of these agents (as yet not fully identified),
which share common structures, may be responsible for some
of the immunologically mediated adverse events seen with
these drugs.
The naphthyridones can be modified by the addition of
cyclopropyl groups in the same way as the quinolones. A five-
membered ring has been added to the molecule of gemi-
floxacin at position 7. Trovafloxacin has an azabicyclo ring,
which improves anti-Gram-positive activity, increases the
half-life and results in resistance to efflux pumps.
8
Developments in potency
The development of these agents can be described in terms
of their potency against Gram-positive, Gram-negative or
atypical bacteria. In terms of anti-Gram-negative potency,
as indicated by MIC
90
s, the activity of quinolones against
Enterobacteriaceae such as
Escherichia coli
and
Klebsiella
spp
.
has not changed significantly since the development of
norfloxacin (Table 2).
6,2231
In terms of their MIC
90
s, the early
drugs (nalidixic acid and cinoxacin) did not have very good
anti-pseudomonal potency. In general, the newer agents have
improved activity against pathogens such as
Mycoplasma
pneumoniae
and other atypical bacteria, and some have
improved activity against Gram-negative anaerobes, such as
Bacteroides fragilis
(Tables 2 and 3). However, many of the
newer agents, such as gatifloxacin, gemifloxacin, gare-
noxacin and moxifloxacin, are not as potent as ciprofloxacin
against
P. aeruginosa
.
One of the more interesting developments in terms of
potency is the area of anti-Gram-positive activity. Although
it is unlikely any fluoroquinolone will have activity against
ciprofloxacin-resistant
Staphylococcus aureus
, a major
development has been in the activity of these drugs against
ciprofloxacin-sensitive
S. aureus, Streptococcus pneumoniae
and Group A streptococci.
10,12,3235
This is one of the key
advances in terms of the development of these agents.
Table 4
2231,3639
lists the fluoroquinolones on the basis of
their MIC
90
s for five Gram-positive species. Nalidixic acid,
cinoxacin and enoxacin have no activity at all, whereas gemi-
floxacin has an MIC
90
of 0.03 mg/L for
S. pneumoniae
, and is
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
M. I. Andersson and A. P. MacGowan
4
equivalent to the earlier compounds in terms of activity
against the majority of Gram-negatives (Table 2). The activ-
ity against enterococci is less clearly defined because differ-
ent species are often classified together, and the proportions
of
Enterococcus faecium
and
Enterococcus faecalis
are not
provided. This is important as
E. faecium
is more resistant
than
E. faecalis
.
Developments in pharmacodynamics and
pharmacokinetics
Pharmacokinetics (PK) refers to changes in drug concen-
tration as the drug moves through the body, whilst pharmaco-
dynamics (PD) refers to how the drug action changes with
concentration or dose.
T
a
bl
e
2
.
Deve
l
opment o
f
qu
i
no
l
one potency aga
i
nst
G
ram-negat
i
ve
b
acter
i
a
MIC
90
(mg/L)
Quinolone
E. coli
Klebsiella
spp.
Enterobacter
/
Citrobacter
spp.
Serratia
spp.
Haemophilus
influenzae P. aeruginosa B. fragilis
Nalidixic acid 8 16 >64 >64 2 >64 >64
Cinoxacin 8 8 >64 >64 2 >64 >64
Enoxacin 0.25 2 1 4 0.12 2 >64
Norfloxacin 0.12 0.5 0.25 2 0.06 2 >64
Ciprofloxacin 0.03 0.25 0.12 0.5 0.03 1 16
Ofloxacin 0.12 0.5 0.5 1 0.03 4 16
Levofloxacin 0.12 0.25 0.5 0.5 0.03 2 8
Temafloxacin 0.06 0.5 0.5 0.5 0.03 1 4
Trovafloxacin 0.06 0.25 0.06 1 0.01 1 0.25
Clinafloxacin 0.01 0.03 0.12 0.25 0.01 0.5 0.25
Sparfloxacin 0.06 0.5 0.5 4 0.03 4 4
Grepafloxacin 0.06 0.12 0.5 2 0.01 8 8
Moxifloxacin 0.06 0.12 1 2 0.06 8 1
Gatifloxacin 0.06 0.25 0.5 1 0.03 4 1
Gemifloxacin 0.03 0.25 0.5 2 0.06 4 ND
Garenoxacin 0.06 0.5 1.0 2 0.03 16 1
Table 3.
Development of quinolone potency against atypical bacteria
MIC
90
(mg/L)
Quinolone
Legionella
pneumophila
M. pneumoniae Chlamydia
spp
. Mycoplasma
hominis
Ureaplasma
urealyticum
Nalidixic acid 0.25 >64 >64 >64 >64
Enoxacin 0.12 4 8 8 16
Norfloxacin 0.06 16 16 8 16
Ciprofloxacin 0.01 2 2 1 4
Ofloxacin 0.03 2 2 0.5 1
Levofloxacin 0.01 1 1 0.25 1
Trovafloxacin 0.01 0.25 0.12 0.5 0.5
Clinafloxacin 0.008 0.03 0.12 0.03 0.12
Sparfloxacin 0.06 0.5 0.5 0.5 0.5
Grepafloxacin 0.01 0.5 0.12 0.12 0.5
Moxifloxacin 0.01 0.12 0.12 0.06 0.25
Gatifloxacin 0.01 0.12 0.25 0.12 0.25
Gemifloxacin 0.008 0.12 0.06 0.01 0.25
Garenoxacin 0.008 0.06 0.016 0.03 0.06
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
Development of the quinolones
5
Nalidixic acid is a drug that is excreted in urine and has very
variable systemic absorption. Essentially it is not a systemic
drug, but a urinary agent. Over time, larger serum area under
the curve (AUC) or peak serum (
C
max
) values are consistently
seen as the quinolones have developed, and oral absorption
has markedly improved compared with that of nalidixic acid
(Table 5).
6,30,31,40,41
The half-life of these agents has also
tended to increase as structural modifications have been made
to the molecule. However, since the activity of quinolones is
concentration dependent, meaning that the extent of bacterial
killing increases as drug concentrations increase, and they
have a prolonged post-antibiotic effect (PAE),
42
a long half-
life is not an absolute prerequisite for once-daily dosing.
14,43,44
No trend in protein binding has become apparent over time
with some agents having a binding of <30% (norfloxacin,
temafloxacin and gatifloxacin) and others >70% (nalidixic
T
a
bl
e
4
.
Deve
l
opment o
f
qu
i
no
l
one potency aga
i
nst
G
ram-pos
i
t
i
ve
b
acter
i
a
MIC
90
(mg/L)
Quinolone
S. aureus
(methicillin sensitive)
S. pneumoniae
Group A
streptococci
Enterococcus
spp.
Clostridium
perfringens
Nalidixic acid >64 >64 >64 >64 >64
Cinoxacin >64 >64 >64 >64 >64
Enoxacin 2 64 64 8 >64
Ciprofloxacin 1 2 1 4 0.5
Ofloxacin 0.5 2 2 2 1
Levofloxacin 0.25 1 1 2 0.5
Temafloxacin 0.25 0.5 0.5 2 1
Sparfloxacin 0.12 0.5 1.0 2 0.25
Grepafloxacin 0.12 0.25 1.0 4 1
Gatifloxacin 0.25 0.25 0.25 1 0.5
Trovafloxacin 0.03 0.12 0.25 1 0.25
Moxifloxacin 0.06 0.12 0.25 2 0.25
Clinafloxacin 0.06 0.12 0.06 0.25 0.12
Gemifloxacin 0.06 0.03 0.06 4 ND
Garenoxacin 0.03 0.12 0.25 0.5 0.25
Tabl e 5 .
Development of quinolone pharmacokinetics
Quinolone
Dose (g)
(frequency
per day)
C
max
(mg/L) AUC (mg
h/L)
Half-life
(h)
Protein binding
(%) Elimination route
Nalidixic acid 1 (
×
4) variable variable 1.5 90 renal
Enoxacin 0.5 (
×
2) 1.5 19 2 60 renal
Enoxacin 0.6 (
×
1) 3.7 29 6 70 renal
Norfloxacin 0.4 (
×
2) 1.5 10 3 15 renal and hepatic
Ciprofloxacin 0.75 (
×
2) 3.5 30 4 40 renal and enteral
Ofloxacin 0.4 (
×
2) 4.8 64 6 40 renal
Levofloxacin 0.5 (
×
1) 5.2 48 7 40 renal
Temafloxacin 0.6 (
×
2) 7.0 134 8 25 renal
Trovafloxacin 0.3 (
×
1) 2.5 40 12 85 hepatic
Clinafloxacin 0.2 (
×
2) 1.6 18 6 40 renal
Sparfloxacin 0.4 (
×
1) 1.0 20 18 40 renal and hepatic
Grepafloxacin 0.4 (
×
1) 1.4 14 14 50 hepatic
Moxifloxacin 0.4 (
×
1) 3.1 30 13 50 hepatic
Gatifloxacin 0.4 (
×
1) 4.0 37 9 20 renal
Gemifloxacin 0.32 (
×
1) 1.0 9 7 60 renal and other
Garenoxacin 0.4 (
×
1) 5.8 59 15 87 renal and other
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
M. I. Andersson and A. P. MacGowan
6
acid, enoxacin, trovafloxacin and garenoxacin). Non-renal
clearance seems to have become a feature of a number of the
newer agents (e.g. trovafloxacin, grepafloxacin, moxifloxacin
and garenoxacin), although many fluoroquinolones and
naphthyridones are eliminated primarily by metabolism and
renal clearance (glomerular filtration and active tubular
secretion). Clearly, a high free-drug
C
max
and AUCs, com-
bined with lower MIC, are a significant advance because they
will have the effect of increasing the value of
C
max
/MIC or
AUC/MIC ratios.
There is always a certain amount of debate as to whether the
C
max
/MIC or the AUC/MIC is more important in terms of
fluoroquinolone bacteriological and clinical outcomes in
man. This is probably not important because the co-variable
nature of these two pharmacokinetic parameters makes it dif-
ficult to identify which one is dominant in determining micro-
biological and clinical outcome. In general, agents with large
AUC also have high
C
max
concentrations. The other area of
considerable pharmacodynamic debate has been what break-
point or critical PK/PD ratio is required to optimize micro-
biological or clinical outcomes. For example, if there is a
range of values, the bigger numbers are more likely to be pre-
dictive of good clinical outcome in terms of the microbiology
and the emergence of resistance.
45
At present it is reasonable
to assume that almost all the drugs that have AUC/MIC ratios
>100 are likely to have useful activity against Enterobac-
teriaceae and
P. aeruginosa
, while a target of free AUC/MIC
of 3040 is required for
S. pneumoniae.
46
For predicting efficacy against most infections, pharmaco-
dynamic variables (for example, AUC/MIC or
C
max
/MIC) are
driven by changes in potency, much more than they are driven
by changes in pharmacokinetics. The MIC changes are some-
times 100-fold between agents, whereas the pharmacokinetic
changes are probably at most five-fold between any of these
agents. For example, as ciprofloxacin has an AUC/MIC
between 25 and 50 for the therapy of
P. aeruginosa
, it can
be anticipated that levofloxacin, trovafloxacin and clina-
floxacin will have similar activity to ciprofloxacin against
P. aeruginosa
(Table 6).
47,48
In contrast, some of the newer
drugs, such as grepafloxacin, moxifloxacin, gatifloxacin,
sparfloxacin, gemifloxacin or garenoxacin, together with
some of the older urinary drugs like norfloxacin, are unlikely
to have significant clinical activity against
P. aeruginosa
at
present doses. Pharmacodynamics would predict that all but
the very early quinolones will have good clinical activity
T
a
bl
e
6
.
Deve
l
opment o
f
qu
i
no
l
one p
h
armaco
d
ynam
i
cs aga
i
nst
G
ram-negat
i
ve pat
h
ogens
AUC/MIC (total)
E. coli H. influenzae P. aeruginosa
<12.5 nalidixic acid nalidixic acid nalidixic acid, enoxacin, norfloxacin, sparfloxacin,
grepafloxacin, moxifloxacin, gatifloxacin, gemifloxacin, garenoxacin
12.525 enoxacin, ofloxacin
2550 ciprofloxacin, levofloxacin, trovafloxacin, clinafloxacin
50100 norfloxacin
>100 all others all others temafloxacin
Table 7.
Development of quinolone pharmacodynamics against Gram-positive pathogens
AUC/MIC
(total)
S. aureus S. pneumoniae
Group A streptococci
<12.5 nalidixic acid, cinoxacin,
norfloxacin
nalidixic acid, cinoxacin, enoxacin,
norfloxacin
nalidixic acid, cinoxacin,
enoxacin,
norfloxacin
12.525 enoxacin ciprofloxacin sparfloxacin, grepafloxacin
2550 ciprofloxacin ofloxacin, levofloxacin, grepafloxacin ciprofloxacin, ofloxacin,
levofloxacin
50100 sparfloxacin
>100 ofloxacin, levofloxacin, temafloxacin,
trovafloxacin, clinafloxacin,
grepafloxacin, moxifloxacin,
gatifloxacin, gemifloxacin,
garenoxacin
temafloxacin, trovafloxacin,
clinafloxacin, moxifloxacin,
gatifloxacin, gemifloxacin,
garenoxacin
temafloxacin, trovafloxacin,
clinafloxacin, moxifloxacin,
gatifloxacin, gemifloxacin,
garenoxacin
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
Development of the quinolones
7
against
E. coli
and
H. influenzae
causing systemic infection
(Table 6).
With respect to the Gram-positive organisms, it is clear that
the urinary drugs are not going to have useful clinical activity
against
S. pneumoniae
,
S. aureus
or
Streptococcus pyogenes
(Table 7). Whereas with ofloxacin, levofloxacin, grepa-
floxacin and sparfloxacin, AUC/MIC ratios of >25 are begin-
ning to predict clinically useful outcomes for non-immuno-
compromised patients with mild to moderate community-
acquired disease. In the context of immunocompromised
patients (those with co-morbidity or ITU patients) or in order
to prevent the emergence of resistance, ratios should probably
be higher. The more recently developed fluoroquinolones,
such as trovafloxacin, gatifloxacin, gemifloxacin, moxi-
floxacin and garenoxacin, are likely to have useful clinical
activity against
S. pneumoniae
, a prediction now supported by
clinical trials data in community-acquired pneumonia
49
and
acute exacerbation of chronic bronchitis (AECB).
50,51
The
small amount of existing clinical data also implies that these
agents are clinically active against ciprofloxacin-susceptible
S. aureus
and Group A streptococci.
3235,52
Toxicology
Many of the more recently developed quinolones do not have
the toxicological disadvantages of the earlier compounds, for
example the QTc prolongation that has limited the use of spar-
floxacin and grepafloxacin. Other effects include a haemo-
lytic uraemic-like syndrome with temafloxacin,
17
a metallic
taste with grepafloxacin,
53
hepatitis with trovafloxacin,
54
unexpected hypoglycaemia with clinafloxacin and tema-
floxacin,
21,55,56
a number of immunologically mediated
adverse events with tosufloxacin,
18
and an immune-mediated
rash in young women with gemifloxacin.
6
To date there are
little toxicological data on garenoxacin.
Indications and use
The pharmacokinetic and
in vitro
potency profiles of fluoro-
quinolones determine the areas of clinical use. This has
shifted from agents used predominantly for the treatment of
urinary tract infections in the 1960s/70s, to more systemic use
in the 1980s/90s, and in the current millennium, to the treat-
ment of respiratory tract infections. Sparfloxacin and grepa-
Table 8.
Proposed classification of fluoroquinolones
Agents Activity Clinical applications
Urinary agents (19601985)
nalidixic acid
cinoxacin
enoxacin
norfloxacin
activity against common
Enterobacteriaceae, short serum
half-lives, renal elimination
main use in UTI
Gram-negative systemic agents (19851995)
ciprofloxacin
ofloxacin
levofloxacin
wide activity against Gram-negatives,
including
P. aeruginosa
, marginal
activity against Gram-positives,
longer serum half-lives
widely used against
tissue-based and urinary
infections
Broad spectrum systemic agents (19902000)
temafloxacin
clinafloxacin
trovafloxacin
wide activity against Gram-negatives,
including
P. aeruginosa
for some
agents, and Gram-positives, for some
agents long serum half-lives, some
activity against anaerobes
widely used against a
broad range of tissue-
based infections
Respiratory agents (1995 onwards)
levofloxacin
sparfloxacin
grepafloxacin
moxifloxacin
gatifloxacin
gemifloxacin
garenoxacin
wide activity against
Enterobacteriaceae, active against
Gram-positives, especially
S.
pneumoniae
, active against atypical
bacteria, variable activity against
anaerobes, long serum half-life
main use in respiratory
tract infection
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
M. I. Andersson and A. P. MacGowan
8
floxacin were the first agents to focus on the respiratory tract,
exploiting their activity against
S. pneumoniae
and atypical
organisms. More recently a further group of agents (moxi-
floxacin, gatifloxacin, gemifloxacin and garenoxacin) with
more convincing potential in the respiratory tract have been
developed. Thus development has moved from the urinary to
the respiratory tract via a miscellaneous group of systemic
indications. Levofloxacin is one example of a drug where
there is a wide range of potential uses, but in terms of market-
ing the main focus appears to be respiratory.
On this basis, quinolones can be classified into four groups
according to clinical use, potency, pharmacokinetics and
pharmacodynamics. The first group consists of urinary agents
that are, in general, the older drugs which are active against
the common Enterobacteriaceae and tend to have short half-
lives and renal elimination (Table 8). The second group
includes the anti-Gram-negative systemic agents such as
ciprofloxacin, levofloxacin and ofloxacin which, according
to their pharmacokinetics and activity, indicate that they can
be used for Gram-negative infection and as anti-pseudomonal
agents. There is, however, debate about their use in various
areas of Gram-positive infection. The third group, of which
trovafloxacin and temafloxacin are the best examples, have
sufficiently broad activity in terms of anti-Gram-negative,
Figure 5.
Development of quinolones.
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
Development of the quinolones
9
anti-Gram-positive, antipseudomonal and anti-anaerobic
activity, to indicate their use for a wide range of tissue-based
infections. The fourth group includes the respiratory agents,
some of which are also in other categories. These drugs have
activity against
S. pneumoniae
and atypical organisms, but
are less active against
P. aeruginosa.
Some of these agents,
such as moxifloxacin and garenoxacin, may be as active as
trovafloxacin against anaerobes, suggesting that with time,
their clinical indications may expand beyond the respiratory
area. The nomenclature of the quinolones is complex, and
these agents have also been classified in terms of generation
57
(discussed in this supplement in the article by P. Ball). How-
ever, whichever classification is used, it is clear that these
agents are not a homogeneous group of antibiotics, and that
important differences exist between them.
Conclusions
Modifications in the basic structure of quinolones have
increased their antibacterial spectrum and potency, as well as
improving bioavailability, making quinolones useful agents
for the treatment of urinary, systemic and respiratory tract
infections. An evolutionary tree is shown in Figure 5. How-
ever, safety concerns continue with some members of this
class and have resulted in the withdrawal of some agents after
marketing (temafloxacin, grepafloxacin, sparfloxacin, trova-
floxacin) or others in late development (clinafloxacin and
gemifloxacin). It is still unclear as to which structure
function relationships have resulted in these problems, so
despite a good deal of progress being made in terms of
in vitro
activity and pharmacodynamics, progress in the area of toxi-
cology has been erratic.
References
1.
Tillotson, G. S. & Blondeau, J. M. (1999). Structure–activity–
function evaluation of the fluoroquinolones. In
Moxifloxacin in Prac-
tice
(Adam, D., Finch, R. G. & Hunter, P. A., Eds), pp. 91101.
Maxim Medical, Oxford, UK.
2.
Domagala, J. M. (1994). Structureactivity and structureside-
effect relationships for the quinolone antibacterials.
Journal of Anti-
microbial Chemotherapy
33
, 685706.
3.
Asahina, Y., Ishizaki, T. & Suzue, S. (1992). Recent advances
in structure activity relationships in new quinolones.
Progress in
Drug Research
38
, 57106.
4.
Tillotson, G. S. (1996) Quinolones: structureactivity relation-
ships and future predictions.
Journal of Medical Microbiology
44
,
3204.
5.
Ball, P., Fernald, A. & Tillotson, G. (1998). Therapeutic
advances of new fluoroquinolones.
Expert Opinion in Investigational
Drugs
7
, 76183.
6.
Lowe, M. N. & Lamb, H. M. (2000). Gemifloxacin.
Drugs
59
,
113747.
7.
Ball, P. (2000). Moxifloxacin (Avelox): an 8-methoxyquinolone
antibacterial with enhanced potency.
International Journal of Clin-
ical Practice
54
, 32932.
8.
Brighty, K. E. & Gootz, T. D. (1997). The chemistry and biologi-
cal profile of trovafloxacin.
Journal of Antimicrobial Chemotherapy
39
,
Suppl. B
, 114.
9.
Dong, Y., Xu, C., Zhao, X., Domagala, J. & Drlica, K. (1998).
Fluoroquinolone action against mycobacteria: effects of C-8 substit-
uents on growth, survival, and resistance.
Antimicrobial Agents and
Chemotherapy
42
, 297884.
10.
Zhao, X., Wang, J. Y., Xu, C., Dong, Y., Zhou, J., Domagala, J.
et al
. (1998). Killing of
Staphylococcus aureus
by C-8-methoxy
fluoroquinolones.
Antimicrobial Agents and Chemotherapy
42
,
9568.
11.
Lu, T., Zhao, X. & Drlica, K. (1999) Gatifloxacin activity against
quinolone-resistant gyrase: allele-specific enhancement of bacterio-
static and bactericidal activities by the C-8-methoxy group.
Anti-
microbial Agents and Chemotherapy
43
, 296974.
12.
Cecchetti, V., Fravolini, A., Lorenzini, M. C., Tabarrini, O., Terni,
P. & Xin, T. (1996). Studies on 6-aminoquinolones: synthesis and
antibacterial evaluation of 6-amino-8-methylquinolones.
Journal of
Medicinal Chemistry
39
, 43645.
13.
Ma, Z., Chu, D. T., Cooper, C. S., Li, Q., Fung, A. K., Wang, S.
et al
. (1999). Synthesis and antimicrobial activity of 4H-4-oxoquino-
lizine derivatives: consequences of structural modification at the
C-8 position.
Journal of Medicinal Chemistry
42
, 420213.
14.
Gootz, T. D. & Brighty, K. E. (1996). Fluoroquinolone anti-
bacterials: SAR mechanism of action, resistance, and clinical
aspects.
Medicinal Research Reviews
16
, 43386.
15.
Lipsky, B. A. & Baker, C. A. (1999). Fluoroquinolone toxicity
profiles: a review focusing on newer agents.
Clinical Infectious
Diseases
28
, 35264.
16.
Jaillon, P., Morganroth, J., Brumpt, I. & Talbot, G. (1996). Over-
view of electrocardiographic and cardiovascular safety data for
sparfloxacin. Sparfloxacin Safety Group.
Journal of Antimicrobial
Chemotherapy
37
,
Suppl. A
, 1617.
17.
Blum, M. D., Graham, D. J. & McCloskey, C. A. (1994). Tema-
floxacin syndrome: review of 95 cases.
Clinical Infectious Diseases
18
, 94650.
18.
Ball, P., Mandell, L., Niki, Y. & Tillotson, G. (1999). Comparative
tolerability of the newer fluoroquinolone antibacterials.
Drug Safety
21
, 40721.
19.
Yamamoto, T., Tanida, T., Ueta, E., Kimura, T., Doi, S. & Osaki,
T. (2001). Pulmonary infiltration with eosinophilia (PIE) syndrome
induced by antibiotics, PIPC and TFLX during cancer treatment.
Oral Oncology
37
, 4715.
20.
Zhanel, G. G., Ennis, K., Vercaigne, L., Walkty, A., Gin, A. S.,
Embil, J.
et al
. (2002). A critical review of the fluoroquinolones:
focus on respiratory infections.
Drugs
62
, 1359.
21.
Rubinstein, E. (2001). History of quinolones and their side
effects.
Chemotherapy
47
,
Suppl. 3
, 38.
22.
Dalhoff, A. (1999).
In vitro
activities of quinolones.
Expert Opin-
ion on Investigational Drugs
8
, 12337.
23.
Reeves, D. S., Bywater, M. J. & Holt, H. A. (1984). The activity
of enoxacin against clinical bacterial isolates in comparison with that
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
M. I. Andersson and A. P. MacGowan
10
of five other agents, and factors affecting that activity.
Journal of
Antimicrobial Chemotherapy
14
,
Suppl. C
, 717.
24.
Newsom, S. W. (1984). The antimicrobial spectrum of nor-
floxacin.
Journal of Antimicrobial Chemotherapy
13
,
Suppl. B
,
2531.
25.
Gruneberg, R. N., Felmingham, D., OHare, M. D., Robbins, M.
J., Perry, K., Wall, R. A.
et al
. (1988). The comparative in-vitro activ-
ity of ofloxacin.
Journal of Antimicrobial Chemotherapy
22
,
Suppl.
C
, 919.
26.
King, A. & Phillips, I. (1986). The comparative in-vitro activity of
eight newer quinolones and nalidixic acid.
Journal of Antimicrobial
Chemotherapy
18
,
Suppl. D
, 120.
27.
Wiedemann, B. & Heisig, P. (1997). Antibacterial activity of
grepafloxacin.
Journal of Antimicrobial Chemotherapy
40
,
Suppl. A
,
1925.
28.
Cunha, B. A., Qadri, S. M., Ueno, Y., Walters, E. A. &
Domenico, P. (1997). Antibacterial activity of trovafloxacin against
nosocomial Gram-positive and Gram-negative isolates.
Journal of
Antimicrobial Chemotherapy
39
,
Suppl. B
, 2934.
29.
Wise, R., Ashby, J. P. & Andrews, J. M. (1988).
In vitro
activity
of PD 127,391, an enhanced-spectrum quinolone.
Antimicrobial
Agents and Chemotherapy
32
, 12516.
30.
MacGowan, A. P. (1999). Moxifloxacin (Bay 12-8039): a new
methoxy quinolone antibacterial.
Expert Opinion on Investigational
Drugs
8
, 18191.
31.
Perry, C. M., Barman Balfour, J. A. & Lamb, H. M. (1999). Gati-
floxacin.
Drugs
58
, 68396.
32.
Berrington, A. W., Koerner, R. J., Perry, J. D., Bain, H. H. &
Gould, F. K. (2001). Treatment of
Staphylococcus aureus
endocar-
ditis using moxifloxacin.
International Journal of Medical Micro-
biology
291
, 2379.
33.
Muijsers, R. B. & Jarvis, B. (2002). Moxifloxacin in uncompli-
cated skin and skin structure infections.
Drugs
62
, 96773.
34.
Parish, L. C., Routh, H. B., Miskin, B., Fidelholtz, J., Werschler,
P., Heyd, A.
et al
. (2000). Moxifloxacin versus cephalexin in the
treatment of uncomplicated skin infections.
International Journal of
Clinical Practice
54
, 497503.
35.
Entenza, J. M., Que, Y. A., Vouillamoz, J., Glauser, M. P. &
Moreillon, P. (2001). Efficacies of moxifloxacin, ciprofloxacin, and
vancomycin against experimental endocarditis due to methicillin-
resistant
Staphylococcus aureus
expressing various degrees of
ciprofloxacin resistance.
Antimicrobial Agents and Chemotherapy
45
, 307683.
36.
Bille, J. & Glauser, M. P. (1991). In-vitro activity of temafloxacin
for Gram-positive pathogens.
Journal of Antimicrobial Chemo-
therapy
28
,
Suppl. C
, 914.
37.
Ridgway, G. L., Salman, H., Robbins, M. J., Dencer, C. & Felm-
ingham, D. (1997). The in-vitro activity of grepafloxacin against
Chlamydia
spp.,
Mycoplasma
spp.,
Ureaplasma
urealyticum
and
Legionella
spp.
Journal of Antimicrobial Chemotherapy
40
,
Suppl.
A
, 314.
38.
Sefton, A. M., Maskell, J. P., Rafay, A. M., Whiley, A. &
Williams, J. D. (1997). The in-vitro activity of trovafloxacin, a new
fluoroquinolone, against Gram-positive bacteria.
Journal of Anti-
microbial Chemotherapy
39
,
Suppl. B
, 5762.
39.
Blondeau, J. M. (1999). A review of the comparative in-vitro
activities of 12 antimicrobial agents, with a focus on five new
respiratory quinolones.
Journal of Antimicrobial Chemotherapy
43
,
Suppl. B
, 111.
40.
Wolf, R., Eberl, R., Dunky, A., Mertz, N., Chang, T., Goulet, J.
R.
et al
. (1984). The clinical pharmacokinetics and tolerance of
enoxacin in healthy volunteers.
Journal of Antimicrobial Chemo-
therapy
14
,
Suppl. C
, 639.
41.
Stass, H. & Kubitza, D. (1999). Pharmacokinetics and elimin-
ation of moxifloxacin after oral and intravenous administration in
man.
Journal of Antimicrobial Chemotherapy
43
,
Suppl. B
, 8390.
42.
Hooper, D. (2000). Quinolones. In
Mandell
,
Douglas
,
and
Bennetts Principles and Practice of Infectious Diseases
, 5th edn
(Mandell, G. L., Bennett J. E. & Dolin, R., Eds), pp. 40423. Church-
ill Livingstone, Philadelphia, USA.
43.
Ball, P. (2000). The quinolones: history and overview. In
The
Quinolones
, 3rd edn (Andriole, V. T., Ed.), pp. 131. Academic
Press, London, UK.
44.
Wise, R., Lister, D., McNulty, C. A., Griggs, D. & Andrews, J. M.
(1986). The comparative pharmacokinetics of five quinolones.
Journal of Antimicrobial Chemotherapy
18
,
Suppl. D
, 7181.
45.
MacGowan, A. P. (1999).
In vitro
models of infection:
pharmacokinetic/pharmacodynamic correlates. In
Proceedings of
the First International Moxifloxacin Symposium
(Mandell, L., Ed.),
pp. 10410. Springer-Verlag, Berlin, Germany.
46.
Ambrose, P. G., Grasela, D. M., Grasela, T. H., Passarell, J.,
Mayer, H. B. & Pierce, P. F. (2001). Pharmacodynamics of fluoro-
quinolones against
Streptococcus pneumoniae
in patients with
community-acquired respiratory tract infections.
Antimicrobial
Agents and Chemotherapy
45
, 27937.
47.
Mouton, J. W., van Ogtrop, M. L., Andes, D. & Craig, W. A.
(1999). Use of pharmacodynamic indices to predict efficacy of
combination therapy
in vivo
.
Antimicrobial Agents and Chemo-
therapy
43
, 24738.
48.
MacGowan, A. P., Wootton, M. & Holt, H. A. (1999). The anti-
bacterial efficacy of levofloxacin and ciprofloxacin against
Pseudo-
monas aeruginosa
assessed by combining antibiotic exposure and
bacterial susceptibility.
Journal of Antimicrobial Chemotherapy
43
,
3459.
49.
Patel, T., Pearl, J., Williams, J., Haverstock, D. & Church, D.
(2000). Efficacy and safety of ten day moxifloxacin 400 mg once
daily in the treatment of patients with community-acquired pneu-
monia. Community Acquired Pneumonia Study Group.
Respiratory
Medicine
94
, 97105.
50.
Wilson, R., Jones, P., Schaberg, T., Davey, P., Allegra, L.,
Izquierdo, J. L.
et al
. (2002). A clinical and outcomes assessment of
the management of Acute Exacerbations of Chronic Bronchitis
(AECB): The MOSAIC Study. In
Programs and Abstracts of the
Twelfth European Respiratory Society Congress
, Stockholm, 2002.
Abstract 3371. European Respiratory Society.
51.
Wilson, R., Schentag, J. J., Ball, P. & Mandell, L. (2002). A
comparison of gemifloxacin and clarithromycin in acute exacerba-
tions of chronic bronchitis and long-term clinical outcomes.
Clinical
Therapeutics
24
, 63952.
52.
Worlitzsch, D., Kaygin, H., Steinhuber, A., Dalhoff, A., Botzen-
hart, K. & Doring, G. (2001). Effects of amoxicillin, gentamicin, and
moxifloxacin on the hemolytic activity of
Staphylococcus aureus
in
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
Development of the quinolones
11
vitro
and
in vivo
.
Antimicrobial Agents and Chemotherapy
45
,
196202.
53.
Stahlmann, R. & Schwabe, R. (1997). Safety profile of grepa-
floxacin compared with other fluoroquinolones.
Journal of Anti-
microbial Chemotherapy
40
,
Suppl. A
, 8392.
54.
Chen, H. J., Bloch, K. J. & Maclean, J. A. (2000). Acute
eosinophilic hepatitis from trovafloxacin.
New England Journal of
Medicine
342
, 35960.
55.
Stahlmann, R. (2002). Clinical toxicological aspects of fluoro-
quinolones.
Toxicology Letters
127
, 26977.
56.
Stratton, C. W. (1998). Pharmacokinetics, pharmacodynamics
and tolerability profile of clinafloxacin.
Clinical Drug Investigation
15
,
Suppl. 1
, 1934.
57.
Ball, P. (2000). Quinolone generations: natural history or natu-
ral selection?
Journal of Antimicrobial Chemotherapy
46
,
Suppl. T1
,
1724.
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
by guest on July 13, 2011jac.oxfordjournals.orgDownloaded from
... In the 1970s-1980s, the quinolone class expanded significantly with the development of fluoroquinolones (FQs), which demonstrate a much broader spectrum of activity and improved pharmacokinetics and pharmacodynamics (PK/PD) compared to the firstgeneration quinolones [1,2]. For over five decades, FQs have been favored as antibiotics due of their high potency, broad-spectrum activity, favorable bioavailability, convenient formulations, high serum concentrations, high tissue penetrations and comparatively low incidence of side effects [1,3]. ...
... On the contrary, microorganisms of the normal oral flora, which generally start the formation of biofilm, are rarely involved in VAP development [139,146]. It is also funda-mental to the timing of VAP development; some studies show that, if it develops a few days (2)(3)(4)(5) after intubation, it is likely caused by antibiotic-sensitive bacteria such as MSSA; on the other hand, if it is developed after five days, it frequently involves multidrug-resistant microorganism like MRSA, P. aeruginosa difficult-to-treat (DTR) and carbapenem-resistant Enterobacterales (CRE) [143,147]. There are many strategies to prevent VAP, but these are not the focus of this review [148,149]. ...
... Their PK/PD characteristics allow their use in different human districts, even the ones hard to normally reach, such as the prostate or the CNS. Their versatility in treating Gram-positive and Gram-negative pathogens makes them the treatment of choice for various infections [1,3]. Furthermore, they are made in different formulations to enhance their use. ...
Article
Full-text available
Background: Biofilm-associated infections frequently span multiple body sites and represent a significant clinical challenge, often requiring a multidisciplinary approach involving surgery and antimicrobial therapy. These infections are commonly healthcare-associated and frequently related to internal or external medical devices. The formation of biofilms complicates treatment, as they create environments that are difficult for most antimicrobial agents to penetrate. Fluoro-quinolones play a critical role in the eradication of biofilm-related infections. Numerous studies have investigated the synergistic potential of combining fluoroquinolones with other chemical agents to augment their efficacy while minimizing potential toxicity. Comparative research suggests that the antibiofilm activity of fluoroquinolones is superior to that of beta-lactams and glycopeptides. However, their activity remains less effective than that of minocycline and fosfomycin. Noteworthy combinations include fluoroquinolones with fosfomycin and aminoglycosides for enhanced activity against Gram-negative organisms and fluoroquinolones with minocycline and rifampin for more effective treatment of Gram-positive infections. Despite the limitations of fluoroquinolones due to the intrinsic characteristics of this antibiotic, they remain fundamental in this setting thanks to their bioavailability and synergisms with other drugs. Methods: A comprehensive literature search was conducted using online databases (PubMed/MEDLINE/Google Scholar) and books written by experts in microbiology and infectious diseases to identify relevant studies on fluoroquinolones and biofilm. Results: This review critically assesses the role of fluoroquinolones in managing biofilm-associated infections in various clinical settings while also exploring the potential benefits of combination therapy with these antibiotics. Conclusions: The literature predominantly consists of in vitro studies, with limited in vivo investigations. Although real world data are scarce, they are in accordance with fluoroquinolones' effectiveness in managing early biofilm-associated infections. Also, future perspectives of newer treatment options to be placed alongside fluoroquinolones are discussed. This review underscores the role of fluoroquinolones in the setting of biofilm-associated infections, providing a comprehensive guide for physicians regarding the best use of this class of antibiotics while highlighting the existing critical issues.
... This is because they offer many of the attributes of an ideal antibacterial, combining high potency, broad-spectrum activity, good bioavailability, oral and intravenous formulations, high serum levels, and large distribution volume, indicating good concentrations in different tissues. 12,13 They play a key role in the treatment of urinary tract infections (UTIs), upper and lower respiratory tract infections (RTI), and sexually transmitted diseases (STDs). Besides their effects in curing skin, so tissue, gastrointestinal, bones, and joint infections, 14,15 they exert their bactericidal activity by targeting bacterial DNA gyrase and topoisomerase IV (Topo IV), where they bind to complexes that are formed between DNA and DNA gyrase or Topo IV, forming a ternary complex of DNA enzymequinolone. ...
Article
Full-text available
The antibacterial efficacy of some newly developed bis- and C3-carboxylic moieties of fluoroquinolone-linked triazole conjugates was studied. Twenty compounds from two different series of triazoles were synthesized using click chemistry and evaluated for their antibacterial activity against a Gram-positive strain, i.e. Enterococcus faecalis (ATCC29212), and its clinical isolate and a Gram-negative bacterial strain, i.e. Escherichia coli (ATCC25922), and its clinical isolate. Among the compounds, 7, 9a, 9d, 9i, 10(a–d), and 10i showed excellent activity with MIC values of up to 6.25 μg mL⁻¹, whereas the control ciprofloxacin showed MIC values of up to 12.5 μg mL⁻¹ towards the various strains. Cytotoxicity was evaluated against Vero cells (kidney epithelial cells of an African green monkey), and results revealed that compounds 9a, 9c, 10g, 10h, and 10 are toxic. Molecular docking and MD analysis were performed using the protein structure of E. coli DNA gyrase B and further corroborated with an in vitro assay to evaluate the inhibition of DNA gyrase. The analysis revealed that compound 10d was a more potent inhibitor of DNA gyrase compared to ciprofloxacin, which was employed as the positive control.
... Discovered in the 1960s, quinolones have since become important therapies in the treatment of community-acquired and severe nosocomial infections [64]. Quinolones pose several desirable characteristics, such as a broad spectrum of activity, limited incidence of severe side effects, high potency, and good bioavailability [64][65][66], and, as such, have been widely prescribed for a variety of infections [64,66]. ...
Article
Full-text available
Antimicrobial resistance (AMR) in Campylobacter species, particularly C. jejuni and C. coli, poses a significant public health threat. These bacteria, which are commonly found in livestock, poultry, companion animals, and wildlife, are the leading causes of foodborne illnesses, often transmitted through contaminated poultry. Extensive exposure to antibiotics in human and veterinary medicine creates selection pressure, driving resistance through mechanisms such as point mutations, horizontal gene transfer, and efflux pumps. Resistance to fluoroquinolones, macrolides, and tetracyclines complicates treatment and increases the risk of severe infections. Drug-resistant Campylobacter is transmitted to humans via contaminated food, water, and direct contact with animals, highlighting its zoonotic potential. Addressing this challenge requires effective interventions. Pre-harvest strategies like biosecurity and immune-based methods reduce bacterial loads on farms, while post-harvest measures, including carcass decontamination and freezing, limit contamination. Emerging approaches, such as bacteriocins and natural antimicrobials, offer chemical-free alternatives. Integrated, multidisciplinary interventions across the food chain are essential to mitigate AMR transmission and enhance food safety. Sustainable agricultural practices, antimicrobial stewardship, and innovative solutions are critical to curbing Campylobacter resistance and protecting global public health. Our review examines the dynamics of antimicrobial resistance in Campylobacter and presents current strategies to mitigate Campylobacter-related AMR, offering valuable insights for antimicrobial control in the poultry industry.
... Fluoroquinolones like ciprofloxacin and ofloxacin are the first choice and highly recommended antibiotics due to highly potency, highly bioavailability, available formulations, highly serum concentrations and mild side effects. Fluoroquinolones inhibit both Gram-negative and Gram-positive bacteria, so they are widely used in the treatment of tuberculosis, anthraxes and other serious bacterial diseases [6]. Rarely, fluoroquinolones affect gastrointestinal tract, nervous system, genome, and eye [7]. ...
Article
Full-text available
This work is an attempt to overcome antimicrobial resistance problem which dispersed worldwide in particular developing countries due to misuse of antibiotics. Actinobacteria were isolated and screened against selected resistant Gram-negative bacteria to detect the powerful antibacterial activity. Identification of the most potent actinobacterial isolate has been carried out using classical and genetical methods. Antibacterial compound has been extracted, purified and characterized using accurate and more specific techniques and instruments. Among forty actinobacterial isolates, only twenty-two isolates could inhibit the growth of Gram-negative bacteria. The most potent isolate Eg-7 was identified as S. griseorubens, which has a typical 16S rRNA gene. The antibacterial compound was extracted using ethyl acetate, and separated by High Performance Liquid Chromatography using methanol and water as a mobile phase. Five active peaks were displayed and retained in the range 40 – 45 min, but the last three peaks were retained at 41.90, 43.43, and 44.54 min, respectively. The crude extract was analyzed by liquid chromatography mass spectrum, where the active peak was displayed at 721.325 m/z. The antibacterial compound was purified using flash column chromatography and gel filtration column chromatography. The active fraction was analyzed by Infra-Red spectrum, where a broad absorption at 3338 cm-1 was displayed. Molecular formula of an antibacterial compound was determined by mass spectrum as C35H26N6O4. Nuclear magnetic resonance analysis was carried out for an antibacterial compound. These results suggest that a new antibacterial compound that similar quinolone could be produced by S. griseorubens and exhibited a higher activity against Gram-negative bacteria.
... unsanitary situations and are provided with a diet that is deficient or unbalanced in vitamins and amino acids. This unequivocally highlights the crucial role of plant antioxidants in alleviating the detrimental effects of such circumstances on avian health and maximizing nutrient utilization (Andersson and Macgowan, 2003;Luangtongkum et al., 2006;Martinez et al., 2006;Billah et al., 2015). Feed and its manufacturing process constitute a substantial proportion of the overall expenses in broilers production. ...
Article
Full-text available
Background: The poultry industry continues to grow due to the availability, affordability, and nutritional value of eggs and meat. Factors influencing this growth include genetics, feed quality, disease prevention, and the use of antibiotics. However, the search for alternatives to antibiotics has gained momentum due to concerns about antibiotic resistance and consumer demand. This study aimed to evaluate the effectiveness of adding various doses of the commercial compound X-VIRUS solution (containing antioxidants and essential amino acids) to drinking water, with the goal of determining the optimal dose for improving broiler productivity. Methods: A total of 360 unsexed Ross 308 broilers were assigned to four treatments. Each treatment group was divided into six pens, with 15 chicks per pen. Treatments included: T1) control group (did not receive any additives), T2) 0.5 ml of X-VIRUS per liter of water, T3) 1 ml of X-VIRUS per liter of water, and T4) 1.5 ml of X-VIRUS per liter of water. Performance, carcass traits, biochemical blood variables and antioxidant capacity of broilers were assessed. Results: Administering X-VIRUS solution at a dose of 1.5 ml/l in broilers' drinking water increased body weight and weight gain compared to the lower doses. Additionally, higher X-VIRUS doses improved feed intake and feed conversion ratios. X-VIRUS solution increased significantly the relative weight of carcass, breast, and thigh in the fifth week. Higher X-VIRUS doses (1 and 1.5ml/l) led to higher antibody titers compared to lower doses. X-VIRUS solution reduced triglyceride, cholesterol, LDL, glucose, ALT and MDA levels and increased HDL, total protein levels and GSH-Px activity, while albumin and globulin levels remained unchanged. Conclusions: administering a dose of 1.5ml/l of X-virus solution showed the best results in the performance, carcass characteristics, blood parameters and antioxidant capacity of broilers.
... Campylobacter infections are generally self-limiting; however, antimicrobial therapies are required for severe infection cases, especially for the elderly and individuals with compromised immune systems (Balaban et al., 2004;Cañas-Duarte et al., 2014;Sulaiman and Lam, 2020). However, C. jejuni is increasingly resistant to clinically important antibiotics, particularly fluoroquinolones (FQs), the most commonly prescribed class of antibiotics for oral treatment of various bacterial infections, including gastroenteritis (Andersson and MacGowan, 2003;Schierenberg et al., 2019). ...
Article
Full-text available
Antibiotic tolerance enables antibiotic-susceptible bacteria to withstand prolonged exposure to high concentrations of antibiotics. Although antibiotic tolerance presents a major challenge for public health, its underlying molecular mechanisms remain unclear. Previously, we have demonstrated that Campylobacter jejuni develops tolerance to clinically important antibiotics, including ciprofloxacin and tetracycline. To identify cellular responses associated with antibiotic tolerance, RNA-sequencing was conducted on C. jejuni after inducing antibiotic tolerance through exposure to ciprofloxacin or tetracycline. Additionally, knockout mutants were constructed for genes exhibiting significant changes in expression levels during antibiotic tolerance. The genes involved in protein chaperones, bacterial motility, DNA repair system, drug efflux pump, and iron homeostasis were significantly upregulated during antibiotic tolerance. These mutants displayed markedly reduced viability compared to the wild-type strain, indicating the critical role of these cellular responses in sustaining antibiotic tolerance. Notably, the protein chaperone mutants exhibited increased protein aggregation under antibiotic treatment, suggesting that protein chaperones play a critical role in managing protein disaggregation and facilitating survival during antibiotic tolerance. Our findings demonstrate that various cellular defense mechanisms collectively contribute to sustaining antibiotic tolerance in C. jejuni, providing novel insights into the molecular mechanisms underlying antibiotic tolerance.
... In addition to the advantages related to the antibacterial activity, FQNs also present a benefit from a chemical point of view. Their structures are relatively easy to synthesize, thus allowing the development of numerous potential derivatives with various advantageous features [14,37,40,41]. Their chemical structure shows excellent complexation properties with metal ions and the ability to form combinations with other active molecules [42,43]. ...
Article
Full-text available
Background/Objectives: Despite the discovery of antibiotics, bacterial infections persist globally, exacerbated by rising antimicrobial resistance that results in millions of cases, increased healthcare costs, and more extended hospital stays. The urgent need for new antibacterial drugs continues as resistance evolves. Fluoroquinolones and tetracyclines are versatile antibiotics that are effective against various bacterial infections. A hybrid antibiotic combines two or more molecules to enhance antimicrobial effectiveness and combat resistance better than monotherapy. Fluoroquinolones are ideal candidates for hybridization due to their potent bactericidal effects, ease of synthesis, and ability to form combinations with other molecules. Methods: This study explored the mechanisms of action for 40 hypothetical fluoroquinolone–tetracycline hybrids, all of which could be obtained using a simple, eco-friendly synthesis method. Their interaction with Escherichia coli DNA Gyrase and similarity to albicidin were evaluated using the FORECASTER platform. Results: Hybrids such as Do-Ba, Mi-Fi, and Te-Ba closely resembled albicidin in physicochemical properties and FITTED Scores, while Te-De surpassed it with a better score. Similar to fluoroquinolones, these hybrids likely inhibit DNA synthesis by binding to enzyme–DNA complexes. Conclusions: These hybrids could offer broad-spectrum activity and help mitigate bacterial resistance, though further in vitro and in vivo studies are needed to validate their potential.
Article
Full-text available
Enrofloxacin (EFX) is a third-generation synthetic fluoroquinolone with a broad spectrum of antibacterial activity but suffers from low water solubility, affecting its bioavailability. This study attempts to enhance the physicochemical and biological properties of enrofloxacin by converting it into multicomponent forms using crystal engineering concepts. Cocrystallization of enrofloxacin with isomeric pyridine-2,n-dicarboxylic acids (n = 3,4,5,6) resulted in four new crystalline salts (1:1): EFX·Py2,3DCA, EFX·Py2,4DCA, EFX·Py2,5DCA·H2O and EFX·Py2,6DCA·H2O; two of these are monohydrates. The protonation of the nitrogen atom of the piperazine moiety and the presence of crystallization water molecules were confirmed by single-crystal X-ray diffraction and Fourier transform infrared spectroscopy. Thermogravimetric analysis provided information on the thermal behaviour of multicomponent forms. The biological studies showed that the obtained salts are characterized by high antibacterial activity against Gram-positive and Gram-negative bacteria, and their haemolytic activity is low. The new salts demonstrate significantly greater solubility in water compared to the parent drug, along with enhanced antibacterial activity; hence, pyridinedicarboxylic acids appear to be efficient cocrystallizing agents for improving the efficacy of pharmaceutical ingredients.
Article
Full-text available
A comprehensive review of advances in the synthesis and biological applications of enoxacin (1, referred to as ENX)-based compounds is presented. ENX, a second-generation fluoroquinolone (FQ), is a prominent 1,8-naphthyridine containing compounds studied in medicinal chemistry. Quinolones, a class of synthetic antibiotics, are crucial building blocks for designing multi-biological libraries due to their inhibitory properties against DNA replication. Chemical modifications at positions 3 and 7 of the quinolone structure can transform antibacterial FQs into anticancer analogs. ENX and its derivatives have been examined for various therapeutic applications, including anticancer, antiviral, and potential treatment against COVID-19. Several synthetic methodologies have been devised for the efficient and versatile synthesis of ENX and its derivatives. This review emphasizes all-inclusive developments in the synthesis of ENX derivatives, focusing on modifications at C3 (carboxylic acid, Part A), C7 (piperazinyl, Part B), and other modifications (Parts A and B). The reactions considered were chosen based on their reproducibility, ease of execution, accessibility, and the availability of the methodology reported in the literature. This review provides valuable insights into the medicinal properties of these compounds, highlighting their potential as therapeutic agents in various fields.
Article
Since its publication in 1979, Principles and Practice of Infectious Diseases (PPID) has become an essential element of every medical library. Infectious diseases clinicians have well-worn home or office copies to which they repeatedly turn. Other less complete infectious diseases texts have come and gone, but with each revision PPID remains the standard. The latest edition, a beautifully packaged two-volume opus of nearly 3000 pages, raises the measure of excellence to a new level.
Chapter
This chapter provides an overview of the quinolone antibacterials. Since the mid-1980s, the fluoroquinolones have become a major group of synthetic antibiotics with activities ranging from the Enterobacteriaceae and opportunists such as Pseudomonas aeruginosa to the Gram-positive pathogens including streptococci and staphylococci. In some areas such as pyelonephritis, gonorrhoea, enteric fevers, oral management of osteomyelitis, and P. aeruginosa infections, they have become drugs of choice, often replacing parenteral therapy with cephalosporins and aminoglycosides. However, bacterial resistance has appeared not only in predictable species such as staphylococci and P. aeruginosa, but also among organisms such as Escherichia coli, which was initially thought unlikely to be thus compromised. The newer agents including the 8-methoxyquinolones moxifloxacin and gatifloxacin, together with gemifloxacin have greatly enhanced anti-pneumococcal potency while retaining potency similar to ciprofloxacin against the enterobacteria. However, quinolone resistance has also been reported, apart from implications in sick elderly patients. Quinolones also exhibit potency against anaerobes and like trovafloxacin may find a major place in intra-abdominal and pelvic infection as monotherapy replacements for cumbersome combinations. The moratorium imposed on fluoroquinolone usage in children, relating to the potential risk of joint toxicity, has remained under review, and an expert committee has published guidelines for use in specified infections where the clinical need outweighs the theoretical risk. These specified infections include bronchopulmonary exacerbations of cystic fibrosis due to P. aeruginosa, typhoid, and paratyphoid fevers in areas of high resistance prevalence, single-dose treatment (and prophylaxis) of cholera, potentially invasive shigellosis and salmonellosis, and complicated urinary tract infection (UTI) caused by multiresistant bacteria.
Article
Community-acquired pneumonia (CAP) remains a common and serious illness with approximately 2–4 million cases reported annually. Management of CAP is therapeutically challenging due to the increasing prevalence of penicillin- and macrolide-resistant pneumococci and β -lactamase producing Haemophilus influenzae, as well as the increased recognition of ‘atypical’ pathogens, such as Chlamydia pneumoniae and Mycoplasma pneumoniae, and the frequent need for empiric therapy.We aimed to evaluate the safety and efficacy of moxifloxacin in the treatment of patients with CAP. To do this we carried out a prospective, uncontrolled, non-blind, Phase III clinical trial, in 27 U.S. centers. Patients included in the study were over 18 years of age with signs and symptoms of CAP confirmed by evidence of a new or progressive infiltrate on chest radiograph. The intervention used was moxifloxacin 400 mg PO once daily for 10 days.Sputum samples were collected pretherapy for Gram stain and culture for typical organisms. Culture and serological testing for Chlamydia pneumoniae and Mycoplasma pneumoniae was also performed. Susceptibility to moxifloxacin was determined by disk diffusion and MIC. Clinical and bacteriological responses were determined at the end of therapy (0–6 days post-therapy), follow-up (14–35 days post-therapy) and overall (end of therapy plus follow-up). Analyses were performed on both valid for efficacy and intent-to-treat populations. The primary efficacy variable was overall clinical resolution.Of 254 patients enrolled in the Study, 196 patients were included in the efficacy analyses. The majority of patients were male (58%) and Caucasian (85%) with a mean age of 49 years (range: 18 to 85 years). Only 3% of patients were hospitalized pretherapy. The most common pretherapy organisms identified, by culture or serology, in the valid for efficacy population (i.e. 147 organisms among 116 patients), were: Chlamydia pneumoniae (n=63; 54%),Mycoplasma pneumoniae (n=29; 25%), Streptococcus pneumoniae (n=14; 12%) and Haemophilus influenzae (n=13; 10%). End of therapy, follow-up and overall clinical resolution rates for the valid for efficacy population were 94%, 93% and 93%, respectively. The 95% CI for the overall clinical resolution rate was 88·1%, 95·9%. The overall bacteriological response for patients diagnosed by culture or serological criteria, was 91% (95% CI=84%, 96%). For patients who only met serological criteria for infection, the overall bacteriological response was 94% (60/64). Bacterial response rates for the four most commonly isolated pathogens were: 89% (56/63) for C. pneumoniae, 93% (27/29) for M. pneumoniae, 93% (13/14) for S. pneumoniae and 85% (11/13) for H. influenzae. Drug-related adverse events were reported in 33% (85/254) of moxifloxacin-treated patients. Nausea (9%), diarrhea (6%) and dizziness (4%) were the most commonly reported adverse events.Atypical organisms were isolated in high frequency among patients with CAP. Moxifloxacin 400 mg once daily for 10 days was effective and well-tolerated in the treatment of these adult patients with CAP. Moxifloxacin offers an effective treatment alternative for CAP due to both typical and atypical bacterial pathogens.
Article
Clinafloxacin is a broad-spectrum and highly potent member of the fluoro-quinolone class of antibiotics, with enhanced activity against difficult-to-treat Gram-negative, Gram-positive, and anaerobic pathogens. It appears that the most important factor in terms of structure-activity is the chloride atom at the C-8 position. Clinafloxacin has a tolerability profile that is similar to that of other quinolones. Structure-tolerability issues related to the C-8 halogen, such as phototoxicity, should not be a major problem for an antimicrobial agent used for nosocomial infections in hospitalised patients. Similar class-related tolerability issues of the quinolone class of antimicrobial agents, such as tendinitis, should not be an important issue in hospitalised patients. The activity of clinafloxacin against multidrug-resistant nosocomial pathogens and anaerobes suggests that this agent would be well suited for empirical therapy of nosocomial infections in seriously ill patients. Initial in vivo animal studies have confirmed that the enhanced in vitro activity of clinafloxacin against difficult-to-treat nosocomial pathogens results in increased in vivo efficacy. The success in animal models has led to the current series of ongoing clinical trials. Published data from these studies to date have confirmed the efficacy of clinafloxacin against difficult-to-treat nosocomial pathogens.
Article
The antimicrobial activity of temafloxacin against aerobic Gram-positive cocci was compared to that of ciprofloxacin, ofloxacin, fleroxacin and pefloxacin using the broth microdilution technique. Temafloxacin was more active than the other four fluoroquinolones, particularly for viridans streptococci and Streptococcus pneumoniae. The MIC90 of temafloxacin was at least four-fold lower than that of ciprofloxacin and ofloxacin for viridans streptococci and penicillin-susceptible pneumococci. The MIC50s and MIC90s of temafloxacin were equal to or lower than those of the other fluoroquinolones for methicillin-susceptible Staphylococcus aureus (MSSA), methicillin-resistant S. aureus (MRSA), and methicillin-susceptible and methicillin-resistant coagulase-negative staphylococci. Temafloxacin was more active against all the other Gram-positive aerobes (except Enterococcus faecalis) tested than the other fluoroquinolones.
Article
The in vitro activity of PD 127,391, a dihalogenated quinolone, was compared with those of ofloxacin, ciprofloxacin, nalidixic acid, gentamicin, and cefuroxime against 525 recent isolates and well-characterized antimicrobial agent-resistant strains. The MICs of PD 127,391 against 90% of members of the family Enterobacteriaceae, Bacteroides fragilis, Haemophilus influenzae, Neisseria sp., and Streptococcus pneumoniae were less than or equal to 0.12 microgram/ml. Some 90% of Pseudomonas aeruginosa and staphylococci were susceptible to 0.25 micrograms of PD 127,391 per ml. Against most strains, PD 127,391 was 2- to 8-fold more active than ciprofloxacin, but it was 64-fold more active than ciprofloxacin against B. fragilis. Strains of members of the family Enterobacteriaceae which were resistant to nalidixic acid were less susceptible to all of the quinolones tested, including PD 127,391. The MIC and minimum lethal concentration of PD 127,391 against three strains of Chlamydia trachomatis were each 0.06 microgram/ml, and the MIC against 90% of 21 strains of Mycobacterium tuberculosis was 1 microgram/ml. PD 127,391 was less active at pH 5, its maximal activity being at pH 7 to 8. The presence of urine at pH 5.9 decreased the bactericidal activity. The protein binding of PD 127,391 was 2 to 7%, and serum had little effect on activity.
Article
The antibacterial activity of ofloxacin, a new fluoroquinolone, was evaluated against a wide range of clinical bacterial isolates and compared with that of nalidixic acid, norfloxacin, enoxacin, pefloxacin and ciprofloxacin by determination of minimum inhibitory concentrations (MICs). Ofloxacin was very active against nalidixic acid-susceptible isolates of the Enterobacteriaceae (MIC⩽0·12 mg/1) and was also active against strains resistant to nalidixic acid (MIC ⩽ 2 mg/1). The activity was similar to norfloxacin, enoxacin and pefloxacin but some four-fold less than that of ciprofloxacin. All of the fluoroquinolones were highly active against Vibrio cholerae (MIC ⩽0·015 mg/1), V. parahaemolyticus (MIC ⩽0·12 mg/1) Aeromonas hydrophila (MIC ⩽0·03 mg/1), Plesiomonas shigelloides (MIC ⩽0·015 mg/1), Campylobacter jejuni (MIC ⩽0·5 mg/1), Neisseria spp., Haemophilus influenzae, H. ducreyi, Bordetella pertussis and Legionella pneumophila (MIC ⩽0·06 mg/1 for all species). Ofloxacin, ciprofloxacin and pefloxacin (MIC ⩽1,2 and 2 mg/1, respectively) showed similar activity against Staphylococcus spp. and were somewhat more active than enoxacin (MIC ⩽4 mg/1) and norfloxacin (MIC ⩽8 mg/1). Ofloxacin was moderately active against β-hacmolytic Streptococcus spp. (MIC ⩽2 mg/1), Corynebacterium diphtheriae (MIC ⩽1 mg/1) and Cory. jeikeium (MIC ⩽2mg/1) and somewhat less active against α- and non-haemolytic Streptococcus spp., Str. pneumoniae and Listeria monocytogenes (MIC ⩽mg/4 for all species) and Str. faecalis (MIC ⩽8 mg/1). The activity of ofloxacin, against these species, was similar to ciprofloxacin and four to eight times greater than norfloxacin, enoxacin and pefloxacin. Ofloxacin, and all of the fluoroquinolones, were less active against anaerobic than aerobic bacteria. Clostridium perfringens (MIC ⩽1 mg/1) was more susceptible to ofloxacin than were other anaerobic species and Cl. difficile (MIC ⩽16 mg/1) was more resistant. Ofloxacin was the most active compound tested against Chlamydia trachomatis SA2f (MIC ⩽0·5 mg/1) with only ciprofloxacin (MIC ⩽1 mg/1) approaching similar activity. Ofloxacin was moderately active against Mycoplasma hominis (MIC ⩽2 mg/1) and Ureaplasma urealyticum (MIC ⩽4 mg/1).
Article
The pharmacokinetics and tissue penetration of five quinolones were studied in volunteers. The compounds were norfloxacin (400 mg po), enoxacin (400 mg iv and 600 mg po), ciprofloxacin (100 mg iv and 500 mg po), ofloxacin (600 mg po) and pefloxacin (400 mg iv). Of the oral agents studied ofloxacin and ciprofloxacin were the most rapidly absorbed (Tmax = 1.2 h) and enoxacin the least (Tmax = 1.9 h). The serum levels attained were highest in the case of ofloxacin (after allowing for the higher dose administered). The serum half-lives were norfloxacin 3.75 h, ciprofloxacin 3.9 h (po), 4.0 h (iv), ofloxacin 7.0 h, enoxacin 6.2 h (po), 5.1 h (iv) and pefloxacin 10.5 h. All agents penetrated blister fluid readily. The 24 h urine recovery was 62% for oral enoxacin, 46.4% for iv enoxacin (plus 12.2% for oxo-enoxacin), 27% for norfloxacin, 30.6% for oral ciprofloxacin, 75.7% for iv ciprofloxacin, 73% for ofloxacin and 4.9% for pefloxacin (plus 9.2% for the norfloxacin metabolite and 17.8% for pefloxacin N-oxide).
Article
The in-vitro antibacterial activity of nalidixic acid and the 4-quinolones, ciprofloxacin, norfloxacin, enoxacin, ofloxacin, Pefloxacin, A-56619, A-56620 and CI-934 was assessed by determination of MICs. The 4-quinolones were all highly active against most isolates of Enterobacteriaceae, including nalidixic acid-resistant strains. Ciprofloxacin (MICs 0·002-2 mg/1) was the most active and A-56619 (MICs 0·008-32 mg/1) was the least active. A-56619, A-56620, ofloxacin, ciprofloxacin and CI-934 were highly active against Acinetobacter strains, Pefloxacin and enoxacin were slightly less active, and a few strains were resistant to norfloxacin. All the compounds, including nalidixic acid, were active against Aeromonas strains (MICs 0·001-0·12 mg/1). Ciprofloxacin (MICs 0·06-1 mg/1) was the most active compound against Pseudomonas aeruginosa; A-56619 and CI-934 (MICs 1-16 mg/1) were the least active against this species.. All the compounds were highly active against Haemophilus influenzae, Branhamella catarrhalis and Neisseria gonorrhoeae but the activity of all the compounds was poor against most isolates of Gardnerella vaginalis. All the 4-quinolones were active against staphylococci and CI-934 (MICs 0·03-0·25 mg/1) was the most active. CI-934 (MICs 0·06-2 mg/1) was also the most active compound against all streptococci. Most streptococci were sensitive also to ciprofloxacin (MICs 0·25-4 mg/1) but there were many isolates resistant to the other 4-quinolones. Against the anaerobic bacteria CI-934 was again the most active compound, particularly against the Gram-positive anaerobic cocci. Pefloxacin, enoxacin and norfloxacin had poor activity against most anaerobes. Ofloxacin, ciprofloxacin, A-56619 and A-56620 had good to moderate activity against all species of anaerobes except the Bacteroides fragilis group, against which none of the compounds was very active.