Sei sulla pagina 1di 21

Pharmacology & Therapeutics 143 (2014) 225245

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


journal homepage: www.elsevier.com/locate/pharmthera

Azithromycin: Mechanisms of action and their relevance for


clinical applications
Michael J. Parnham a,b,c,, Vesna Erakovic Haber d, Evangelos J. Giamarellos-Bourboulis e,f, Gianpaolo Perletti g,h,
Geert M. Verleden i, Robin Vos i
a

Fraunhofer Institute for Molecular Biology and Applied Ecology, Project Group Translational Medicine and Pharmacology, Frankfurt am Main, Germany
Institute of Pharmacology for Life Scientists, Goethe University Frankfurt, Frankfurt am Main, Germany
of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
dFidelta d.o.o., Zagreb, Croatia
e4th Department of Internal Medicine, University of Athens, Medical School, Athens, Greece
fIntegrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
gBiomedical Research Division, Department of Theoretical and Applied Sciences, University of Insubria, Busto A., Varese, Italy
hDepartment of Basic Medical Sciences, Ghent University, Ghent, Belgium
iRespiratory Division, Lung Transplantation Unit, University Hospitals Leuven and Department of Clinical and Experimental Medicine, KU Leuven, Belgium
b

cInstitute

article

info

Available online 11 March 2014


Keywords:
Azithromycin
Macrolide antibiotic
Mechanisms of action
Pharmacokinetics
Immunomodulation
Clinical efcacy

abstract
Azithromycin is a macrolide antibiotic which inhibits bacterial protein synthesis, quorum-sensing and reduces
the formation of biolm. Accumulating effectively in cells, particularly phagocytes, it is delivered in high
concentrations to sites of infection, as reected in rapid plasma clearance and extensive tissue distribution.
Azithromycin is indicated for respiratory, urogenital, dermal and other bacterial infections, and exerts
immunomodulatory effects in chronic inammatory disorders, including diffuse panbronchiolitis, posttransplant bronchiolitis and rosacea. Modulation of host responses facilitates its long-term therapeutic benet
in cystic brosis, non-cystic brosis bronchiectasis, exacerbations of chronic obstructive pulmonary disease
(COPD) and non-eosinophilic asthma.
Initial, stimulatory effects of azithromycin on immune and epithelial cells, involving interactions with
phospholipids and Erk1/2, are followed by later modulation of transcription factors AP-1, NFB, inammatory
cytokine and mucin release. Delayed inhibitory effects on cell function and high lysosomal accumulation
accompany disruption of protein and intracellular lipid transport, regulation of surface receptor expression, of
macrophage phenotype and autophagy. These later changes underlie many immunomodulatory effects of
azithromycin, contributing to resolution of acute infections and reduction of exacerbations in chronic airway
diseases. A sub-group of post-transplant bronchiolitis patients appears to be sensitive to azithromycin, as may
be patients with severe sepsis. Other promising indications include chronic prostatitis and periodontitis, but
weak activity in malaria is unlikely to prove crucial. Long-term administration of azithromycin must be balanced
against the potential for increased bacterial resistance. Azithromycin has a very good record of safety, but recent
reports indicate rare cases of cardiac torsades des pointes in patients at risk.
2014 Elsevier Inc. All rights reserved.

Abbreviations: AECOPD, acute exacerbations of COPD; AP-1, activator protein-1; ARDS, acute respiratory distress syndrome; AUC, area under the plasma concentration-versus-time
curve; BAL, broncho-alveolar lavage; BOS, bronchiolitis obliterans syndrome; CAD, cationic amphiphilic drugs; CAP, community acquired pneumonia; CBP, chronic bacterial prostatitis;
CF, cystic brosis; CFTR, CF transmembrane conduction regulatory protein; C max, peak plasma concentration; COPD, chronic obstructive pulmonary disease; COX, cyclo-oxygenase;
cPLA2, cytoplasmic phospholipase A2; DNFB, dinitrouorobenzene; DPB, diffuse panbronchiolitis; DTH, delayed type hypersensitivity; ERK, extracellular signal-regulated kinase; FGF, broblast growth factor; GM-CSF, granulocytemacrophage colony stimulating factor; GVH, graft versus host reaction; HR, hazard ratio; IL, interleukin; i.n, intranasal; i.p, intraperitoneal; i.t,
intratracheal; i.v, intravenous; JNK, c-Jun NH(2)-terminal kinase; LC3, microtubule-associated protein 1A/light chain 3; LOS, length of stay; LPS, bacterial lipopolysaccharide; LRTI, lower
respiratory tract infection; MAPK, mitogen-activated protein kinase; MDR1, multidrug resistance protein 1; MIC, minimum inhibitory concentration; MLSbK, macrolides, lincosamines,
streptogramin B and ketolides; MMP, metalloproteinase; MODS, multiple organ dysfunction syndrome; MPO, myeloperoxidase; MUC5AC, mucin 5AC; MV, mechanical ventilation;
NFB, nuclear factor kappaB; NGU, non-gonococcal urethritis; NK, natural killer cells; NR, not reported; OR, odds ratio; p.o, oral; PG, prostaglandin; PI, phosphatidylinositol; PI3K,
phosphoinositide-3-kinase; PK, pharmacokinetic; PMNL, polymorphonuclear leukocytes; PS, phosphatidylserine; RCT, randomized controlled clinical trial; ROS, reactive oxygen species;
SAA, serum amyloid A protein; SMC, smooth muscle cell; SREBP, sterol regulatory element binding protein; STAT, signal transducers and activators of transcription; STD, sexually transmitted disease; t1/2, plasma half-life; TLR, Toll-like receptor; T max, time to Cmax; TNF, tumor necrosis factor alpha; Th, T helper cell; VAP, ventilation assisted pneumonia; V d, volume of
distribution; VEGF, vascular endothelial growth factor.
Corresponding author at: Fraunhofer IME-TMP, Theodor-Stern-Kai 7, D-60596 Frankfurt am Main, Germany. Tel.: +49 69 6301 84234.
E-mail addresses: michael.parnham@ime.fraunhofer.de (M.J. Parnham), Vesna.ErakovicHaber@glpg.com (V.E. Haber), egiamarel@med.uoa.gr (E.J. Giamarellos-Bourboulis),
gianpaolo.perletti@uninsubria.it (G. Perletti), geert.verleden@uzleuven.be (G.M. Verleden), robin.vos@uzleuven.be (R. Vos).

http://dx.doi.org/10.1016/j.pharmthera.2014.03.003
0163-7258/ 2014 Elsevier Inc. All rights reserved.

226

M.J.M.J.
Parnham
Parnham
et al.et/ al.
Pharmacology
/ Pharmacology
& Therapeutics
& Therapeutics
143 143
(2014)
(2014)
225245
225245

Leclerq, 2002).Contents
In S. pyogenes there are three types of inducible
phenotype (a) iMLSbK-A, (b) iMLSbK-B, which is highly resistant to
azithromycin (MICN128 g/mL) and (c) iMLSbK-C with a medium resis1.Introduction
. . . . . .against
. . . . . . azithromycin
. . . . . . . . . . . . (Giovanetti
...
tance prole (MICs
up to 16 g/mL)
2.Antibacterial mechanisms of action . . . . . . . . . . . . . . . .
et al., 1999; Giovanetti
et al., 2002). In addition to methylation of
3.Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . .
23rRNA, resistance
can be achieved by mutation of various genes coding
4.Immunomodulatory activities . . . . . . . . . . . . . . . . . . .
for ribosomal components.
This type ofinresistance
been observed
5.Clinical applications
respiratoryhas
infections
and airway inammation
infrequently in S.6.Infections
pneumonia,ofS.thepyogenes
H.and
inuenzae,
the most infections .
urogenitaland
tract
sexually-transmitted
common mutations
occurring
in domain
V rRNA
7.Clinical
application
in sepsis
. . . and
. . . .proteins
. . . . . . . L4
. . . and
..
L22 (Tait-Kamradt
et al.,clinical
2000; Franceschi
8.Other
applicationset. .al.,
. . . 2004).
...............
Mef transporter9.Safety
(efux .pump)
. . . . . .expression
. . . . . . . . . .typically
. . . . . . . .results
. . . . . in a lower
10.Conclusions
. . . .14. . . and
. . . .15-membered
. . . . . . . . . . . . macrolide
....
level of specic resistance
towards
Conict
of
interest
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
. . .is. .referred
....
antibiotics (MICs 432 ug/mL) and this type of resistance
Acknowledgments
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
......
to as the M phenotype (Leclercq, 2002).
References
. . . . . . . . . antibiotics
. . . . . . . . . .is. .linked
. . . . . .with
. . . . an
Increased clinical
use of macrolide
increase in pneumococcal macrolide resistance. The Alexander Project,
monitoring antibiotic resistance among respiratory pathogens, reported
a global rate of pneumococcal macrolide resistance between 16.5% and
21.9% for 1996 and 1997 (Felmingham & Gruneberg, 2000), increasing
to 24.6% from 1998 to 2000 (M. R. Jacobs & Johnson, 2003). Over this
period, macrolide-exceeded penicillin-resistance in 19/26 countries.
The PROTEKT US surveillance study reported similar ndings. From
2000 to 2004, erythromycin resistance among S. pneumoniae isolates
1. Introduction
remained high (Jenkins et al., 2008), mef(A) being the most common
macrolide resistance genotype, accounting for N60% of tested isolates
Azithromycin,
a second
generation
(Farrell & Jenkins,
2004).
Althoughmacrolide,
macrolidebroad-spectrum
resistance rates have
antibacterial,
hasstabilized,
received increasing
attention
in recent
yearswith
because
subsequently
the prevalence
of clonal
isolates
a comof additional
effects
on
host-defence
reactions
and
chronic
human and
bined erm(B) and mef(A) genotype and a high-level
of macrolide
diseases.
It
is
the
prototype
15-membered
lactone
ring
azalide,
synthemultidrug resistance has increased.
sized Distribution
in the early 1980s
as a semi-synthetic
derivativevaries
of erythromycin.
of macrolide
resistance genotypes
between and
Discovered
around the
same
time by
researchers
at Pzer
in the
United
within countries.
In the
studies
mentioned
above,
highest
resistance
States
(Bright
&
Hauske,
1984)
and
at
PLIVA,
Croatia
(Kobrehel
al.,
rates were in Asia and some European countries (France, Italy, et
Spain,
1982),
PLIVAbeing
patented
rstand
and2560%,
licensed respectively.
the compound
to Pzer.resistance
Belgium),
N70%
Macrolide
With
much
improved
pharmacokinetic
over erythromycin,
was
common
among
S. pneumoniaeproperties
isolates (77.281.9%)
in Japan
azithromycin
became the
most
widely used
during PROTEKT
years
19992004,
the broad-spectrum
erm(B) genotypeantibacteaccounting
rialfor
in North
America.
Pzer
Inc's
Arthur
E.
Girard
and
Gene
Michael
most resistance (Inoue et al., 2008). In Portugal (19942002),
Bright,
togetherofwith
PLIVA's Slobodan
Djokic (posthumously)
andcorrelatemergence
macrolide-resistant
S. pneumoniae
strains clearly
Gabrijela
in 2000
American
Chemical
ed withKobrehel,
increasedreceived
azithromycin
usethe
(Dias
& Canica,
2004).Society's
awardInofaHeroes
of Chemistry
who have
promoted human
in
recent study
on commensal
Staphylococcus
aureuswelfare
in Europe,
the except
area offor
health
for their
of Zithromax
(azithromycin).
penicillin,
the discovery
highest recorded
resistance
rate was to
Azithromycin
the same
mechanism
antibacterial
azithromycin shares
(from 1.6%
in Sweden
to 16.of9%
in France) action
(den Heijer
as other
(Allen, 2002),
butclarithromycin
accumulates more
et al.,macrolide
2013). Asantibiotics
usage of azithromycin
and
has increased
effectively
in
phagocytes,
thus
being
delivered
in
high
concentrations
for indications beyond RTI, resistance has started to emerge in nonto sites
of
infection
(Miossec-Bartoli
et
al.,
1999;
Wilms
et
al.,
2006).
respiratory pathogens. For instance, macrolide treatment of healthy
It also
inhibits in
bacterial
quorum-sensing
reduces formation
volunteers,
comparison
to placebo, and
signicantly
increased of
the
biolm
and mucus
production, whichoral
extend
its range for
of antibacterial
proportion
of macrolide-resistant
streptococci
up to 180 days
actions
(Tateda
et
al.,
2001;
Hoffmann
et
al.,
2007).
As
an
antibiotic,
after treatment (Malhotra-Kumar et al., 2007). Consequently, as with
azithromycin
is indicated
for respiratory,
dermal
other
all antibiotics,
azithromycin
should beurogenital,
administered
for alland
relevant
bacterial
infections,
butconsideration
has benecialofeffects
in chronicbenets
inammatory
conditions
with due
the associated
and risks.
disorders such as diffuse panbronchiolits, bronchiolitis obliterans and
rosacea. Efcacy in these conditions is ascribed to immunomodulatory
effects on innate and adaptive immune responses. Modulation of host
response reactions also accounts, at least partially, for benecial effects
in cystic brosis, non-cystic brosis bronchiectasis, bronchial obliterans
syndrome (BOS) and chronic obstructive pulmonary disease (COPD).
Azithromycin is well-tolerated and has a very good record of safety.
Although macrolides have a class warning for potential cardiac QT prolongation, azithromycin does not show this effect under experimental
conditions (Milberg et al., 2002). Until recently, only a handful of cases
of QT prolongation had been reported for patients treated with the
drug (Kezerashvili et al., 2007). This is mainly because azithromycin,
unlike other macrolide antibiotics, does not interact with CYP3A4,
despite a minor interaction with the anti-coagulant warfarin (Kanoh
& Rubin, 2010; Mergenhagen et al., 2013). Recently, evidence for
increased risk of QT prolongation with azithromycin has appeared, but
mainly
in patients with greater susceptibility to adverse cardiac effects
3. Pharmacokinetics
(Giudicessi & Ackerman, 2013).
We review here the antibacterial actions and pharmacokinetics of
Early pharmacokinetic
(PK) studies on azithromycin,
low
azithromycin,
as well as its immunomodulatory
effects and indicating
the
plasma levels,
were in
incorrectly
interpreted
as reecting
poor PK propermechanisms
involved,
comparison
to other macrolide
antibiotics.
Azithromycin
is now uses
known
to have a largedrawing
volume attention
of distribution,
Weties.
discuss
the main clinical
of azithromycin,
achievingindications
high tissueand
concentrations
is its
efciently
delivered to sites
to emerging
emphasisingand
how
pharmacokinetic
of infection. Compared with older generation macrolides, it is more
stable in acidic media and has a longer half-life, allowing for once a
day or even single dose treatment (Amsden, 1995, 2001). Moreover,
azithromycin is barely metabolized, with no active metabolites and
does not induce CYPs (Amsden, 1995; Nightingale, 1997).

3.1. Cellular accumulation


Concentrations of azithromycin achieved in tissues, are up to 100fold higher than those in plasma (Foulds et al., 1990; Shepard &

Falkner, 1990) as a result of high cellular accumulation, particularly in


phagocytes, in which at least 200-fold higher intracellular than extracellular concentrations can be achieved (Bosnar et al., 2005; Stepanic et al.,
. . . . specic
. . . . accumulation
. . . . . of
. .azithromycin
. . . . occurs
226 at
2011). Consequently,
. . . . . . . . . . . . . . . . . . .
226
sites of inammation
(Miossec-Bartoli et al., 1999; Wilms et al.,
2006).
. . . . . . . . . . . . . . . . . . .
227226
In drug-free. medium,
preloaded cells release azithromycin slowly,N80%
. . . . . . . . . . . . . . . . . .
229226
being retained,
for the
. . so. cellular
. . . accumulation
. . . . . . and
. .retention
. . . account
. .
233227
long half-life. of
. azithromycin
. . . . . . in. vivo
. . (Bosnar
. . . et
. al.,
. . 2005;
. . Stepanic
236227
et al., 2011).. Accumulation
. . . . . . of. azithromycin
. . . . . .in .polymorphonuclear
. . . .
237227
cells (PMNLs)
site of in. . acts
. .as. an. innate
. . .targeted
. . . delivery
. . . .system
. . .to the
238228
fection. Degranulation
. . . . . of. PMNs
. . . by. bacteria
. . . .then
. .releases
. . . azithromycin
238228
. . . . space,
. . . where
. . .it exerts
. . . its
. antibacterial
. . . . . activity.
239229
into the extracellular
. . also
. . accumulates,
. . . . . . to. a .less
. and
. . varying
. . . extent,
.
239229
Azithromycin
in other
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
239229
cells such as epithelial cells, broblasts, lymphocytes, hepatocytes
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
239229
(Bosnar et al., 2005; Gladue & Snider, 1990; Matijasic et al., 2012;
Stepanic et al., 2011). Fibroblasts, due to their wide distribution, have
been proposed as a potential reservoir for azithromycin, slowly releasing or passing it to nearby phagocytes for transport to the site of infection (Gladue & Snider, 1990; McDonald & Pruul, 1991). Based on such
retention in epithelial lining and bronchoalveolar cells, macrolides,
including azithromycin, nd veterinary use for the treatment of pneumonia (Villarino & Martin-Jimenez, 2013).
Most and
intracellular
azithromycinactions
is located
in acidic
organelles
properties
immunomodulatory
contribute
to the
effects such
as lysosomes. Initially shown to concentrate in the cytoplasm and plasobserved.
malemma of PMNLs and monocytes (Wildfeuer et al., 1993), the vast
majority of azithromycin in these cells is associated with the granular
2. Antibacterial
mechanisms
of Miossec-Bartoli
action
fraction (Carlier
et al., 1994;
et al., 1999). Recent data
support its lysosomal localization in hepatocytes (Matijasic et al.,
2012). In keeping with lysosomal accumulation, azithromycin causes
2.1.
InhibitorypHactions
lysosomal
change and functional impairment in murine macrophages (Nujic et al., 2012a).
The exact mechanism
cellular accumulation
remains
unclear. One
Azithromycin,
like other of
macrolide
antibiotics, inhibits
bacterial
possibility
is passive
transmembrane
transport
andthe
trapping
by of
protonprotein
synthesis
by binding
to and interfering
with
assembly
acidic
organelles,
as this
inhibited
simply
by polyneutralizatheation
50S in
large
ribosomal
subunit
andcan
thebe
growth
of the
nascent
tion ofchain
lysosomal
pH, markedly
increasing
azithromycin
efux
(Hand &
peptide
(Champney
& Burdine,
1998; Champney
et al.,
1998;
Hand,et2001).
Moreover,
macrolide
cellular accumulation
and retention
Hansen
al., 2002).
It binds
at the polypeptide
exit tunnel, close
to
be predicted
by experimentally
determined
physicochemical
thecan
peptidyl
transferase
center (PTC) on
the 23S rRNA,
but does not
parameters
and the
calculated
of positively
charged atoms
inhibit
PT activity,
in contrast
to number
larger macrocyclic
antibiotics.
The
(Stepanic
et al., 2011).leads
Like to
other
cationic
amphiphilic
(CADs),
basicity
of azithromycin
faster
penetration
of the drugs
outer memmacrolides
mayeffective
degradeentrance
cell membranes,
together thereby
with which
the
branes
and more
into the bacteria,
enhancing
drugs against
are carried
to lysosomesbacteria
(Baciu(Dinos
et al., 2006).
activity
Gram-negative
et al., 2001).
An active
mediated
protein(s),
Binding
sitesuptake
on themechanism,
bacterial ribosome
forby
thetransporter
structurally
different is
supportedlincosamines,
by dependencestreptogramin
of accumulation
onketolides
cell viability,
temperamacrolides,
B and
(MLSbK)
overpH and extracellular
calcium
(Gladue
al., 1989;
lapture,
signicantly,
so that changes
in a concentration
single ribosomal
regionetsimultaMtairag
et al.,
1995; Pascual
et al., 1995).
Macrolides
neously
alter
susceptibility
to various
MLSbK
antibiotics.also compete for
cell accumulation
(Munic
et al., 2010).agent
Theyagainst
are inhibitors
of ATPAlthough
ineffective
as a bactericidal
Pseudomonas
binding cassette
transporters
as P-glycoprotein
(multidrug
aeruginosa
at clinically
relevantsuch
concentrations,
azithromycin
inhibits
protein
1, growth-stimulating,
MDR1) (Wang et al.,quorum-sensing
2000; Yasuda etcomal., 2002;
theresistance
generation
of both
Asakura
al., 2004;
Municwhich
et al.,protects
2010), multidrug
resistance from
associated
pounds,
andetalginate
biolm
the micro-organism
protein actions
2 (MRP2)
(Sugie
2004)
and organic
anion-transporting
antibiotic
(Tateda
et et
al.,al.,
2001;
Hoffmann
et al.,
2007). Efcacy
polypeptide,
OATP (Garver
et al., factor
2008) production
and substrates
MDR1.
against
both P. aeruginosa
virulence
andfor
biolm
Ranking as
of well
macrolides
for cellular
accumulation
is proportional
formation,
as its ability
to decrease
the minimum
inhibitoryto
cellular expression
of anti-pseudomonas
MDR1, its overexpression
decreasing
macrolide
concentration
(MIC) of
agents (Lutz
et al., 2012)
2- to 80-fold,
with azithromycin
areaccumulation
related to ribosomal
proteinparticularly
synthesis inhibition
(Kohler etand
al.,erythromycin (Munic et al., 2010).
2007).
It seems likely
bothmoderate
passive and
activeagainst
mechanisms
are inAzithromycin
alsothat
shows
activity
the malaria
parvolved,
variability
in macrolide
accumulation
resulting
changes
asite,
Plasmodium
spp.,
exhibited as
delayed killing,
whichfrom
is achieved
in
acidic
organelles
or
in
different
transporter
proteins.
via the apicoplast (Dahl & Rosenthal, 2007). Clinical studies, however,
failed to demonstrate even equivalence of three-day treatment with
azithromycin to other antimalarials or drug combinations (van Eijk &
Terlouw, 2011).

2.2. Resistance phenotypes


Bacteria resist azithromycin in two ways: a) by changing the target/
binding site via methylation of key rRNA nucleotides or mutation of
some ribosomal components and b) by efux pump activity, thereby
decreasing its intrabacterial accumulation.
3.2. Pharmacokinetic
Ribosome
methylation properties
is the most important resistance mechanism
for all MLSbK antibiotics (Sutcliffe & Leclerq, 2002). The MLSbK-II
phenotype, characterized by the presence of Erm methylases, is highly
Extensive data are available on the clinical PK of various formularesistant to all MLSbK antibiotics, including azithromycin, which
tions of azithromycin, both under normal and pathologic conditions
induces erm genes in Streptococci and in Staphylococci (Sutcliffe &
(Table 1). Knowledge of the salient kinetic features is crucial to an
understanding of its therapeutic effects.

227

228

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

Azithromycin shows plasma protein binding of approximately 30%


and relatively low oral bioavailability (1737%) (Foulds et al., 1990;
Luke & Foulds, 1997). Following a single standard 500 mg oral dose,
peak plasma concentrations of 0.20.4 mg/L are attained, with a Tmax
of 24 h that may be delayed signicantly in elderly subjects (Coates
et al., 1991) (Table 1). Single oral doses of 1.5 g azithromycin attain
peak plasma concentrations as high as 1.46 g/mL (Amsden, 1996).
The kinetics of azithromycin are best described by a 3-compartment
pharmacokinetic model (Ballow et al., 1998; Pene Dumitrescu et al.,
2013).
When administered as 500 mg extended, rather than immediate
release tablets, a threefold higher AUC024 and twofold higher Cmax
may be attained (Liu et al., 2011; Lucchi et al., 2008) (Table 2). Plasma
clearance of azithromycin is remarkably high (50 L/h), and the volume
of distribution (Vd) is approximately 30 L/kg (Singlas, 1995). Hence, low
plasma concentrations are the consequence of rapid blood-tissue distribution. The plasma half-life of azithromycin is approximately 70 h or 50 h
following oral or intravenous formulations, respectively (Lode et al.,
1996). This remarkable pharmacokinetic characteristic of azithromycin
allows infrequent dosing (in bacterial urethritis or cervicitis, single
administration may be sufcient), and ensures optimal patient compliance and sustained exposure to the drug. In fact, the AUC024 after a
single 500 mg oral dose is 3 gh/mL (Matzneller et al., 2013), reaching
as high as 811 gh/mL after intravenous administration of 500 mg
(Luke & Foulds, 1997; Rodvold et al., 2003). After a standard dose of
500 mg/day for 3 days, much of the drug is present in blood leukocytes
(Matzneller et al., 2013; Pene Dumitrescu et al., 2013). On day 3, blood
concentration was double that in plasma and Vd to the periphery was
2980 L, compared with 439 L in plasma. After 30 days, drug concentrations were 4-fold higher in blood than in plasma. In healthy volunteers,
much less drug is present in tissues (Matzneller et al., 2013). Release is
likely induced by inammatory stimuli.
Azithromycin is mainly eliminated unchanged in the feces by both
biliary excretion and trans-intestinal secretion. Only about 6% (oral
dose) to 12% (i.v.) is recovered unchanged in the urine (Singlas,
1995). Using ileostomy uid sampling and correction for the fraction
of i.v. azithromycin recovered in the uid, approximately 45% of an
oral dose of 500 mg was unabsorbed after intestinal passage up to the
ileal stoma (Luke & Foulds, 1997). Thus, incomplete absorption, rather
than a rst-pass effect, is the major cause of the sub-optimal bioavailability of azithromycin (Luke & Foulds, 1997).
The inactive descladinose and the active 9a-N-desmethyl metabolites are frequently recovered because of acidic degradation in the gut
(Luke & Foulds, 1997; Raines et al., 1998). Hepatic biotransformation
is marginal (Curatolo et al., 2011). Importantly, azithromycin is neither
metabolized by nor an inhibitor of CYP3A4 and therefore, does not
interact with many commonly used medications that are oxidatively
metabolized by CYP3A4 (Shakeri-Nejad & Stahlmann, 2006). Other

factors than drug metabolism are thought to account for serious adverse
effects seen with azithromycin (Fleet et al., 2013) (see Section 9).
PK parameters may be affected considerably by co-administration of
azithromycin with food, prolongation of Tmax, 50% reduction of Cmax,
and of AUC072h having been observed in fed, compared to fasting patients (Table 1) (Curatolo et al., 2011). Azithromycin pharmacokinetics,
in keeping with low hepatic metabolism, are not signicantly affected
by mild or moderate hepatic impairment or class-A or -B liver cirrhosis
(Mazzei et al., 1993), nor by mild or moderate renal insufciency
(Hofer et al., 1995; Singlas, 1995). PK parameters are also virtually
unchanged in patients with diabetes (Ernst et al., 2000). Hence, no
dose modications of azithromycin appear to be necessary in patients
suffering from these conditions. However, severe renal impairment
noticeably increases Cmax and AUC0120 (http://www.medicines.org.
uk/emc/medicine/26131). Interestingly, in such patients on permanent
peritoneal dialysis due to oliguric end-stage renal failure (Kent et al.,
2001), the Cmax of azithromycin increased but Tmax appeared to be in
the normal range.

3.3. Tissue distribution


The distribution of azithromycin into inamed tissues and compartments has been extensively investigated. In bronchial washings and
lung tissue biopsy specimens (Di Paolo et al., 2002), in lungs, in sinus
mucosal tissue (Ehnhage et al., 2008; Fang et al., 2009), in middle ear
uids in children with otitis media (Pukander & Rautianen, 1996;
Scaglione et al., 1999), in the aqueous humor and in conjunctival biopsy
specimens (Tabbara et al., 1998), in tissues lining periodontal pockets
(Gomi et al., 2007), and in inammatory skin blisters (Ballow et al.,
1998), azithromycin concentrations were always higher than those in
plasma. This tissue accumulation offsets the sub-optimal absorption of
the drug. In contrast to many other antibiotics, macrolides penetrate
well into prostatic tissue and seminal uids. For example, Cmax of
azithromycin and clarithromycin in human prostatic tissue after 500
or 750 mg oral dosing is 2.54 and 3.83 g/ml, respectively, about 20and 2-fold higher than in plasma (Foulds et al., 1991; Giannopoulos
et al., 2001). This is important with regard to the use of azithromycin
in prostatitis (see Section 6.2).
In summary, azithromycin concentrates to a considerable extent in
many inamed tissues and uids. In this respect, its extensive uptake
into tissue-inltrating leukocytes is a signicant contributing factor
(see Sections 3.1 and 4.1.2).

3.4. Pediatric pharmacokinetics


The AUC0 of azithromycin (2 g dose) is reduced in pregnant
women (Salman et al., 2010), and plasma half-life is similar in both
placenta and fetus (70 h), though considerably shorter in amniotic

Table 1
Azithromycin pharmacokinetic data extracted from a sample of studies published between the years 1996 and 2013.

Dose/route (mg)
Cmax (g/mL)
tmax (h)
AUC (gh/mL)
t1/2 (h)
Ke (h1)
Clplasma (L/h)
F (%)
Vd (L/Kg)

(Luke & Foulds, 1997)

(Amsden, 1996)

(Rodvold et al., 2003)

(Matzneller et al., 2013)

(Ballow et al., 1998)

(Coates et al., 1991)

500 IV

500 oral

1500 oral

500 oral

500 IV

500 oral

500 oral

0.21
2.5
1.27

1.46

13.1

0.54

11.2

8.2

0.46
2.92
3.05

0.27

500 oral young


(1840 years)
0.41
2.50
2.5

89.5 L/Kg

109.2

3.40
0.7
11
50.2
0.00138
46.6
17.8

78.6

Cmax, peak plasma concentration; tmax, time- to Cmax; Ke, pharmacokinetic elimination constant; Cl, plasmatic clearance; F, oral bioavailability; Vd, distribution volume.
AUC, area under- (denite integral of-) the plasma concentration-versus-time curve
AUC024.
AUC0.

500 mg oral elderly


(6585 years)
0.38 g/mL
3.80 h
3 gh/mL

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

229

Table 2
Pharmacokinetic data for azithromycin administered in different oral formulations or in fasting vs. fed patients.
(Lucchi et al., 2008)

(Liu et al., 2011)

Dose/route

500 mg immediate
release oral

2 g extended
release oral

Cmax (mg/L)
tmax (h)
AUC024 (gh/mL)
Clplasma (L/h)
Vd (L)

0.39
4
3.1

0.94
4
10

500 mg IR oral

0.40
2
2.67

518

uid (3 h) and umbilical cord serum (12 h) (Ramsey et al., 2003). The
serum half-life in extremely preterm infants is shorter (58 h) than at
full term or in adults. Importantly, preterm infants, treated with
10 mg/kg intravenous azithromycin, appear to be overexposed (about
3.2-fold) to the drug compared to older children (age: 0.52 years)
(Jacobs et al., 2005; Hassan et al., 2011) and clearance is low (0.18 L/h
in 1 kg pre-term neonate vs 0.98 L/h/kg in older children). This may
be attributed to the immature biliary excretion pathways in preterm
neonates (Hassan et al., 2011).
In pediatric subjects (mean 24 months) given an immediate-release
formulation of azithromycin (30 mg/kg), plasma concentrations and
drug exposure were higher than in adults (Liu et al., 2011), necessitating
a dose adjustment (Table 1). When an extended release formulation
was administered to children, the Cmax was still elevated compared to
adult subjects, but the AUC024 was lower (Liu et al., 2011).

4. Immunomodulatory activities
Early studies on macrolide antibiotics and host defense revealed
inhibitory effects on neutrophils in vitro, on experimental inammation
(Culic et al., 2001) and particularly actions on cytokine release (Bartold
et al., 2013). While azithromycin was shown in the late 1980s to inhibit
inammation and lysosomal enzyme release in arthritic rats (Carevic &
Djokic, 1988), subsequent studies were carried out predominantly with
erythromycin, clarithromycin and roxithromycin. All macrolide antibiotics inhibit various standard models of inammation (Ianaro et al.,
2000; Culic et al., 2001; Ivetic Tkalcevic et al., 2012) and more recent
studies have sought to clarify immunomodulatory mechanisms in vivo
(Table 3) and at the molecular level.

4.1. Cellular functions


Modulation of host defense by azithromycin and other macrolide
antibiotics occurs through interaction with structural cells, such as
epithelial or endothelial cells, smooth muscle cells or broblasts, as
well as with leukocytes (macrophages, polymorphonuclear leukocytes
or neutrophils, mononuclear leukocytes or monocytes, T cells and
dendritic cells). Immunomodulatory effects on cellular functions of the
macrolide antibiotics as a class were reviewed a few years ago (Kanoh
& Rubin, 2010).

4.1.1. Structural cells


In epithelial cells, azithromycin maintains integrity and the
transepithelial resistance in the face of permeability induced by
virulence factors from P. aeruginosa, in conjunction with rearrangement
of tight junction adhesion molecules, including claudins (Asgrimsson
et al., 2006; Halldorsson et al., 2010). Increased cell integrity is also
accompanied by reduced mucin secretion (Imamura et al., 2004),
altered signal transduction (MAPK pathway) (see Section 4.2.4) and
attenuated expression of inammatory (e.g. IL-6 and 8, MMP-1, 2, 9,
10 and 13, and GM-CSF), lipid metabolism and cell cycle pathways
(Gielen et al., 2010; Murphy et al., 2008b; Ribeiro et al., 2009). In
bronchial epithelial cells infected with rhinoviruses, azithromycin
stimulated interferon and interferon-stimulated gene expression, in

2 g extended
release oral

(Muto et al., 2011)

(Curatolo et al., 2011)

2 g extended
release oral

500 mg oral fast dissolving


formula in fasting/fed patients

0.72
3.5
7.87
122

0.4/0.2
2/4
103
1830

association with reduced rhinovirus proliferation and release (Gielen


et al., 2010). Such activities undoubtedly contribute to therapeutic
benet in respiratory infections.
In airway smooth muscle cells, azithromycin inhibits interleukin (IL)17-induced IL-8 and 8-isoprostane release, whereas dexamethasone
failed to attenuate the IL-8 production (Vanaudenaerde et al., 2007).
On the other hand, azithromycin and dexamethasone (more strongly)
both reduce broblast growth factor (FGF)-induced vascular endothelial
growth factor (VEGF) production through interaction with p38 mitogenactivated protein kinase (MAPK)-signaling (Willems-Widyastuti et al.,
2013). Azithromycin also demonstrates an anti-proliferative and autophagic effect (Stamatiou et al., 2009) (Section 4.2.2), and directly relaxes
pre-contracted airway smooth muscle cells (Daenas et al., 2006).

4.1.2. Leukocytes
In neutrophils, subsequent to its pronounced accumulation (see
Section 3.1), slow efux of azithromycin prolongs its effects on the
cells in vitro and in vivo (Bosnar et al., 2005; Culic et al., 2002). This
extensive intracellular retention of azithromycin, for up to 28 days in
human neutrophils after standard dosing, leads to initial stimulation
of neutrophil degranulation and phagocytosis-associated oxidative
burst, further contributing to its antibacterial activity (Culic et al.,
2002). Acute inhibitory effects of azithromycin include downregulation of neutrophil chemokine production (IL-8, GRO-, MPO);
late effects comprise attenuation of neutrophil oxidative burst responses, down-regulation of myeloperoxidase (MPO) production,
increased neutrophil apoptosis and attenuation of chemokine (IL-8)and leukotriene (LT)B4-dependent and chemokine-independent
neutrophil chemotactic responses by inhibition of transcription factors,
nuclear factor kappa-B (NF-B) and activator protein-1 (AP-1) (Culic
et al., 2001; Culic et al., 2002; Tamaoki et al., 2004; Tsai & Standiford,
2004). Azithromycin also inhibits prostaglandin (PG)E 2-synthesis by
suppressing expression of prostaglandin synthetic enzymes (COX-1
and COX-2) in both lipopolysaccharide (LPS)-stimulated polymorphonuclear leukocytes and monocytes (Miyazaki et al., 2003). In the latter,
azithromycin decreases tumor necrosis factor- (TNF-) and granulocytemacrophage colony stimulating factor (GM-CSF) production
(Khan et al., 1999).
In sputum cells (81.7% neutrophils, 12.9% monocytes/macrophages)
isolated from steroid-nave patients with COPD, azithromycin and
other macrolides inhibited, in a concentration-dependent manner,
the release of a variety of inammatory cytokines and chemokines
(Marjanovic et al., 2011). The macrolides were more broadly inhibitory
than the corticosteroid, dexamethasone, the PDE4 inhibitor, roumilast,
and the p38 kinase inhibitor, SB203580. Azithromycin was less effective
than clarithromycin and roxithromycin, but its broad inhibitory activity
may provide corticosteroid-sparing effects in treatment of respiratory
inammation (see Section 5.4). An extensive list of the effects of
azithromycin on cytokine production, generally, was published recently
(Bartold et al., 2013). Azithromycin inhibits neutrophilic inammation
in vivo induced by LPS given by various routes (Table 3). Lung
neutrophilia induced by intranasal LPS is inhibited by azithromycin in
association with a reduction in lung GM-CSF and IL-1, both of which
were also inhibited by azithromycin in macrophages in vitro (Bosnar

230

M.J.M.J.
Parnham
Parnham
et al.et/ al.
Pharmacology
/ Pharmacology
& Therapeutics
& Therapeutics
143 143
(2014)
(2014)
225245
225245

action of
azithromycin
on broblast
MPR300 (and/ordendritic
on membrane
et al.,
(Kanoh
2009).
&Subsequently,
Rubin, 2010; Shinkai
the action
et on
al.,lung
2008).
neutrophilia
Mainly based
was on
localized
studies on
complex
(MHC)
class II molecules
in LPS-stimulated
cells,
phospholipid
uidity)receptor
may account
for its
inhibitionIL-12
of theproduction
pinocytosis
to the
mucus
alveolar
and cytokine
macrophages
production
(Bosnar
byetairway
al., 2011).
epithelial and inammatory
and
attenuates Toll-like
(TLR)-4
expression,
ofallostimulatory
macromolecules
and their
transport potential
from the for
plasma
membrane
In
cells,
alveolar
the authors
macrophages,
concluded
azithromycin
that macrolide
attenuates
antibiotics,
LPS-induced
including
and
capacity,
suggesting
modulation
of to
endo/lysosomes
(Tyteca
et al.,et2001).
Acute
effects
on MPR300
production
azithromycin,
and expression
exert biphasic
of pro-inammatory
stimulatory thencytokines
inhibitorythrough
effects inhion
allogeneic
responses
(Iwamoto
al., 2011,
2013).
Animal
studies may
also explain
the paradoxical
by azithromycin
of lysosomal hybition
MAPK
of AP-1
kinase
(Bosnar
(ERK,etJNK,
al., 2011;
p38) Meyer
activity,etwith
al., 2009)
subsequent
(see Section
modulatory
4.2.4)efsuggest
that azithromycin
may increase
also modulate
Th2 cell responses
activity by
in broblasts
(Gerbaux et al., 1996)
the early
andfects
it inhibits
on NFB
arachidonate
and AP-1release
transcription
and metabolism
factor activities.
in LPS-stimulated
Crosstalk between
in drolase
vivo, probably
effects on antigen-presenting
cellsand
(Table
3). In-release
and prolonged
depletion
circulatingcould
neutrophil
enzymes
macrophages
ERK and p38
(Banjanac
was suggested
et al., 2012).
to cause
Azithromycin
mutually counteracting
increases phagoeffects.
duction
of a modulatory
DCofphenotype
explainlysosomal
ndings in
healthy
after volunteers
azithromycin
of human
volunteers
(Culic et
al.,IL-2
2002).
cytosis,
Azithromycin
probably and
by upregulating
to a less extent
the other
macrophage
macrolide
mannose
antibiotics,
receptor,
interacts
human
thattreatment
azithromycin
enhanced
proliferative
and
In
this
context,
inhibition
of
neutrophil
lysosomal
enzyme
release
CD206
with (Hodge
phospholipids
et al., 2008)
with pronounced
and makes lysosomes
effects on lysosomal
in macrophages
function and
receptor responses of blood mononuclear cells (MNC) to pokeweed by
neutrophils
was the
rst
anti-inammatory
more
these
resistant
must be
to oxidant
incorporated
challenge
into any
(Persson
consideration
et al., 2012).
of theAzithromycin
intracellular
mitogen
(Tomazic
et al.,
1993).
An inhibitoryeffect
effectof
onazithromycin
antigenever to be by
described
(Carevic
& Djokic,
1988).
alsoeffects
attenuates
of azithromycin.
T helper-1 cell (Th-1) responses following LPS or
presentation
DCs almost
certainly
explains
the Interestingly,
reduction by Niemann
Pick
C2
protein
(NPC2),
which
transports
cholesterol,
glycolipids and
interferon- (IFN-) stimulation of macrophages, shifting polarization
azithromycin pretreatment of the serum titre of specic anti-PCV7,
lysophosphatidic
acid immunized
out of lysosomes,
is also transported
to endo/
of activated macrophages towards the alternative/anti-inammatory
IgG1
antibodies in mice
with polyvalent,
polysaccharide,
lysosomes from
the Golgi
apparatus
by (Fernandez
MPR300 (Willenborg
et al.,
M2-phenotype, which plays a role in directing Th-2 responses and coorpneumococcal
conjugate
vaccine
(PCV7)
et al., 2004).
This
2005),also
so azithromycin
potentially
affect extra-lysosomal
lipid
dination of repair following inammation (Murphy et al., 2008a;
nding
indicates that could
treatment
of individuals
with azithromycin
transport.
In
fact,
expression
of
both
NPC1
and
NPC2
and
of
various
Yamauchi et al., 2009). Enhancement of M2-like macrophage generashould be avoided when an immunization procedure is planned.
Interactions
with
phospholipids
lysosomes of mice with
lipid
and cholesterol
metabolism
in human
cells was
tion4.2.1.
is observed
in vivo
after
azithromycinand
pretreatment
In human
natural killer
(NK) cellsgenes
stimulated
withepithelial
IL-15, azithroAzithromycin
and
other
macrolides
readily
penetrate
into
phosphoincreased
by incubation
with azithromycin
(Ribeiro et al., 2009).
Since
a cystic brosis-like disease (Legssyer et al., 2006).
mycin
inhibited
cytotoxic function
through down-regulation
of perforin
lipidmodulation
bilayers, the
of the desosamine
being inserted into has
the hydrolysophosphingolipids,
like azithromycin
Sections
4.1.2 and
4.3.5),
The
byN-3
azithromycin
of monocytes/macrophages
expression
(Lin et al., 2012).
Inammatory (see
cytokine
production
was
not
phobic
domain
andinthe
macrocycle
occupying
a position
at the
modify but
macrophage
differentiation
et al.,
2011), they
been
studied
in detail
classically
activated
human
blood monocytes
inhibited,
it was in the
transformed(Yamamoto
NK cell line,
NK-92.
hydrophobichydrophilic
interface
(Montenez
et
al.,
1996,
1999).
could
mediate
the
actions
of
azithromycin.
primed with IFN- and activated with LPS (Vrancic et al., 2012).
It should be noted that the pleiotropic effects of azithromycin are not
For azithromycin,
this position allows themanner,
positively-charged
Azithromycin,
in a concentration-dependent
distinctivelyamino
limited to a single anatomic site or organ or by the presence of microbes.
group
in
the
macrocycle
to
neutralize
the
negatively
charged
groups
inhibited the gene expression and/or release of M1 macrophage
For instance, azithromycin, like dexamethasone, inhibits the production
in the(CCR7,
phosphate
head ofand
theIL-12p70),
acidic phospholipids,
including
markers
CXCL11
but not TNF
or IL-6phosand enof the acute phase protein serum amyloid-A (SAA) during sterile inamphatidylinositol
(PI),
preferentially
found
at
the
inner
surface
of
the
hanced expression and/or release of M2 macrophage markers (IL-10
mation in mice (Glojnaric et al., 2007). SAA, like C-reactive protein
cell
membrane
bilayer
(Montenez
et
al.,
1999).
These
negatively
and CCL18), and the pan-monocyte marker CD163. Modulation of the
(CRP), is a pentraxin synthesized by hepatocytes after IL-1 or IL-6
charged
phospholipids
bind
signaling
proteins
to
the
inner
plasmaToll-like receptor (TLR) 4 signalling pathway, NF-kB and signal transstimulation, and can be inhibited indirectly by azithromycin through relemma
and
regulate
many
cellular
functions,
including
phagocytosis
ducer and activator of transcription-1 (STAT-1) activation were shown
duction of cytokine release at the inamed site. Nevertheless, given the
(Yeung
&
Grinstein,
2007).
Consequently,
neutralization
by
azithroto be involved. The inhibitory prole of azithromycin was similar to
strong accumulation of azithromycin in hepatocyte lysosomes, it may
mycin
of
the
negative
charge
could
conceivably
initiate
many
cellular
that of the lysosomotropic drug chloroquine, indicating that lysosomal
also directly inhibit hepatic pentraxin and cyto-/chemokine synthesis
effects.
4.2.2. Autophagy
accumulation of the macrolide played a crucial role (see Section 4.2.1).
(Matijasic
et al., 2012).
PI
contains
high
amounts
of
arachidonic
acid,
and
in
LPS-stimulated
Autophagy is a process whereby ubiquitin-tagged or HspIn dendritic cells, azithromycin modulates differentiation and LPSmacrophages,
azithromycin,
like
an
inhibitor
of
cytoplasmic
phospholichaperoned, often damaged, cytosolic components or intracellular
induced maturation towards a regulatory phenotype (e.g. enhanced
pasewith
A2 (cPLA2),
arachidonate
release and
eicosanoid
pathogens are engulfed in autophagosomes and transported to
IL-10)
increased inhibited
phagocytic
capacity (Polancec
et al.,
2012; production
but
not
cPLA2
expression,
probably
preventing
cPLA2
access
to
its
lysosomes for degradation (Randow & Munz, 2012). Fusion of
Sugiyama et al., 2007). Furthermore, azithromycin inhibits expression
membrane
substrates
(Banjanac
et
al.,
2012).
Reported
inhibition
by
autophagosomes with lysosomes is mediated by the delipidated
of co-stimulatory (CD40 and CD86) and major histocompatibility
macrolide antibiotics, including azithromycin, of arachidonate release
autophagy protein, microtubule-associated protein 1A/1B-light
and metabolism in LPS-stimulated neutrophils and mononuclear cells
chain 3 (LC3), which binds to phospholipids from apopototic parti(Miyazaki et al., 2003; Sato et al., 2007) was probably due to effects on
cles phagocytosed by leukocytes, thereby facilitating the engulfsignaling further downstream from TLR4.
ment of the resulting efferocytotic vesicles by lysosomes (Florey
Interaction of azithromycin with phospholipid head groups
& Overholtzer, 2012). In smooth muscle cells (SMC), azithromycin,
decreases their mobility and the uidity and elasticity of the plasma
at a clinically relevant concentration (10 M), induced autophagy
4.2.
Cell
signaling
processes
membrane (Fa et al., 2007; Tyteca et al., 2003), accounting for many
and
inhibited
actively
proliferating cells (Stamatiou et al., 2009).
effects of the macrolide on cellular functions. These include altered
This autophagy-mediated inhibition of SMC proliferation may
membrane insertion of markers into macrophage membranes and
contribute
to respiratory
effects of azithromycin.
The
mechanisms
of the immunomodulatory
actions of macrolide
slowing of their trafcking into lysosomes; inhibition of uid phase enIn contrast,
in broblasts
and human
macrophages,
azithromycin
antibiotics,
including
azithromycin
have been
reviewed previously
docytosis, but not phagocytosis, of macro-molecules; down-regulation
blocks autophagy, measured by accumulation of lipidated LC3 (LC3-II),
and delayed recycling of surface transferrin receptors, but not Fc reby reducing acidication of autophagosomes and lysosomes (Renna
ceptors (Tyteca et al., 2002, 2003). Moreover, anti-inammatory effects
et al., 2011). Particle-induced generation of TNF and intraTable
of3a range of synthetic macrolide derivatives on macrophages were
cellular mycobacterial killing by the macrophages was inhibited. In
Recent
studies onwith
the anti-inammatory/immunomodulatory
effects of azithromycin
in animal
correlated
their phospholipid binding properties
and number
of models in vivo.
cystic brosis (CF) patients treated for 6 months with azithromycin,
positively charged centers (Munic et al., 2011).
mycobacterial killing was also reduced and LC3-II increased (Renna
Reference
Model
Azithromycin dosing
Variables changed
Extensive lysosomal accumulation of azithromycin
results from
et al., 2011). The clinical signicance of this nding is questionable
proton-driven
concentration
of
its
dicationic
form
in
the
acidic
(pH
5)
lungstudy
leukocytes,
inammatory
cytokines,
on chronic
azithromycin
(Tsai et al., 2004)
Murine P.aeruginosa lung infection
s.c. from day 3 because, in a nested casecontrol
lysosomal milieu. Here, azithromycin, like other cationic amphiphilic
counts
treatment in a total of 27,112 not
CFbacterial
patients,
the longer the treatment
lung leukocytes, MPO,
drugs (CAD), inhibits lysosomal phospholipase A1 by blocking phoswith azithromycin, the less likely
were patients to have non(Legssyer et al., 2006)
CFTR-mutated mouse P.aeruginosa
p.o. 4 wks
lung M2-like macrophages
pholipid substrate access, causing phospholipidosis,
(Binder
et inammatory
al., 2013). cytokines,
lung infection the accumulation
Pretreatment tuberculous mycobacterial infections
lung bacterial
counts,
of phospholipids in lysosomes (Kosol et al.,
2012;
Van
Bambeke
et
al.,
Murine P.aeruginosa lung infection
s.c. from days 1 to Azithromycin
5
survival, lung
effects on autophagy
mayefferocytotic
depend onmacrophages
the intracel(Tsai
et al.,Phospholipidosis,
2009)
1996).
in turn, correlates with phospholipid binding

mortality,
lung
leukocytes,
lular site of action. Accumulation of azithromycin ininammatory
muscle cells is
cytokines, not bacterial counts
by a range of macrolide derivatives (Munic et al., 2011).
many-fold less than in broblasts
leukocytes,
in which
accumu lungand
leukocytes,
inammatory
cytokines,
not viral load
(Yamada
et al., 2013)
Murine
MDR for
A.baumanii
pneumonia
s.c. from days 1 to 6
Annother
mechanism of action may also
account
phospholation is predominantly lysosomal
(Matzneller et al., 2013). Effects
survival
lipidosis. Following phosphorylation of their mannose residues in the
lung leukocytes,
inammatory
cytokines,
on SMC, thus, may involve surface
membrane
effects, while
in the
(Beigelman
et al., 2010)lysosomal enzymes are transported
Murine Sendai to
virus
pneumonia by
s.c. from days 1 to 7
Golgi apparatus,
endosomes
other two cells, azithromycin may target predominantly lysosomal
(Tong
et al., 2011)
Murine Bacillus
pyocyaneus
shock
1 h before
the multifunctional
mannose-6-phosphate/insulin
growth
receptor
II
(Geudens et al., 2008)
Murine lung ischemiareperfusion
p.o. 14 days degradation processes. Macrolides bind with low afnity to
receptor, MPR300 (Kornfeld, 1992). Here,
the
acidic
endosome
environinjury
Pretreatment valosin-containing protein (VCP), a crucial mediator of intracellular
ment causes enzyme release and transfer Neonatal
to lysosomes,
while
MPR300
mouse lung
hypoxia
From days 1 to protein
10
degradation and autophagy (Ju & Weihl, 2010; Nujic et al.,
is returned
to the Golgi apparatus. All phospholipidosis-inducing
CAD
mortality,
lung leukocytes, inammatory 2012b; Tresse et al., 2010) which may account for some long-term
(Ballard
et al., 2007)
cytokines,
tested, including erythromycin, prevent MPR300 recycling from the
effects of azithromycin on autophagy.
(Terao et al., 2003)Murine i.p. LPS inammationi.g. 20 min lung NO, plasma NO & inammatory cytokines,
endosomes to the Golgi apparatus, causing lysosomal depletion and exRecently, azithromycin, clarithromycin and erythromycin
(Hao et al., 2013)Pretreatment plasma kynurenine & inammatory cytokines,
tracellular
secretion
of lysosomal
enzymes
(Ikeda30etminal.,
2008).
Such
an mortality
(Ivetic
Tkalcevic
et al., 2006)Murine
i.p. or i.v.
LPS shockp.o.
before
plasma
TNF,
were found to sensitize to cytotoxic and pro-apoptotic effects
(Bosnar et al., 2009; Bosnar et al., 2011) Murine i.n. LPS lung inammationp.o./i.p.4 h lung leukocytes, inammatory cytokines
of bortezomib by blocking autophagic ux in myeloma cells
PretreatmentIL-1 from alveolar macrophages
(Moriya et al., 2013), suggesting a potential use for macrolides
(Fernandez-Robredo et al., 2013)Rat subconjunctival LPS3 days topical pretreatment ocular inammation
in oncology.
(Glojnaric et al., 2007)Murine sterile s.c. AgNO3 inammation p.o. from 1 to 3 days plasma SAA & inammatory cytokines,
(Beigelman et al., 2009)Murine OVA asthmas.c. from 1 day before or after challenge lung inammation, BAL leucocytes & Th2 cytokines
(Ivetic Tkalcevic et al., 2012)Murine acute OXA skin DTHTopical after challenge ear edema, leukocytes, IL-4
Murine chronic skin DTH, repeated OXA Topical after each challenge ear mast cells, IgE, IL-4, not leukocytes,
(Plesko et al., 2010)Murine DNFB colon DTHFrom before or after challenge colitis
(Iwamoto et al., 2013)Murine allogeneic GvH diseasep.o. from day 2 to 2 graft resistance, due to effect on DCs?
(Fernandez et al., 2004)Murine immunization to pneumococcal i.p. 1 h before specic anti-PCV7 IgG1 antibodies
vaccine PCV7

CFTR: cystic brosis transmembrane conduction regulatory protein; DNFB: dinitrouorobenzene; GvH: graft versus host disease to transplanted bone marrow; i.g.: intragastric; i.n.:
intranasal; i.p.: intraperitoneal; i.v.: intravenous; MDR: multidrug resistant; MPO: myeloperoxidase; OVA: ovalbumin; OXA: oxazolone; p.o.: oral; s.c.: subcutaneous.

231

232

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

4.2.3. Apoptosis
Apoptosis is a crucial non-inammatory process for terminating the
life of cells, particularly immunocompetent cells. Interactions exist between apoptosis and autophagy in that the proteins, autophagy protein
5 (Atg5) and autophagy inducing protein, Beclin-1 are involved in both
processes (Fimia & Piacentini, 2010). It is, therefore, not surprising that
effects of azithromycin have been observed on apoptosis.
In SMC, azithromycin initially induces autophagy (Stamatiou et al.,
2009) and then apoptosis after prolonged incubation (Stamatiou et al.,
2010). At concentrations of 100 M or higher, azithromycin induces
apoptosis in neutrophils (Koch et al., 2000), an action shared by many
antibiotics (Healy et al., 2002); in human lymphocytes by inhibiting expression of Bcl-xL (Ishimatsu et al., 2004; Mizunoe et al., 2004); and in
NK cells (Lin et al., 2012). Induction of apoptosis is thus related to
prolonged, extensive accumulation, particularly at high doses, as seen
with circulating human neutrophils (Culic et al., 2002). This likelihood
is supported by studies on monocytes and macrophages.
Despite accumulating several hundred-fold in macrophages,
azithromycin does not readily induce apoptosis in these cells. To the
contrary, in human bronchial alveolar lavage (BAL) macrophages, lysosomal membrane permeabilization and ROS-induced cell death were
inhibited by long-term azithromycin treatment, an action conrmed
in vitro (Persson et al., 2012). Macrophages from azithromycin-treated
patients, in fact, show enhanced ability to phagocytose apoptotic cells.
First observed in patients with chronic obstructive pulmonary disease
(COPD), azithromycin in vitro restored to normal the reduced ability
of alveolar macrophages from COPD patients to phagocytose apoptotic
neutrophils (Hodge et al., 2006). Administered for 12 weeks to COPD
patients, the macrolide also enhanced alveolar macrophage efferocytotic capacity and mannose receptor (CD 206) expression, together
with a decrease in epithelial cell apoptosis (Hodge & Reynolds, 2012;
Hodge et al., 2008). This enhancement of macrophage efferocytosis is
now attributed to the ability of azithromycin to promote the expression
of the M2 macrophage phenotype (Section 4.1.2).

4.2.4. MAP kinases and AP-1


Kanoh and Rubin (2010) stated in their review thatthere have been
so many different reported effects of macrolides on cell signaling pathways that attempts to put these actions together as one mechanism is
difcult. In fact, all macrolides seem not to have similar ranges or degrees of activity. In a majority of the reports, clarithromycin was the
macrolide tested, so it is important to consider azithromycin separately.
The clearest indication for involvement of the Erk1/2 MAP kinase
signalling pathway has been obtained from studies on mucin production by airway epithelial cells. In these cells, azithromycin inhibited
mucin 5 AC (MUC5AC) secretion and the increase in Erk1/2 phosphorylation induced by some, but not all stimuli (Ishimotoet al., 2009; Luo
et al., 2011; Morinaga et al., 2009). Subsequent transcription of the
MUC5AC gene, in H. inuenzae-or EGF-stimulated cells, was inhibited
by azithromycin through inhibition of AP-1 activation (Araki et al.,
2010; Nie et al., 2012). The same signaling pathway is linked with inhibitory effects of azithromycin on IL-8 production in epithelial cells
(Srivastava et al., 2011) and on human neutrophil chemotaxis (Tsai
et al., 2004).
Subsequent inhibition of AP-1 binding to DNA by azithromycin has
been associated with inhibition of IL-8 production in CF airway cells
(Cigana et al., 2006); inhibition of proliferation in human vascular
SMCs, (Miller et al., 2000); suppression of IL-12p40 expression and
generation by M1-type macrophages (Yamauchi et al., 2009); and
inhibition of IL-1 production in LPS-stimulated macrophages (Bosnar
et al., 2011). Clearly, for inhibition of mucin and cytokine production
and of neutrophil chemotaxis, the ERK1/2, AP-1 signaling pathway is
an important target of azithromycin. The effect on AP-1 binding, though,
is probably indirect as basal, unstimulated AP-1 binding and IL-8
production is not affected by azithromycin (Cigana et al., 2006; Miller
et al., 2000).

4.2.5. NFB pathway


In no study on MUC5AC secretion by airway epithelial cells was an inhibitory effect of azithromycin observed on NFB activation, despite
clear NFB activation by all stimuli (Araki et al., 2010; Morinaga et al.,
2009; Nie et al., 2012). However, apart from CF cells, a clear correlation
exists, in epithelial cells, between inhibition of IL-8 generation, NFB activation and DNA binding (Cigana et al., 2006; Matsumura et al., 2011;
Saint-Criq et al., 2012). This (indirect) NFB inhibition involves an
action on the receptor signal-transmitting Rho GTPase, Ras-related C3
botulinum toxin substrate 1 (Rac-1) (Matsumura et al., 2011). Thus,
while azithromycin acts on epithelial mucin by targeting AP-1, inhibition of IL-8 involves both a direct action on AP-1 and indirect on NFB
activation, apparently preceded by inhibition of membrane Rac-1,
supporting an action of azithromycin close to the phospholipid cell
membrane.
It is unclear whether NFB is a potential target for azithromycin inhibition of leukocyte cytokine production (Aghai et al., 2007; Ikegaya
et al., 2009). Several macrolide antibiotics, though, inhibit NFB activation in a variety of cells in vitro (Kanoh & Rubin, 2010), particularly
when the incubation period is long (Shinkai et al., 2008) and upstream
signaling has occurred. Moreover, in classically activated human monocytes, the macrolide only reduced NFB activation at relatively high
concentrations (50 M) (Vrancic et al., 2012). Since both AP-1 and
NFB are both dependent on upstream signaling processes, many of
the observations on their inhibition by azithromycin may be secondary
to direct cell membrane effects of the macrolide (see Section 4.2.1).

4.2.6. Gene expression


An overview of changes induced by azithromycin, at clinically relevant concentrations, was gained from human airway epithelial cells
stimulated with CF lung supernatant (Ribeiro et al., 2009). In conrmation of other studies, genes for mucin production, MMPs, CXCL3, CXCL6,
IL-7, tissue plasminogen activator (tPA) and cell cycle were downregulated. Genes for CXCR7, IL-1ra, IL-6 signal transducer, transferrin
receptor (CD71) and VEGF were upregulated. IL-8 expression and secretion was increased only by short-term (6 h) incubation. In keeping
with effects of azithromycin on lipid metabolism, the expression of
many proteins regulating lipid and cholesterol metabolism were upregulated. Prominent among these were SREBP1 (sterol regulatory element binding protein 1) and SREBP2, master activating transcription
factors which control the expression of many other genes for cholesterol
metabolism. Effects of azithromycin on membrane phospholipids
and SREBP1/2 may be linked, SREBP1 phosphorylation and downregulation of cell cycle genes being regulated by effects on MAP kinases.
In summary, direct effects of azithromycin on charged phospholipds
in plasma and lysosomal membranes seem to be initial targets of the antibiotic, with associated changes in the expression of lipid metabolising
genes, such as SREBP, autophagy and apoptosis. Effects on Rac-1, MAPK
kinases and other enzymes activated by membrane changes probably
determine downstream effects on transcription factors AP-1 and NFB,
depending on the cell type and membrane stimulus (Fig. 1). Additional
direct effects of azithromycin on signaling molecules cannot be ruled
out. However, only valosin containing protein (VCP), a protein involved
in autophagy, has been identied as a low afnity (KD = 1.8 104 M)
azithromycin binding protein (Nujic et al., 2012b). No high afnity binding proteins for azithromycin were detected.

4.3. Role of state and phase of inammation


Results of several investigations indicate that the immunomodulatory effects of azithromycin and other macrolide antibiotics vary according to the presence and phase of an inammatory response.

4.3.1. Distinction between non-inamed and inammatory conditions


The activation state of immune cells determines their response
to azithromycin. Thus, the macrolide antibiotics, as a class, stimulate

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

neutrophil and macrophage responses in healthy rodents, but generally


inhibit inammation and leukocyte responses in experimental inammatory models (Culic et al., 2001). This dichotomy was clearly seen in
a study in which pretreatment with azithromycin had no effect on
lung inammatory cytokines, but increased IL-10 concentrations in
healthy mice, while in mutant 508 CF mice, azithromycin inhibited
lung inammation and pro-inammatory cytokines, but had no effect
on IL-10 (Legssyer et al., 2006). This may have been due to effects on
predominantly resident macrophages in healthy and M1-like macrophages in inamed CF mice.
This difference between healthy and inamed conditions is also seen
in humans. Standard dosing of healthy volunteers with azithromycin for
3 days stimulated blood neutrophil degranulation and oxidative burst,
but not in similarly treated COPD patients (Culic et al., 2002; Parnham
et al., 2005). In untreated endothelial cells, azithromycin increases
(Millrose et al., 2009), whereas in LPS-stimulated epithelial cells or neutrophils, macrolides suppressed adhesion molecule expression (Khair
et al., 1995; Lin et al., 2000). It seems likely that administered early during a bacterial infection, azithromycin could promote host defense by
activating nave leukocytes and endothelial cells, unprimed by cytokines. At a later stage, when cells are activated by inammatory stimuli,
immunomodulation by azithromycin would lead to inhibition of inammation and promotion of its resolution (Parnham, 2005).

4.3.2. Time dependency of effects


The concept of a biphasic, time-dependent immunomodulatory
action of azithromycin and other macrolide antibiotics is supported by
data in several publications. Initial stimulatory and later enhancing
effects have been observed with clarithromycin on IL-8 production
and Erk1/2 activation in bronchial epithelial cells (Shinkai et al., 2006)
and with erythromycin in vitro or roxithromycin given to mice on
mononuclear cell IL-1 and IL-2 production (Kita et al., 1990; Konno
et al., 1992). Similarly, in Candida albicans infected mice and in healthy
human volunteers, a merely transient stimulation of neutrophil chemotaxis was seen with erythromycin (Fernandes et al., 1984), a nding also
obtained with azithromycin in healthy human volunteers (Culic et al.,
2002). In the latter study, initially enhanced blood neutrophil activation,
2.5 h after azithromycin, changed to a signicant reduction after
128 days.
Early, mainly stimulatory effects of azithromycin, especially on
neutrophils, are probably mediated by interactions with phospholipids
and Erk1/2, followed by later modulation of transcription factors AP-1
and NFB. Subsequent delayed inhibitory effects on cell function are
likely related to lysosomal accumulation of the macrolide, with disruption of protein and lipid transport through the Golgi apparatus and ultimate effects on surface receptor expression, including macrophage
phenotype changes and autophagy (see Section 4.2) (Fig. 2).

4.3.3. Resolution of inammation


Since azithromycin can drive the generation of an anti-inammatory
M2 macrophage phenotype (Section 4.1.2), it is pertinent that it also
promotes the resolution of experimental inammation. In 508 CF
mice with LPS lung inammation, azithromycin pretreatment inhibited
inammation 48 h after LPS, when lung IL-10 levels were increasing, i.e.
during the resolution phase (Legssyer et al., 2006). This effect on the
resolution process was further claried in murine, self-limiting,
zymosan-induced, peritoneal inammatory cell inltration, which
peaks at 48 h and is inhibited by azithromycin pretreatment (NavarroXavier,et al., 2010). Administered at 72 h, azithromycin inhibited
remaining leukocyte inltration and also enhanced the ratio of M2/M1
macrophages in the peritoneal exudate. In contrast to other antiinammatory drugs, azithromycin did not decrease the modulatory
cytokine, IL-10, in exudates. These data strengthen the proposal that
apart from acute anti-inammatory effects on neutrophils, the major
target for the immunomodulatory effects of azithromycin is the

mononuclear phagocyte, promoting an inammation-resolving M2


macrophage phenotype or a regulatory DC phenotype.
5. Clinical applications in
respiratory infections and airway inammation
The unique imunomodulatory properties of azithromycin, its broad
antibacterial spectrum and exceptional pharmacokinetics, resulting
in extensive and sustained tissue penetration, largely account for its
disease-modifying effects in infectious or non-infectious inammatory
airways diseases. These coincident properties also explain its indication
for upper and lower respiratory tract infections (e.g. acute bacterial
sinusitis, community-acquired pneumonia), in which macrolides exert
therapeutic benets not solely explainable by antibacterial activities
(Amsden, 2005).

5.1. Upper and lower respiratory tract infections


With a broad antibacterial spectrum and oral bioavailability,
macrolides are the second most commonly used antibiotic class in
many European countries (Adriaenssens et al., 2011), exceeded only
by penicillins. Based on their efcacy against the main respiratory
pathogens (S. pneumoniae, S. pyogenes), macrolides are often used for
empirical treatment of respiratory tract infections. They are also active
against other bacteria including Streptococcus pneumoniae, Mycoplasma
pneumoniae, Chlamydia spp., Legionella pneumophila, Niesseria
gonorrhoeae, Bordatella pertussis, Campylobacter jejuni, Treponema
pallidum, Ureaplasma urealyticum, Corynebacterium diphtheria and
Listeria monocytogenes. Azithromycin has an even broader spectrum,
being active against Haemophilus inuenza, Moraxella catarrhalis, nontuberculosis mycobacteria, Bartonella henselae, Rhodococcus equi,
Toxoplasma spp., Cryptosporidium spp. and Plasmodium species
(Blondeau et al., 2002). H. inuenzae, M. catarrhalis, S. pneumoniae,
as well as Chlamydophila pneumonia and M. pneumonia are particularly
susceptible. Studies in mice indicate that even with multidrug resistant bacterial strains, clinical benet may result from the immunomodulatory actions of the drug (Yamada et al., 2013).
Azithromycin does not inhibit growth of Pseudomonas aeruginosa
in vitro, but shows efcacy in vivo mainly because of inhibition of
quorum-sensing and biolm (Hoiby, 2011) (see Section 2). Given
prophylactically, azithromycin, therefore, reduces the incidence
of ventilation-associated pneumonia, particularly in patients with
rhamnolipid-positive isolates (van Delden et al., 2012), conrming
previous ndings in P. aeruginosa-induced lung inammation in mice
(Table 3). Promotion by azithromycin of macrophages with a regulatory
M2-like phenotype in animals (Feola et al., 2010; Hoffmann et al., 2007)
suggests that immunomodulation also contributes to the benet seen
clinically in P. aeruginosa infection.
Clinical experience with azithromycin in the treatment of upper
and lower respiratory tract infections has been reviewed previously
(Mulholland et al., 2012; Panpanich et al., 2008; Wierzbowski et al.,
2006). Accumulation in leukocytes at inamed sites means that
oral dosing either at 500 mg daily for 3 days or as a single dose of
11.5 mg is often sufcient.
Azithromycin is effective in microbial eradication in patients
with presumed bacterial lower respiratory tract infections (LRTI),
community-acquired pneumonia and acute bacterial sinusitis (Henry
et al., 2003; Lakos et al., 2012; Panpanich et al., 2008; Yanagihara
et al., 2009). In acute sinusitis, there are few randomized studies of
azithromycin, but the clinical and bacteriologic efcacy and safety of
azithromycin is generally good and comparable to or even slightly
better than that of amoxicillin/clavulanate (Marple et al., 2010) or
levooxacin (Murray et al., 2005). Azithromycin therapy was better
tolerated, with less frequent adverse reactions and lower cost, compared to amoxicillin/clavulanate (Henry et al., 2003; Karpov, 1999). A
common clinical nding is the coexistence of sinusitis, periodontal

233

234

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

Phospholipid
bilayer penetration

Lysosomal
enzyme &
ROS release

Inflammatory
stimulus

Neutralizes negative
PLs, reduces FA release,
decreases membrane fluidity

MAPKs (ERKs)
Lysosomal
accumulation

Increased
phagocytosis?

Rac-1?

Lipid
remodeling

Surface
molecule
recycling

Phospholipidosis

SREBP1
AP-1

Lysosome

+LC3-II

NFB

High concs

+VCP
+ MPR
Autophagy

- Bcl

Enzymes
Apoptosis

Lipid
transport

Golgi
ER

Cytokine, mucin and


inflammatory gene expression
depending on stimulus

Fig. 1. Effects of azithromycin on cell signaling. Azithromycin penetrates the cell membrane bilayer and stabilizes the membrane, reducing uidity. The cationic moiety of azithromycin
comes into close proximity to the negatively charged phospholipids in the inner leaet of the membrane, neutralising their charge. This results in reduced fatty acid release and also to
liberation of enzymes bound by electrostatic charge to the membrane. Lipid remodeling leads to modulation of signaling pathways, particularly of MAPK kinases, such as ERK1/2 (possibly
involving Rac-1) with subsequent inhibition of the activation of transcription factors including AP-1 and NFB. The signaling pathway most affected is likely to be dependent on the
particular cell, its activation state and the stimulus by which it is activated. Lysosomal enzyme release and initial induction of the oxidative burst of neutrophils are a consequence of
the actions on membrane lipids. Membrane lipid remodelling also disrupts surface molecule recycling and probably phagocytosis. Molecules dependent on negatively charged
phospholipids are also affected, including LC3-II, with subsequent inuences on lysosomal uptake and autophagy. The latter may also be inuenced, with time, by low afnity binding
of azithromycin to VCP. Azithromycin accumulates in lysosomes, modulating MPR transport of enzymes and lipids and through lipid remodeling in the lysosome membranes ultimately
leads to phospholipidosis. Apoptosis may be induced by high concentrations of azithromycin, possibly by compromising the action of the apoptosis inhibiting Bcl molecules. [ indicates
inhibition].

disease and/or bronchiectasis (Brook, 2006), which may all benet


simultaneously from azithromycin therapy (Sections 5.3 and 8.2).
Immunomodulation by macrolides has been proposed repeatedly to
contribute to decreased length of hospitalization and increased survival
in CAP and CAP-related sepsis (Amsden, 2005; Giamarellos-Bourboulis
et al., 2008; Metersky et al., 2007; Restrepo et al., 2009; Tessmer et al.,
2009). Recent meta-analyses indicate that while macrolides do decrease
mortality among hospitalized CAP patients, in RCTs or in guidelineconcordant studies a difference from other antibiotics was not evident
(Asadi et al., 2012; Asadi et al., 2013; Skalsky et al., 2013). As discussed
later for sepsis (Section 7), disease severity may determine sensitivity to
antibiotics. A recent study on 187 patients with pneumonia due to
S. pneumoniae, clearly conrmed that azithromycin signicantly
reduces mortality rate (Shorr et al., 2013). This study differed from
previous studies in being limited to a single causative agent.
Azithromycin effectively reduces respiratory symptoms in chronic
or recurrent respiratory tract infections due to so-called atypical bacteria, such as chronic Chlamydia pneumoniae or Mycoplasma pneumoniae

(Branden et al., 2004; Esposito et al., 2005; Mulholland et al., 2012).


Long-term, low-dose azithromycin, though, lacked efcacy in children
with acute respiratory syncytial virus bronchiolitis (Kneyber et al.,
2008; Pinto et al., 2012) or in chronic rhinosinusitis (Videler et al.,
2011). Nevertheless, an increasing number of small scale studies suggest that azithromycin and other macrolides are of benet in viral infections because of their immunomodulatory properties (Min & Jang,
2012). There is considerable interest, particularly in Japan, in using
macrolides to prevent the cytokine storm which occurs in patients during respiratory viral epidemics (Bermejo-Martin et al., 2009). Certainly,
azithromycin inhibits and stimulates the resolution of lung inammation in mice infected with mouse parainuenza virus type 1 (Sendai
virus) without affecting viral load (Beigelman et al., 2010). It may well
also enhance anti-viral defences, since in contrast to erythromycin and
telithromycin, azithromycin signicantly increases interferon generation and interferon-stimulated gene expression, reducing virus replication and growth in human bronchial epithelial cells infected with
rhinoviruses 16 and 1B (Gielen et al., 2010).

236

M.J.M.J.
Parnham
Parnham
et al.et/ al.
Pharmacology
/ Pharmacology
& Therapeutics
& Therapeutics
143 143
(2014)
(2014)
225245
225245

per5.2.
3 months
Diffuse
inpanbronchiolitis
comparison to a decrease of 0.10% per 3 months in the
placebo group (Altenburg et al., 2013). Macrolide resistance rate was
88% in azithromycin-treated individuals and 26% in the placebo group.
Initialwith
reports
on clinical
immunomodulation
byazithromycin
macrolides apIn children
non-CF
bronchiectasis,
once-weekly
for
peared
in the late
and 1990s,
whenexacerbations,
it became clear
the
up to
24 months
also 1980s
decreased
pulmonary
butthat
again
prognosis
of
patients
with
diffuse
panbronchiolitis
(DPB)
improved
drawith increased carriage of azithromycin-resistant bacteria (Valery
matically
implementing
treatment
with
macrolides.
et al.,
2013). after
Overall,
these RCTslong-term
conrm the
benecial
effects
with
This was rst
documented with
erythromycin,
demonstrating
signiazithromycin
or erythromycin
described
in prior, smaller,
retrospective
cant in
reduction
broncho-alveolar
lavage
(BAL)
of IL-1,
IL-8
studies
non-CF in
bronchiectasis
(Anwar
et al.,
2008;uid
Serisier
& Martin,
and Although
neutrophils
et al.,the
1996).
Sputumetiology
volumeof
in the
DPB
is reduced
2011).
not(Sakito
addressing
underlying
disease,
and lung function
by macrolides,
with marked
reduction of
azithromycin
may be improved
a therapeutic
option in patients
with frequent
neutrophil activation
lung
inltration,
of pro-inammatory
exacerbations
(Spagnoloand
et al.,
2013;
Suresh Babu
et al., 2013). cytokines and MMPs. This benet is seen despite the relatively high incidence of resistance to macrolides in Japan (Kobayashi et al., 1993;
Kudoh, 2004). Because of the established treatment of DPB with other
macrolides in Japan, azithromycin has been used less frequently, but
was recently reported to be effective and safe for long-term therapy
(Li et al., 2011).
Treatment of DPB is the most striking example of the introduction of
5.4.macrolide-therapy
Chronic obstructive
pulmonary
disease and
asthma
to chronic
respiratory
diseases.
However, the experience in DPB is almost exclusively based on non-randomized, controlled
clinical trials
(RCTs) or retrospective
studies (Kudoh
et al., antibi1998; Li et al.,
Short-term
azithromycin
has been an alternative
to rst-line
2011;
Yang
et al., 2013).
Drawing
on this of
experience
in DPB, long-term,
otics
in the
treatment
of acute
exacerbations
COPD (AECOPD)
for sevazithromycin therapy
subsequently
introduced
in CF
erallow-dose
years (Butorac-Petanjek
et al., was
2010;
Yamaya et al.,
2012). The
(Saiman
et al.,is2003),
bronchiectasis
(Anwar
2008),
benet
achieved
greaternon-CF
than with
other therapies
suchetasal.,
corticosteCOPD
(Blasi et al.,
2010; Hodge
al., 2008;
Parnham
et al., 2005),
roids
or roumilast
(Spagnolo
et al., et
2013).
Long-term
azithromycin
(Piacentini
et al.,
infectious
(Naidoo
bothasthma
decreased
frequency
of 2007),
exacerbations
andbronchiolitis
enhanced quality
of &
2009) though
and BOS
following
or hematopoietic
stem cell
lifeBryant,
in AECOPD,
with
a small lung
incidence
of hearing decrements
transplantation
(Gerhardt
et al., 2003;
et al.,
2008;toVos
et al.,
(Albert
et al., 2011).
Azithromycin
given Gottlieb
for several
months
patients
2010).
Clinical
evidenceefferocytosis
for immunomodulatory
effects and applicawith
COPD
also increased
in airway macrophages,
intions of
in inammatory
airway
is briey
dicating
an azithromycin
immunomodulatory
component
to its disorders
efcacy (Hodge
&
discussed
here based on landmark and important clinical trials.
Reynolds,
2012).
Azithromycin ameliorates bronchial hyperresponsiveness and airway neutrophil inltration in some children with asthma (Piacentini
et al., 2007), but failed to reduce rates of severe exacerbations and
LRTI in adult patients with severe asthma (Brusselle et al., 2013). Corticosteroids have limited efcacy in COPD or neutrophilic asthma and in a
subgroup analysis of patients with non-eosinophilic severe asthma,
azithromycin signicantly lowered rates of severe exacerbations and
LRTI requiring treatment with antibiotics (Brusselle et al., 2013).
Modulation of neutrophilic inammation probably accounts for these
ndings in asthma since azithromycin inhibits airway inammation in
a non-infectious, ovalbumin-induced mouse model of allergic asthma
(Beigelman et al., 2009) (see Section 4.3.3).
Despite broad inhibitory activity in experimental studies (see
Section 4.1), efcacy of azithromycin in clinical airways inammation
is limited. Clinical bronchodilatory effects have not been reported.
Azithromycin has a place in the treatment of COPD exacerbations, but
other than possibly contributing to corticosteroid-sparing in highly
selected patients, its potential role in the treatment of asthma remains
unclear (Spagnolo et al., 2013; Suresh Babu et al., 2013).

5.5. Post-transplant bronchiolitis


In lung transplant recipients with post-transplant bronchiolitis,
azithromycin for 36 months reduced local airway inammation (cytokines, neutrophils), oxidative stress and matrix remodelling (Verleden
et al., 2006, 2011a,b). It attenuates both neutrophilia and plasma levels
of multiple pro-inammatory chemokines (Federica et al., 2011). These
ndings with azithromycin have even led to the denition of novel
phenotypes of post-transplant chronic lung allograft dysfunction,
based on factors such as severity and degree of neutrophil inltration
(Gottlieb et al., 2008; Vanaudenaerde et al., 2008a,b; Vos et al., 2010).
Interestingly, prophylactic azithromycin treatment following lung
transplantation resulted in signicantly higher chronic rejection-free
survival and lower airway neutrophilia and systemic CRP levels in
comparison to placebo (Vos et al., 2011). This again conrms that
azithromycin attenuates inammation resulting from allo- and nonallo-immunologic events in the lung. Long-term RCTs are needed to
establish the routine use of azithromycin in this condition.

Cystic
brosis conditions
and non-cystic brosis bronchiectasis
5.6.5.3.
Other
respiratory
Macrolides
As in DPB,
(i.e.pulmonary
clarithromycin)
neutrophil
appear
inltration
to modulate
predominates
cryptogenic
in
organizing
patients with
pneumonia,
CF and radiation-related
non-CF bronchiectasis
bronchiolitis
and thisobliterans
encouraged
organizing
many investigators
pneumoniato(Mann
study et
azithromycin
al., 2005; Stover
in this&indication.
Mangino, In
2005)
an RCT
and
inpulmonary
251 patients
brosis
chronically
(Wuyts
infected
et al., 2010).
with P.This
aeruginosa,
is not surprising
standard 3 day
given
azithromycin
that in these
treatment
conditions,
weekly
the for
distal
24 airways
weeks signicantly
and alveoli initially
improved
may
FEV1
be involved
and reduced
in anrisk
inammatory
of exacerbations
process.
(Saiman et al., 2003). SubIn
sequently,
summary,when
numerous
treatment
pulmonary
was extended
disorders
to 12
sharing
months,
common
FEV1 remained
innate
unchanged
inammatory
in patients
mechanisms
not infected
canwith
potentially
P. aeruginosa,
benet from
although
the exaceranti-inammatory
bation and coughand
were
immunomodulatory
signicantly decreased
properties
(Clement
of azithromycin.
et al., 2006;
Azithromycin
Saiman et al.,
may,
2010).
therefore,
While prove
pulmonary
to be function,
a new milestone
thus, only
in the
improves
treatment
on azithromycin
of chronic pulmonary
treatmentdisorders,
for up to always
6 months
taking
and not
intothereafter,
account the
reincrease
duced in
exacerbations
macrolide resistance.
and reduced
In this
S. aureus
respect,
counts,
futurepresumably
development
related
of
non-antibacterial,
to immunomodulation,
anti-inammatory
are consistent
macrolide
ndingsderivatives
(Southern may
et al.,be2011).
crucial
The FEV1
(Bosnar
evolution
et al., 2012;
is explained
Kobayashi
by aetdecrease
al., 2013;inMencarelli
FEV1 during
et al.,
the initial
2011;
inammatory
Tomaskovic
phase,
et al.,which
2013)then
(Erakovic
increases
Haber
withetdecreasing
al., in press).
inammation after azithromycin is initiated, FEV1 subsequently reaching a
steady-state or plateau phase (depending on % predicted for each
individual). Azithromycin is generally considered an important part
of the treatment armamentarium for CF, particularly in patients chronically infected with P. aeruginosa (Spagnolo et al., 2013; Suresh Babu
et al., 2013).
6. Infections
urogenital in non-CF bronchiectasis is now well
The roleof
ofthe
azithromycin
tract
and sexually-transmitted
established
with three recentinfections
RCTs. In the EMBRACE trial, compared to
placebo, azithromycin decreased event-based exacerbation rates in
adult patients with non-CF bronchiectasis, whereas changes in FEV1
Over baseline
90% of an
dose oftotal
azithromycin
is excreted
via the
hepatofrom
andoral
in SGRQ
score during
the 6-month
treatment
biliary
system
(Ballow
et al., 1998;
al.,BAT
1990;
Singlas,
1995).
period
did not
differ (Wong
et al.,Foulds
2012). et
The
trial
conrmed
that
Since
drug is found
the urine,
is
dailyvery
uselittle
of azithromycin
forunchanged
12 monthsinresults
in a azithromycin
lower rate of innotfectious
suitableexacerbations
for diseases involving
signicant
bacteruria.
Due
to
its
optibut azithromycin also increased FEV1 by 1.03%
mal systemic distribution and tissue penetration, though, azithromycin
accumulates up to 100-fold in a variety of tissues of urological interest
(Foulds et al., 1990; Foulds et al., 1991).

6.1. Urethritis
Their considerable penetration into human cells makes macrolides
particularly suited for treatment of infections caused by obligate intracellular pathogens, like Chlamydia spp. For several years, azithromycin
has been the rst-line drug for treatment of primary non-gonococcal
urethritis (www.uroweb.org/gls/pdf/15_Urological_Infections.pdf),
mainly caused, in both sexes, by Chlamydia trachomatis, Ureaplasma
urealyticum or Mycoplasma genitalium.
Essential clinical evidence was provided in 1995 for this indication
in men (Stamm et al., 1995). A single 1 g oral dose of azithromycin,
for empirical treatment of acute non-gonococcal urethritis syndrome,
was as effective as a standard 7-day course of twice-daily 100 mg doxycycline in achieving clinical cure. Microbiological and overall clinical
cure rates with azithromycin were 83% and 81%, respectively (Stamm
et al., 1995).
A meta-analysis of RCTs conrmed the therapeutic equivalence of
the two regimens for chlamydial urethritis in men and women (Lau &
Qureshi, 2002). However, patient compliance to doxycycline for 7
days is often hampered by gastrointestinal side effects (Thorpe et al.,
1996). Since poor compliance is detrimental to resolution of the infection and may increase resistance, in our opinion, the 100% compliance
with a single-dose azithromycin regimen may minimize such risks.
Whereas azithromycin and doxycycline are equally active against
chlamydial infections, the macrolide achieved higher rates of eradication and clinical remission in patients with non-gonococcal urethritis
(NGU) caused by Mycoplasma genitalium (Falk et al., 2003; Mena et al.,
2009).
In women, chlamydial cervicitis/urethritis is highly prevalent, and
may occur concurrently with onset of preterm labor, preterm rupture
of membranes, spontaneous abortion, fetal death, postpartum endometritis, salpingitis, with risks to the neonate (Pitsouni et al., 2007). A single 1 g dose of azithromycin eradicates the infection in over 95% of cases
and is equivalent in terms of efcacy and symptom resolution to oneweek, once-daily 100 mg doxycycline (Thorpe et al., 1996). A metaanalysis (Pitsouni et al., 2007), involving 587 pregnant women, demonstrated high and equivalent efcacy of azithromycin, erythromycin and

Fig. 2. Proposed time course of azithromycin actions on infection and chronic inammation.

235

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

amoxicillin against chlamydial cervicitis/urethritis. However, azithromycin was associated with fewer gastrointestinal adverse events,
fewer total adverse events, and better compliance.

6.2. Prostatitis
Chronic bacterial prostatitis (CBP) is a difcult-to-eradicate biolm
disease, dramatically affecting quality of life and requiring long-term
antibacterial therapy for weeks or even months. Eradication of causative
pathogens is hampered by poor penetration of most antibacterial agents
into the prostate gland. Fluoroquinolones are currently the agents of
choice, due to adequate prostatic penetration, but recent RCTs show
that macrolides are becoming a primary therapeutic option for CBP
(Kolumbic Lakos et al., 2011; Magri et al., 2007, 2010; Skerk et al.,
2002, 2003, 2004, 2006). The rationale is the excellent penetration
and high concentration of this macrolide (see Sections 3.1 and 3.2) in
prostatic tissue and secretions, together with its powerful anti-biolm
and immunomodulatory activities. Consequently, azithromycin is
recommended as a therapeutic option for CBP in specic cases by
the European Urological Association (www.uroweb.org/gls/pdf/15_
Urological_Infections.pdf). The clinical and preclinical evidence
supporting azithromycin as a valuable treatment option for CBP has
been reviewed in detail (Perletti et al., 2011).

6.3. Other sexually transmitted diseases (STDs)


Macrolides and azithromycin in particular, are used to treat a number of STDs including syphilis, granuloma inguinale (Donovanosis)
and chancroid. In the latter two diseases, azithromycin is recommended
as rst-line therapy (http://www.cdc.gov/std/treatment/2010/toc.
htm).

6.4. Glomerulonephritis
Crescentic glomerulonephritis is a severe renal complication of
infectious endocarditis, frequently caused by Gram-positive cocci or
Gram-negative Bartonella species (Kannan & Mattoo, 2001). The condition is not a urinary tract infection per se, but rather derives from immune activation by bacterial antigens. Bartonella endocarditis immune
complexes are deposited in the glomeruli cause necrotizing crescentic
glomerulonephritis. Interestingly, this form of glomerulonephritis may
improve with antibiotic therapy (Bookman et al., 2004). Resolution of
glomerulonephritis and normalization of serum creatinine levels was
achieved in several cases by aggressive therapy with azithromycin and
a parenteral cephalosporin (ceftriaxone) or by tobramycin/doxycycline
combination (Bookman et al., 2004). The efcacy of azithromycin
against facultative intracellular Bartonella may be explained by good
intracellular penetration of the macrolide (Massei et al., 2005).

7. Clinical application in sepsis


Sepsis is an overwhelming inammatory host response to a bacterial
stimulus. Several animal studies have shown a signicant antiinammatory effect of azithromycin (Table 3) or clarithromycin in experimental sepsis. Pretreatment of mice with azithromycin improved
survival and attenuated plasma levels of TNF in LPS-induced shock
(Ivetic Tkalcevic et al., 2006; Tong et al., 2011) and additional immunomodulatory effects have been described for clarithromycin. Administered i.v. immediately after cecal ligation and puncture in rabbits,
clarithromycin reduced apoptosis in circulating lymphocytes and
production of TNF by circulating monocytes, especially within the
rst 4 h after treatment (Atmatzidis et al., 2011). Pretreatment with
clarithromycin, but not levooxacin, also inhibited lung inammation
and adhesion molecule expression due to high pressure ventilation in
mice (Amado-Rodriguez et al., 2013). Similar clinically relevant studies
on azithromycin have yet to be performed.

It is estimated that severe sepsis and septic shock, mainly due to CAP,
annually affect 377 of 100,000 members of the general population;
mortality is 3550% making sepsis a leading cause of death, more common than malignancies and heart disorders (Kotsaki & GiamarellosBourboulis, 2012). Current guidelines recommend intravenous antibiotic treatment as early as possible, because with most antibiotic classes,
effectiveness decreases with time and sepsis severity (Dellinger et al.,
2013). Several retrospective analyses of prospectively collected data
have assessed the outcome of monotherapy over combination therapy
of severe CAP. The primary endpoint of these analyses was whether
inclusion of a macrolide (azithromycin, clarithromycin or erythromycin,
where recorded) in the combination affects mortality. The summary of
the seven studies published after 2006 is shown in Table 4. Four studies
indicated a signicant decrease in risk of death with the intake of one
macrolide (Martin-Loeches et al., 2010; Metersky et al., 2007; Restrepo
et al., 2009; Tessmer et al., 2009); two showed a signicant decrease in
duration of hospitalization (Ambroggio et al., 2012; Wilson et al., 2012);
and only one failed to show any benet (Karhu et al., 2013).
Two prospective, RCTs have evaluated the impact of macrolide
administration on the outcome of severe infections. In both trials, the
macrolide was clarithromycin, but in view of the striking outcomes,
they are discussed here as indicators of the potential use of other
macrolides, such as azithromycin. The hypothesis was that clarithromycin, administered at a dose of 1 g within 1 h on consecutive days,
should reach maximum serum levels (10 g/mL) sufcient to modulate
circulating monocyte function. In the rst study (GiamarellosBourboulis et al., 2008), 200 patients with ventilator-associated pneumonia (VAP) received placebo or clarithromycin for three days. Isolated
bacterial pathogens were multidrug-resistant species of Acinetobacter
baumannii, of Pseudomonas aeruginosa and of Klebsiella pneumoniae,
not included in the antimicrobial spectrum of clarithromycin. The
macrolide signicantly reduced by 5.5 days, time until resolution of
VAP (p = 0.011); by 6.5 days, time until weaning from mechanical
ventilation (p = 0.049); and the relative risk of death by septic shock
and multiple organ dysfunction (MODS; from 19.00 with placebo to
3.78 with clarithromycin; p = 0.048). No difference in incidence of
serious adverse events was seen.
Immunoparalysis develops during severe sepsis and drives towards
multiple organ dysfunction syndrome (MODS) and a lethal outcome
(Boomer et al., 2011). Crucially, clarithromycin treatment was accompanied by a reversal of immunoparalysis, most prominently 4 days
after start of treatment and among patients with septic shock and
MODS. Compared with placebo, clarithromycin decreased the serum
IL-10/TNF ratio; increased monocyte CD86 expression, a marker
of antigen-presentation efciency; and increased IL-6 production by
LPS-stimulated monocytes (Spyridaki et al., 2012). Reversal of immunoparalysis was unexpected as attenuation of inammatory responses in
patients would have been anticipated, based on the results of animal
studies with macrolides (Table 3). It seems that, like azithromycin,
clarithromycin can modulate monocyte function, depending on the
inammatory environment (see Section 4.1.2). Alternatively, these
ndings, at least in part, may represent an epiphenomenon arising
from improvement in the clinical course of sepsis following treatment.
In the second RCT study (Giamarellos-Bourboulis et al., 2013), 600
patients were enrolled with sepsis due to primary or secondary Gramnegative bacteremia, acute pyelonephritis or acute intrabdominal
infections, all with microbiologically-proven or clinically-suspected
Gram-negative bacteria insensitive to clarithromycin. Treatment was
extended to four days and the ndings of the rst RCT were conrmed.
Mortality due to septic shock and MODS was reduced by clarithromycin
to 53.6% compared to 73.1% with placebo (p = 0.020). Furthermore,
among patients with severe sepsis/shock, median time to infection
resolution was 6 days with clarithromycin and 10 days with placebo
(p = 0.037) and was accompanied by a signicant shortening of hospitalization time and reduction of costs. Treatment with clarithromycin
did not impact the safety of the patients.

237

238

M.J.M.J.
Parnham
Parnham
et al.et/ al.
Pharmacology
/ Pharmacology
& Therapeutics
& Therapeutics
143 143
(2014)
(2014)
225245
225245

Current
et al., 2013).
management
The benet
of sepsis
of long-term
remains macrolide
largely supportive.
treatmentTherapy
of DPB patargets
tients
theconsiderably
underlying infection
outweighsand
thisaims
concern,
to support
particularly
the function
since resistance
of
failing
to macrolides
organs. Allisagents
alreadydeveloped
high in Japan,
over the
where
last DPB
two decades
is demographically
that
inhibit
signicant
exaggerated
(see Section
pro-inammatory
5.2). In addition,
responses
the motilin-agonistic
have failed to prolong
effects
survival
of theorantibiotic
were even
result
harmful.
in an increased
The fact that
incidence
clarithromycin
of adverse
was
gastrointesable to
reverse
tinal the
effects
immunoparalytic
on prolonged state
treatment
arising
(Cameron
during severe
et al., sepsis,
2012). in
The
andevelRCT
opment
in patients
of novel,
withnon-antibacterial,
VAP (Spyridaki etnon-motilin
al., 2012), raises
receptor
hopes
binding,
that
macrolides,
immunomodulatory
including azithromycin,
macrolides may
mayovercome
offer a signicant
these concerns
advance in
the (Cameron
management
et al.,
of sepsis.
2012); Their
Erakovic
efcacy
Haberinetthis
al.,condition
in press).may
A further
help safety
explain
issuethe
with
benet
long-term
of macrolides
azithromycin
in patients
treatment
withisCAP.
the possible
The good
occurrence
efcacy
of azithromycin
in a small number
in severe
of patients
inammatory
of hearing
cases
defects
of post-transplantation
(Li et al., 2013).
bronchiolitis
For many
(see
years,
Section
azithromycin
5.5), suggests
has been
that there
one of
may
thebe
safest
similarities
broad
in the
spectrum
mechanisms
antibiotics.
involved
In contrast
in effects
to of
other
macrolides
macrolide
in antibiotics,
the two
not
conditions.
interacting with CYP3A4 means that it exhibits no metabolic interactions with most other commonly used drugs. It is N20-fold less potent
an inhibitor of hERG potassium channels than other macrolide antibiotics (Giudicessi & Ackerman, 2013). However, in an observational,
non-randomized study of patients enrolled into a Tennessee Medicaid
program, patients receiving azithromycin had an incidence of cardiovascular death 2.88-fold higher than those taking no antibiotic, and
8. Other clinical applications
2.49-fold higher than in patients on amoxicillin (Ray et al., 2012). Subsequently, the FDA introduced a black box warning about the potential
ofdisease
fatal arrhythmias with azithromycin, advising health profes8.1.risk
Skin
sionals to consider the risk of fatal heart rhythms with azithromycin
when considering treatment options for patients who are already at
Macrolide
antibiotics, are
widelyAused
for post-marketing
the topical treatment
risk for cardiovascular
events.
further
safetyofsurveilacne
and using
rosacea
and population-based
their use for thesehealthcare
and other databases
skin disorders
has
lance
seven
in Denmark,
been
reviewed
& Zedan,
2012).
In several among
small studies,
Italy,
and the(Alzolibani
Netherlands,
identied
azithromycin,
other medioralcations,
azithromycin
(for suspect
24 weeks)
provided
signicant
improvementacute
as a prime
for an
association
with drug-induced
in intractable
In one
healthy
volunteer-controlled
study, stanmyocardialrosacea.
infarction
(Coloma
et al.,
2013). The US observational
dard
dosing
azithromycin
4 weeks
to 17 patients
rosacea,
study
waswith
criticized,
though,for
because
Medicaid
patientswith
generally
reduced
score and
ameliorated
oxidave
burst
have ainammatory
higher comorbidity
than
patients inthe
theraised
general
population
(chemiluminescence)
of leukocytes
biopsies from
skinprospective
lesions
(Giudicessi & Ackerman,
2013). in
Importantly,
a large
study
(Bakar
et al.,to2007).
Since noDanish
microbiological
carried
in young
middle-aged
people at aanalyses
low-riskwere
of underlying
out,cardiovascular
it is unclear whether
the
effects
observed
were
due
to
antidisease revealed that the risk of cardiovascular death
inammatory
or treatment
antibacterial
rosacea
considered
due to 5-day
wasactions.
15.4 in Currently,
comparison
to 85.4isper
million
to be
a
disease
of
dysregulated
immunity.
In the absence
of 2013).
azithromycin courses in the innate
Tennessee
study (Svanstrom
et al.,
RCTs,
the
place
of
azithromycin
in
the
treatment
of
rosacea
remains
to
Thus, the risk of a cardiovascular event with azithromycin in otherwise
be established.
healthy patients is very small, but in patients at risk (i.e. idiopathic or
drug-induced baseline QT prolongation, familial long QT syndrome, or
multiple risk factors) azithromycin should be considered with care
and polypharmacy should be avoided (Giudicessi & Ackerman, 2013).

The
comparative
mechanismefcacy
of action
with
is considered
other drugs,toazithromycin
be a combination
is only
of recomantibacterial,
mendedimmunomodulatory
for use in DPB, CF,and
non-CF
biolm-inhibiting
bronchiectasiseffects,
and bronchiolitis.
associated
with Early,
the prolonged
mainly stimulatory
accumulation
effects
of azithromycin
of azithromycin
in inamed
on immune
tissues
and
(Wang,
epithelial
2010).
cells
The
areimproved
probablysafety
mediated
of azithromycin
by interactions
in comparison
with phosphoto
thelipids
current
andrecommended
Erk1/2. Later adjuvant
modulation
antibiotic
of transcription
treatmentfactors
with amoxiAP-1 and
cillin
NFB,
and is
metronidazole
accompaniedisbya further
inhibition
added
of inammatory
incentive to the
cytokine
introduction
and
of mucin
the macrolide.
release. Subsequent delayed inhibitory effects on cell function
are
Azithromycin
likely related
is widely
to the high
usedlysosomal
to treat gingival
accumulation
overgrowth,
of theamacrolide,
sideeffect
withofdisruption
cyclosporin
of protein
administration.
and lipidOnly
transport
3 RCTs
through
have the
beenGolgi
carried
apparaouttus
using
and aultimate
short treatment
effects on
period,
surface
butreceptor
clear reduction
expression,
of tissue
including
overgrowth
macrophage
was reported
phenotype
in allchanges,
studies and
anduse
autophagy.
of azithromycin
These later
for this
changes,
purpose
particularly
is recommended
on macrophage
(Hirsch
andetdendritic
al., 2012).
cell phenotype, appear to
underlie many in vivo experimental and clinical immunomodulatory
effects of azithromycin, contributing to resolution of acute infections
of the airways, genital tract and skin, as well as to reduction of exacerbation frequency in chronic airway diseases.
Immunomodulation with long-term azithromycin to reduce exacer8.3.
Ophthamology
bations
in COPD, CF and non-CF bronchiectasis patients must be
balanced against the potential for increased bacterial resistance. However, a sub-group of post-transplant bronchiolitis patients with proAzithromycin is used in ophthalmology for topical treatment of
minent neutrophil inltration appears to be particularly sensitive to
bacterial conjunctivitis and for off-label use in chronic blepharitis
long-term prophylactic therapy with azithromycin, as may be patients
(inammation of the eyelid) (Gebre et al., 2012). In the latter, applicawith severe sepsis treated acutely with the drug. Other promising indition of 1% azithromycin for 2 weeks reduced redness and swelling
cations include chronic prostatitis, rosacea and periodontitis.
and improved meibomian gland secretion (Luchs, 2010), probably
Although well-tolerated with a very good record of safety, azithrodue to immunomodulatory effects, also observed in a study of
mycin has recently been associated with rare cases of torsades des
topical azithromycin in LPS-induced ocular inammation in the rat
pointes in patients at risk of cardiac arrhymias and polypharmacy with
(Fernandez-Robredo et al., 2013).
azithromycin is generally to be avoided.

8.4. Gastric motility


Erythromycin is a stimulator of gastric motility as a consequence of
agonist activity at motilin receptors (Peeters et al., 1989). Azithromycin
similarly stimulates gastric motility and in view of its better safety prole to that of erythromycin, has been proposed as a treatment for gastric
motility
disorders
(Mertens et al., 2009; Rohof et al., 2012). Recently,
Conict
of interest
azithromycin has been shown to have similar potency to erythromycin
as an agonist at human motilin receptors, causing prolonged release of
GPP and
RVhuman
declaregastric
that they
havemuscle
no conicts
acetylcholine
from
antrum
(Broadof&interest.
Sanger,
2013).
Acknowledgments

8.2. Dentistry

MJP has collaborated with Novo Nordisk and serves as an advisor to


9. Safety
Leo Pharma A/S and Xellia Pharmaceuticals ApS. VEH is an employee of
Fildelta d.o.o. and both MJP and VEH are previous employees of PLIVA
and
GlaxoSmithKline.
has received
unrestricted
educational
grants
A
concern
about the useEGB
of macrolide
antibiotics
for long-term
treatfrom
SA (program
70/3/7447
of the University
of Athens).
ment
ofAbbott
chronicHellas
inammatory
diseases,
is the possibility
of increased
He alsoresistance,
serves as an
advisor
AbbVie SA.
bacterial
though
the to
relationship
between long-term use
and increased resistance is still uncertain (Cameron et al., 2012). A recent meta-analysis of six RCTs on the effects of long-term azithromycin
in chronic lung diseases, revealed that while bacterial colonization decreased
by 45%, there was a 2.7-fold increase in bacterial resistance (Li
References

Over the last 20 years, a number of studies, only a few them RCTs,
have reported benecial effects of azithromycin in adjuvant treatment
of periodontal disease (Bartold et al., 2013; Hirsch et al., 2012). Microbiological studies indicate that azithromycin is highly effective against
Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans
(Bartold et al., 2013).
The balance of opinion is that azithromycin relieves infection and
inammation and reduces pocket depth, particularly in aggressive
forms of periodontal disease, but further controlled studies are needed.
10. Conclusions

Adriaenssens, N., Coenen, S., Kroes, A. C., Versporten, A., Vankerckhoven, V., Muller, A.,
et al. (2011). European Surveillance of Antimicrobial Consumption (ESAC): systemic
antiviral use in Europe. J Antimicrob Chemother 66, 18971905.
As a broad-spectrum antibacterial, azithromycin shares the same
Aghai,
Z. H., Kode, A., Saslow, J. G., Nakhla, T., Farhath, S., Stahl, G. E., et al. (2007).
Table 4
mechanism
of action
as published
other macrolide
its range
of
Azithromycin
suppresses
activation of nuclear factor-kappa B and synthesis of proSummary
of retrospective
studied
after 2006 antibiotics
on the impact and
of macrolide
administration
on the outcome of community-acquired
pneumonia
(CAP).
inammatory cytokines in tracheal aspirate cells from premature infants. Pediatr
activity is extended through inhibition of bacterial quorum-sensing
Res
62,
483488.
and biolm. Accumulating more effectively
than other macrolides in
Reference
Setting
Arms (n patients)
Administered
macrolide
of macrolide
Albert, R. K., Connett,
J., Bailey, W.
C., Casaburi, R., Cooper, J. A.,Effect
Jr., Criner,
G. J., et intake
al. (2011).
cells, particularly circulating phagocytes, it is delivered in high concenAzithromycin for prevention of exacerbations of COPD. N Engl J Med 365, 689698.
-lactam
(15809)
(Ambroggio
al., 2012)
Hospitalizedfeature,
children combined with
NR
Reduction
of Schnfeld,
LOS by one day
trations toet sites
of infection. This important
Allen, N. E. (2002). Effects
of macrolide antibiotics on ribosome function.
In W.
-lactam + macrolide (4934)& H. A. Kirst (Eds.), Macrolide antibiotics (pp. 261280). Basel: among
31%Verlag.
of children
Birkhuser
the extended plasma half-life of azithromycin, often allows effective
(p b 0.0001)
Altenburg, J., de Graaff, C. S., Stienstra, Y., Sloos, J. H., van Haren,1218
E. H., yKoppers,
R. J., et al.
single-dose administration for acute bacterial infections. Cellular accuNo
difference
(2013). Effect of azithromycin maintenance treatment on infectious exacerbations
mulation,
accounting for rapid plasma
clearance
extensive tissue
-lactam + quinolone (104) among patients with
(Karhu
et al., 2013)
Severe
sepsis due and
to CAP
Azithromycin
or erythromycin
non-cystic brosis
bronchiectasis: the BAT randomized con-lactam + macrolide (106) trolled trial. JAMA 309, 12511259.
distribution, offsets the drug's moderate bioavailability.
-lactam + quinolone (54)Alzolibani, A. A., & Zedan, K. (2012). Macrolides in chronic inammatory skin disorders.
The antibiotic
are facilitated
its immuno(Martin-Loeches
et al.,effects
2010) of azithromycin
Severe sepsis
and MV dueby
to CAP
Reduction of risk of death
-lactam + macrolide (46) Mediators InammNR
2012, 159354.
modulatory actions on innate and adaptive immune responses. The
(HR:
p: 0.03)
Monotherapy (601)
Amado-Rodriguez, L., Gonzalez-Lopez, A., Lopez-Alonso, I., Aguirre,
A.,0.44,
Astudillo,
A.,
Risk of death
Combination therapy (1408)Batalla-Solis, E., et al. (2013). Anti-inammatory effects of clarithromycin
drug appears to exert a biphasic action
which may serve to promote
in after 30 days
(Metersky et al., 2007)
Bacteremic pneumonia
Azithromycin (n = 231)
with
macrolide
intake 0.61
ventilator-induced Clarithromycin
lung injury. Respir
initial host defence and later reduce bystander tissue injury and
(n =Res
65) 14, 52.
0.007)
Ambroggio, L., Taylor,
J. A., Tabb, (n
L. =P.,32)
Newschaffer, C. J., Evans, (p:
A. A.,
& Shah, S. S. (2012).
Erythromycin
promote inammation resolution. Benecial effects mediated by
Reduction
of 90-day mortality
Comparative effectiveness
of empiric beta-lactam monotherapy and
beta-lactamNR
(12.5%
vs 33.8%, p b 0.0001)
immunomodulation are also seen in a variety of chronic inammatory
macrolide combination therapy in children hospitalized with community-acquired
(Restrepo et al., 2009)
Severe sepsis due to CAP
Non-macrolide (133)
Independent predictor of lower
disorders. These include DPB, post-transplant bronchiolitis (particularly Macrolide (104)
pneumonia. J Pediatr 161, 10971103.
mortality
14 days
Amsden, G. W. (1995). Macrolides versus azalides: a drug interaction
update. at
Ann
severe inammatory forms) and rosacea. The ability of azithromycin to -lactam (908)
(OR: 0.53, p: 0.031)
Pharmacother
29,
906917.
(Tessmer
et
al.,
2009)
Hospitalized
patients
NR
-lactam + macrolide (946)
modulate host response reactions contributes to benecial effects in
Reduction
of
mean LOS
Amsden, G. W. (1996). Erythromycin, clarithromycin, and azithromycin: are the differ(15.9 days vs 24.7 days,
CF, non-CF bronchiectasis, COPD and asthma. Currently, based on
ences real? Clin Ther 18, 5672 (discussion 55).
p b 0.0001)
(Wilson et al., 2012)

Severe sepsis due to CAP

-lactam + quinolone (908)


-lactam + macrolide (946)

HR: hazard ratio; LOS: length of stay; MV: mechanical ventilation; NR: not reported; OR: odds ratio.

NR

239

240

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

Amsden, G. W. (2001). Advanced-generation macrolides: tissue-directed antibiotics. Int J


Antimicrob Agents 18(Suppl. 1), S11S15.
Amsden, G. W. (2005). Anti-inammatory effects of macrolidesan underappreciated
benet in the treatment of community-acquired respiratory tract infections and
chronic inammatory pulmonary conditions? J Antimicrob Chemother 55, 1021.
Anwar, G. A., Bourke, S. C., Afolabi, G., Middleton, P., Ward, C., & Rutherford, R. M. (2008).
Effects of long-term low-dose azithromycin in patients with non-CF bronchiectasis.
Respir Med 102, 14941496.
Araki, N., Yanagihara, K., Morinaga, Y., Yamada, K., Nakamura, S., Yamada, Y., et al. (2010).
Azithromycin inhibits nontypeable Haemophilus inuenzae-induced MUC5AC expression and secretion via inhibition of activator protein-1 in human airway epithelial cells. Eur J Pharmacol 644, 209214.
Asadi, L., Eurich, D. T., Gamble, J. M., Minhas-Sandhu, J. K., Marrie, T. J., & Majumdar, S. R.
(2013). Impact of guideline-concordant antibiotics and macrolide/beta-lactam combinations in 3203 patients hospitalized with pneumonia: prospective cohort study.
Clin Microbiol Infect 19, 257264.
Asadi, L., Sligl, W. I., Eurich, D. T., Colmers, I. N., Tjosvold, L., Marrie, T. J., et al. (2012).
Macrolide-based regimens and mortality in hospitalized patients with communityacquired pneumonia: a systematic review and meta-analysis. Clin Infect Dis 55,
371380.
Asakura, E., Nakayama, H., Sugie, M., Zhao, Y. L., Nadai, M., Kitaichi, K., et al. (2004).
Azithromycin reverses anticancer drug resistance and modies hepatobiliary excretion of doxorubicin in rats. Eur J Pharmacol 484, 333339.
Asgrimsson, V., Gudjonsson, T., Gudmundsson, G. H., & Baldursson, O. (2006). Novel effects of azithromycin on tight junction proteins in human airway epithelia.
Antimicrob Agents Chemother 50, 18051812.
Atmatzidis, S., Koutelidakis, I., Chatzimavroudis, G., Louis, K., Pistiki, A., Roditis, K., et al.
(2011). Clarithromycin modulates immune responses in experimental peritonitis.
Int J Antimicrob Agents 37, 347351.
Baciu, M., Sebai, S. C., Ces, O., Mulet, X., Clarke, J. A., Shearman, G. C., et al. (2006). Degradative transport of cationic amphiphilic drugs across phospholipid bilayers. Philos
Transact A Math Phys Eng Sci 364, 25972614.
Bakar, O., Demircay, Z., Yuksel, M., Haklar, G., & Sanisoglu, Y. (2007). The effect of
azithromycin on reactive oxygen species in rosacea. Clin Exp Dermatol 32,
197200.
Ballard, H. O., Bernard, P., Qualls, J., Everson, W., & Shook, L. A. (2007). Azithromycin protects against hyperoxic lung injury in neonatal rats. J Investig Med 55, 299305.
Ballow, C. H., Amsden, G. W., Highet, V. S., & Forrest, A. (1998). Pharmacokinetics
of oral azithromycin in serum, urine, polymorphonuclear leucocytes and inammatory vs non-inammatory skin blisters in healthy volunteers. Clin
Drug Investig 15, 159167.
Banjanac, M., Munic Kos, V., Nujic, K., Vrancic, M., Belamaric, D., Crnkovic, S., et al. (2012).
Anti-inammatory mechanism of action of azithromycin in LPS-stimulated J774A.1
cells. Pharmacol Res 66, 357362.
Bartold, P. M., du Bois, A. H., Gannon, S., Haynes, D. R., & Hirsch, R. S. (2013). Antibacterial
and immunomodulatory properties of azithromycin treatment implications for periodontitis. Inammopharmacology 21, 321338.
Beigelman, A., Gunsten, S., Mikols, C. L., Vidavsky, I., Cannon, C. L., Brody, S. L., et al. (2009).
Azithromycin attenuates airway inammation in a noninfectious mouse model of allergic asthma. Chest 136, 498506.
Beigelman, A., Mikols, C. L., Gunsten, S. P., Cannon, C. L., Brody, S. L., & Walter, M. J. (2010).
Azithromycin attenuates airway inammation in a mouse model of viral bronchiolitis. Respir Res 11, 90.
Bermejo-Martin, J. F., Kelvin, D. J., Eiros, J. M., Castrodeza, J., & Ortiz de Lejarazu, R. (2009).
Macrolides for the treatment of severe respiratory illness caused by novel H1N1
swine inuenza viral strains. J Infect Dev Ctries 3, 159161.
Binder, A. M., Adjemian, J., Olivier, K. N., & Prevots, D. R. (2013). Epidemiology of
nontuberculous mycobacterial infections and associated chronic macrolide use
among persons with cystic brosis. Am J Respir Crit Care Med 188, 807812.
Blasi, F., Bonardi, D., Aliberti, S., Tarsia, P., Confalonieri, M., Amir, O., et al. (2010). Longterm azithromycin use in patients with chronic obstructive pulmonary disease and
tracheostomy. Pulm Pharmacol Ther 23, 200207.
Blondeau, J. M., DeCarolis, E., Metzler, K. L., & Hansen, G. T. (2002). The macrolides. Expert
Opin Investig Drugs 11, 189215.
Bookman, I., Scholey, J. W., Jassal, S. V., Lajoie, G., & Herzenberg, A. M. (2004). Necrotizing
glomerulonephritis caused by Bartonella henselae endocarditis. Am J Kidney Dis 43,
e25e30.
Boomer, J. S., To, K., Chang, K. C., Takasu, O., Osborne, D. F., Walton, A. H., et al. (2011). Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306,
25942605.
Bosnar, M., Bosnjak, B., Cuzic, S., Hrvacic, B., Marjanovic, N., Glojnaric, I., et al. (2009).
Azithromycin and clarithromycin inhibit lipopolysaccharide-induced murine pulmonary neutrophilia mainly through effects on macrophage-derived granulocytemacrophage colony-stimulating factor and interleukin-1beta. J Pharmacol Exp Ther 331,
104113.
Bosnar, M., Cuzic, S., Bosnjak, B., Nujic, K., Ergovic, G., Marjanovic, N., et al. (2011).
Azithromycin inhibits macrophage interleukin-1beta production through inhibition
of activator protein-1 in lipopolysaccharide-induced murine pulmonary neutrophilia.
Int Immunopharmacol 11, 424434.
Bosnar, M., Kelneric, Z., Munic, V., Erakovic, V., & Parnham, M. J. (2005). Cellular uptake
and efux of azithromycin, erythromycin, clarithromycin, telithromycin, and
cethromycin. Antimicrob Agents Chemother 49, 23722377.
Bosnar, M., Kragol, G., Kostrun, S., Vujasinovic, I., Bosnjak, B., Bencetic Mihaljevic, V.,
et al. (2012). N-substituted-2-O,3-N-carbonimidoyl bridged macrolides: novel
anti-inammatory macrolides without antimicrobial activity. J Med Chem 55,
61116123.

Branden, E., Koyi, H., Gnarpe, J., Gnarpe, H., & Tornling, G. (2004). Intermittent
azithromycin treatment for respiratory symptoms in patients with chronic Chlamydia
pneumoniae infection. Scand J Infect Dis 36, 811816.
Bright, G.M. & Hauske, J.R. (1984). Azahomoerythromycin D derivative and intermediates
thereof. US Patent 4,465,674 A.
Broad, J., & Sanger, G. J. (2013). The antibiotic azithromycin is a motilin receptor agonist in
human stomach: comparison with erythromycin. Br J Pharmacol 168, 18591867.
Brook, I. (2006). Sinusitis of odontogenic origin. Otolaryngol Head Neck Surg 135, 349355.
Brusselle, G. G., Vanderstichele, C., Jordens, P., Deman, R., Slabbynck, H., Ringoet, V., et al.
(2013). Azithromycin for prevention of exacerbations in severe asthma (AZISAST): a
multicentre randomised double-blind placebo-controlled trial. Thorax 68, 322329.
Butorac-Petanjek, B., Parnham, M. J., & Popovic-Grle, S. (2010). Antibiotic therapy for exacerbations of chronic obstructive pulmonary disease (COPD). J Chemother 22,
291297.
Cameron, E. J., McSharry, C., Chaudhuri, R., Farrow, S., & Thomson, N. C. (2012). Long-term
macrolide treatment of chronic inammatory airway diseases: risks, benets and future developments. Clin Exp Allergy 42, 13021312.
Carevic, O., & Djokic, S. (1988). Comparative studies on the effects of erythromycin A and
azithromycin upon extracellular release of lysosomal enzymes in inammatory processes. Agents Actions 25, 124131.
Carlier, M. B., Garcia-Luque, I., Montenez, J. P., Tulkens, P. M., & Piret, J. (1994). Accumulation, release and subcellular localization of azithromycin in phagocytic and nonphagocytic cells in culture. Int J Tissue React 16, 211220.
Champney, W. S., & Burdine, R. (1998). Azithromycin and clarithromycin inhibition of 50S
ribosomal subunit formation in Staphylococcus aureus cells. Curr Microbiol 36,
119123.
Champney, W. S., Tober, C. L., & Burdine, R. (1998). A comparison of the inhibition of
translation and 50S ribosomal subunit formation in Staphylococcus aureus cells by
nine different macrolide antibiotics. Curr Microbiol 37, 412417.
Cigana, C., Nicolis, E., Pasetto, M., Assael, B. M., & Melotti, P. (2006). Anti-inammatory effects of azithromycin in cystic brosis airway epithelial cells. Biochem Biophys Res
Commun 350, 977982.
Clement, A., Tamalet, A., Leroux, E., Ravilly, S., Fauroux, B., & Jais, J. P. (2006). Long term
effects of azithromycin in patients with cystic brosis: a double blind, placebo controlled trial. Thorax 61, 895902.
Coates, P., Daniel, R., Houston, A. C., Antrobus, J. H., & Taylor, T. (1991). An open study to
compare the pharmacokinetics, safety and tolerability of a multiple-dose regimen of
azithromycin in young and elderly volunteers. Eur J Clin Microbiol Infect Dis 10,
850852.
Coloma, P. M., Schuemie, M. J., Triro, G., Furlong, L., van Mulligen, E., BauerMehren, A., et al. (2013). Drug-induced acute myocardial infarction: identifying prime suspects from electronic healthcare records-based surveillance
system. PLoS One 8, e72148.
Culic, O., Erakovic, V., Cepelak, I., Barisic, K., Brajsa, K., Ferencic, Z., et al. (2002).
Azithromycin modulates neutrophil function and circulating inammatory mediators
in healthy human subjects. Eur J Pharmacol 450, 277289.
Culic, O., Erakovic, V., & Parnham, M. J. (2001). Anti-inammatory effects of macrolide antibiotics. Eur J Pharmacol 429, 209229.
Curatolo, W., Liu, P., Johnson, B. A., Hausberger, A., Quan, E., Vendola, T., et al. (2011). Effects of food on a gastrically degraded drug: azithromycin fast-dissolving gelatin capsules and HPMC capsules. Pharm Res 28, 15311539.
Daenas, C., Hatziefthimiou, A. A., Gourgoulianis, K. I., & Molyvdas, P. A. (2006).
Azithromycin has a direct relaxant effect on precontracted airway smooth muscle.
Eur J Pharmacol 553, 280287.
Dahl, E. L., & Rosenthal, P. J. (2007). Multiple antibiotics exert delayed effects against
the Plasmodium falciparum apicoplast. Antimicrob Agents Chemother 51,
34853490.
Dellinger, R. P., Levy, M. M., Rhodes, A., Annane, D., Gerlach, H., Opal, S. M., et al. (2013).
Surviving sepsis campaign: international guidelines for management of severe sepsis
and septic shock: 2012. Crit Care Med 41, 580637.
den Heijer, C. D., van Bijnen, E. M., Paget, W. J., Pringle, M., Goossens, H., Bruggeman, C. A.,
et al. (2013). Prevalence and resistance of commensal Staphylococcus aureus, including meticillin-resistant S. aureus, in nine European countries: a cross-sectional study.
Lancet Infect Dis 13, 409415.
Di Paolo, A., Barbara, C., Chella, A., Angeletti, C. A., & Del Tacca, M. (2002). Pharmacokinetics of azithromycin in lung tissue, bronchial washing, and plasma in patients given
multiple oral doses of 500 and 1000 mg daily. Pharmacol Res 46, 545550.
Dias, R., & Canica, M. (2004). Emergence of invasive erythromycin-resistant Streptococcus
pneumoniae strains in Portugal: contribution and phylogenetic relatedness of serotype 14. J Antimicrob Chemother 54, 10351039.
Dinos, G. P., Michelinaki, M., & Kalpaxis, D. L. (2001). Insights into the mechanism of
azithromycin interaction with an Escherichia coli functional ribosomal complex. Mol
Pharmacol 59, 14411445.
Ehnhage, A., Rautiainen, M., Fang, A. F., & Sanchez, S. P. (2008). Pharmacokinetics of
azithromycin in serum and sinus uid after administration of extended-release and
immediate-release formulations in patients with acute bacterial sinusitis. Int J
Antimicrob Agents 31, 561566.
Erakovic Haber, V., Bosnar, M., & Kragol, G. (2014). Design of novel classes of macrolides
for neutrophil dominated inammatory diseases. Future Medicinal Chemistry 6 in
press.
Ernst, E. J., Klepser, M. E., Klepser, T. B., Nightingale, C. H., & Hunsicker, L. G. (2000). Comparison of the serum and intracellular pharmacokinetics of azithromycin in healthy
and diabetic volunteers. Pharmacotherapy 20, 657661.
Esposito, S., Bosis, S., Faelli, N., Begliatti, E., Droghetti, R., Tremolati, E., et al. (2005). Role of
atypical bacteria and azithromycin therapy for children with recurrent respiratory
tract infections. Pediatr Infect Dis J 24, 438444.

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245


Fa, N., Lins, L., Courtoy, P. J., Dufrene, Y., Van Der Smissen, P., Brasseur, R., et al. (2007). Decrease of elastic moduli of DOPC bilayers induced by a macrolide antibiotic,
azithromycin. Biochim Biophys Acta 1768, 18301838.
Falk, L., Fredlund, H., & Jensen, J. S. (2003). Tetracycline treatment does not eradicate Mycoplasma genitalium. Sex Transm Infect 79, 318319.
Fang, A. F., Palmer, J. N., Chiu, A. G., Blumer, J. L., Crownover, P. H., Campbell, M. D., et al.
(2009). Pharmacokinetics of azithromycin in plasma and sinus mucosal tissue following administration of extended-release or immediate-release formulations in adult
patients with chronic rhinosinusitis. Int J Antimicrob Agents 34, 6771.
Farrell, D. J., & Jenkins, S. G. (2004). Distribution across the USA of macrolide resistance
and macrolide resistance mechanisms among Streptococcus pneumoniae isolates collected from patients with respiratory tract infections: PROTEKT US 20012002.
J Antimicrob Chemother 54(Suppl. 1), i17i22.
Federica, M., Nadia, S., Monica, M., Alessandro, C., Tiberio, O., Francesco, B., et al. (2011).
Clinical and immunological evaluation of 12-month azithromycin therapy in chronic
lung allograft rejection. Clin Transplant 25, E381E389.
Felmingham, D., & Gruneberg, R. N. (2000). The Alexander Project 19961997: latest
susceptibility data from this international study of bacterial pathogens from
community-acquired lower respiratory tract infections. J Antimicrob Chemother
45, 191203.
Feola, D. J., Garvy, B. A., Cory, T. J., Birket, S. E., Hoy, H., Hayes, D., Jr., et al. (2010).
Azithromycin alters macrophage phenotype and pulmonary compartmentalization
during lung infection with Pseudomonas. Antimicrob Agents Chemother 54,
24372447.
Fernandes, A. C., Anderson, R., Theron, A. J., Joone, G., & Van Rensburg, C. E. (1984). Enhancement of human polymorphonuclear leucocyte motility by erythromycin
in vitro and in vivo. S Afr Med J 66, 173177.
Fernandez-Robredo, P., Recalde, S., Moreno-Orduna, M., Garcia-Garcia, L., ZarranzVentura, J., & Garcia-Layana, A. (2013). Azithromycin reduces inammation in a rat
model of acute conjunctivitis. Mol Vis 19, 153165.
Fernandez, A. D., Elmore, M. K., & Metzger, D. W. (2004). Azithromycin modulates murine
immune responses to pneumococcal conjugate vaccine and inhibits nasal clearance
of bacteria. J Infect Dis 190, 17621766.
Fimia, G. M., & Piacentini, M. (2010). Regulation of autophagy in mammals and its interplay with apoptosis. Cell Mol Life Sci 67, 15811588.
Fleet, J. L., Shariff, S. Z., Bailey, D. G., Gandhi, S., Juurlink, D. N., Nash, D. M., et al. (2013).
Comparing two types of macrolide antibiotics for the purpose of assessing
population-based drug interactions. BMJ Open 3.
Florey, O., & Overholtzer, M. (2012). Autophagy proteins in macroendocytic engulfment.
Trends Cell Biol 22, 374380.
Foulds, G., Madsen, P., Cox, C., Shepard, R., & Johnson, R. (1991). Concentration of
azithromycin in human prostatic tissue. Eur J Clin Microbiol Infect Dis 10, 868871.
Foulds, G., Shepard, R. M., & Johnson, R. B. (1990). The pharmacokinetics of azithromycin
in human serum and tissues. J Antimicrob Chemother 25(Suppl. A), 7382.
Franceschi, F., Kanyo, Z., Sherer, E. C., & Sutcliffe, J. (2004). Macrolide resistance from the
ribosome perspective. Curr Drug Targets Infect Disord 4, 177191.
Garver, E., Hugger, E. D., Shearn, S. P., Rao, A., Dawson, P. A., Davis, C. B., et al. (2008). Involvement of intestinal uptake transporters in the absorption of azithromycin and
clarithromycin in the rat. Drug Metab Dispos 36, 24922498.
Gebre, T., Ayele, B., Zerihun, M., Genet, A., Stoller, N. E., Zhou, Z., et al. (2012). Comparison
of annual versus twice-yearly mass azithromycin treatment for hyperendemic trachoma in Ethiopia: a cluster-randomised trial. Lancet 379, 143151.
Gerbaux, C., Van Bambeke, F., Montenez, J. P., Piret, J., Morlighem, G., & Tulkens, P. M.
(1996). Hyperactivity of cathepsin B and other lysosomal enzymes in broblasts exposed to azithromycin, a dicationic macrolide antibiotic with exceptional tissue accumulation. FEBS Lett 394, 307310.
Gerhardt, S. G., McDyer, J. F., Girgis, R. E., Conte, J. V., Yang, S. C., & Orens, J. B. (2003). Maintenance azithromycin therapy for bronchiolitis obliterans syndrome: results of a pilot
study. Am J Respir Crit Care Med 168, 121125.
Geudens, N., Timmermans, L., Vanhooren, H., Vanaudenaerde, B. M., Vos, R., Van De
Wauwer, C., et al. (2008). Azithromycin reduces airway inammation in a murine
model of lung ischaemia reperfusion injury. Transpl Int 21, 688695.
Giamarellos-Bourboulis, E. J., Mylona, V., Antonopoulou, A., Tsangaris, I., Koutelidakis, I.,
Marioli, A., et al. (2013). Effect of clarithromycin in patients with suspected Gramnegative sepsis: results of a randomized controlled trial. J Antimicrob Chemother,
http://dx.doi.org/10.1093/jac/dkt475.
Giamarellos-Bourboulis, E. J., Pechere, J. C., Routsi, C., Plachouras, D., Kollias, S.,
Raftogiannis, M., et al. (2008). Effect of clarithromycin in patients with sepsis and
ventilator-associated pneumonia. Clin Infect Dis 46, 11571164.
Giannopoulos, A., Koratzanis, G., Giamarellos-Bourboulis, E. J., Panou, C., Adamakis, I., &
Giamarellou, H. (2001). Pharmacokinetics of clarithromycin in the prostate: implications for the treatment of chronic abacterial prostatitis. J Urol 165, 9799.
Gielen, V., Johnston, S. L., & Edwards, M. R. (2010). Azithromycin induces anti-viral responses in bronchial epithelial cells. Eur Respir J 36, 646654.
Giovanetti, E., Brenciani, A., Burioni, R., & Varaldo, P. E. (2002). A novel efux system in
inducibly erythromycin-resistant strains of Streptococcus pyogenes. Antimicrob
Agents Chemother 46, 37503755.
Giovanetti, E., Montanari, M. P., Mingoia, M., & Varaldo, P. E. (1999). Phenotypes and
genotypes of erythromycin-resistant Streptococcus pyogenes strains in Italy and
heterogeneity of inducibly resistant strains. Antimicrob Agents Chemother 43,
19351940.
Giudicessi, J. R., & Ackerman, M. J. (2013). Azithromycin and risk of sudden cardiac death:
guilty as charged or falsely accused? Cleve Clin J Med 80, 539544.
Gladue, R. P., Bright, G. M., Isaacson, R. E., & Newborg, M. F. (1989). In vitro and in vivo
uptake of azithromycin (CP-62,993) by phagocytic cells: possible mechanism of delivery and release at sites of infection. Antimicrob Agents Chemother 33, 277282.

Gladue, R. P., & Snider, M. E. (1990). Intracellular accumulation of azithromycin by cultured human broblasts. Antimicrob Agents Chemother 34, 10561060.
Glojnaric, I., Cuzic, S., Erakovic-Haber, V., & Parnham, M. J. (2007). The serum amyloid A
response to sterile silver nitrate in mice and its inhibition by dexamethasone and
macrolide antibiotics. Int Immunopharmacol 7, 15441551.
Gomi, K., Yashima, A., Iino, F., Kanazashi, M., Nagano, T., Shibukawa, N., et al. (2007). Drug
concentration in inamed periodontal tissues after systemically administered
azithromycin. J Periodontol 78, 918923.
Gottlieb, J., Szangolies, J., Koehnlein, T., Golpon, H., Simon, A., & Welte, T. (2008). Longterm azithromycin for bronchiolitis obliterans syndrome after lung transplantation.
Transplantation 85, 3641.
Halldorsson, S., Gudjonsson, T., Gottfredsson, M., Singh, P. K., Gudmundsson, G. H., &
Baldursson, O. (2010). Azithromycin maintains airway epithelial integrity during
Pseudomonas aeruginosa infection. Am J Respir Cell Mol Biol 42, 6268.
Hand, W. L., & Hand, D. L. (2001). Characteristics and mechanisms of azithromycin accumulation and efux in human polymorphonuclear leukocytes. Int J Antimicrob Agents
18, 419425.
Hansen, J. L., Ippolito, J. A., Ban, N., Nissen, P., Moore, P. B., & Steitz, T. A. (2002). The structures of four macrolide antibiotics bound to the large ribosomal subunit. Mol Cell 10,
117128.
Hao, K., Qi, Q., Hao, H., Wang, G., Chen, Y., Liang, Y., et al. (2013). The pharmacokinetic
pharmacodynamic model of azithromycin for lipopolysaccharide-induced
depressive-like behavior in mice. PLoS One 8, e54981.
Hassan, H. E., Othman, A. A., Eddington, N. D., Duffy, L., Xiao, L., Waites, K. B., et al. (2011).
Pharmacokinetics, safety, and biologic effects of azithromycin in extremely preterm
infants at risk for ureaplasma colonization and bronchopulmonary dysplasia. J Clin
Pharmacol 51, 12641275.
Healy, D. P., Silverman, P. A., Neely, A. N., Holder, I. A., & Babcock, G. E. (2002). Effect of
antibiotics on polymorphonuclear neutrophil apoptosis. Pharmacotherapy 22,
578585.
Henry, D. C., Riffer, E., Sokol, W. N., Chaudry, N. I., & Swanson, R. N. (2003). Randomized
double-blind study comparing 3- and 6-day regimens of azithromycin with a 10day amoxicillin-clavulanate regimen for treatment of acute bacterial sinusitis.
Antimicrob Agents Chemother 47, 27702774.
Hirsch, R., Deng, H., & Laohachai, M. N. (2012). Azithromycin in periodontal treatment:
more than an antibiotic. J Periodontal Res 47, 137148.
Hodge, S., Hodge, G., Brozyna, S., Jersmann, H., Holmes, M., & Reynolds, P. N. (2006).
Azithromycin increases phagocytosis of apoptotic bronchial epithelial cells by alveolar macrophages. Eur Respir J 28, 486495.
Hodge, S., Hodge, G., Jersmann, H., Matthews, G., Ahern, J., Holmes, M., et al. (2008).
Azithromycin improves macrophage phagocytic function and expression of mannose
receptor in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 178,
139148.
Hodge, S., & Reynolds, P. N. (2012). Low-dose azithromycin improves phagocytosis of
bacteria by both alveolar and monocyte-derived macrophages in chronic obstructive
pulmonary disease subjects. Respirology 17, 802807.
Hofer, D., Koeppe, P., & Paeske, B. (1995). Pharmacokinetics of azithromycin in normal
and impaired renal function. Infection 23, 356361.
Hoffmann, N., Lee, B., Hentzer, M., Rasmussen, T. B., Song, Z., Johansen, H. K., et al. (2007).
Azithromycin blocks quorum sensing and alginate polymer formation and increases
the sensitivity to serum and stationary-growth-phase killing of Pseudomonas
aeruginosa and attenuates chronic P. aeruginosa lung infection in Cftr(/) mice.
Antimicrob Agents Chemother 51, 36773687.
Hoiby, N. (2011). Recent advances in the treatment of Pseudomonas aeruginosa infections
in cystic brosis. BMC Med 9, 32.
Ianaro, A., Ialenti, A., Mafa, P., Sautebin, L., Rombola, L., Carnuccio, R., et al. (2000).
Anti-inammatory activity of macrolide antibiotics. J Pharmacol Exp Ther 292,
156163.
Ikeda, K., Hirayama, M., Hirota, Y., Asa, E., Seki, J., & Tanaka, Y. (2008). Drug-induced
phospholipidosis is caused by blockade of mannose 6-phosphate receptor-mediated
targeting of lysosomal enzymes. Biochem Biophys Res Commun 377, 268274.
Ikegaya, S., Inai, K., Iwasaki, H., Naiki, H., & Ueda, T. (2009). Azithromycin reduces tumor
necrosis factor-alpha production in lipopolysaccharide-stimulated THP-1 monocytic
cells by modication of stress response and p38 MAPK pathway. J Chemother 21,
396402.
Imamura, Y., Yanagihara, K., Mizuta, Y., Seki, M., Ohno, H., Higashiyama, Y., et al. (2004).
Azithromycin inhibits MUC5AC production induced by the Pseudomonas aeruginosa
autoinducer N-(3-Oxododecanoyl) homoserine lactone in NCI-H292 Cells.
Antimicrob Agents Chemother 48, 34573461.
Inoue, M., Farrell, D. J., Kaneko, K., Akizawa, K., Fujita, S., Kaku, M., et al. (2008). Antimicrobial susceptibility of respiratory tract pathogens in Japan during PROTEKT years 15
(19992004). Microb Drug Resist 14, 109117.
Ishimatsu, Y., Kadota, J., Iwashita, T., Nagata, T., Ishii, H., Shikuwa, C., et al. (2004).
Macrolide antibiotics induce apoptosis of human peripheral lymphocytes in vitro.
Int J Antimicrob Agents 24, 247253.
Ishimoto, H., Mukae, H., Sakamoto, N., Amenomori, M., Kitazaki, T., Imamura, Y., et al.
(2009). Different effects of telithromycin on MUC5AC production induced by
human neutrophil peptide-1 or lipopolysaccharide in NCI-H292 cells compared
with azithromycin and clarithromycin. J Antimicrob Chemother 63, 109114.
Ivetic Tkalcevic, V., Bosnjak, B., Hrvacic, B., Bosnar, M., Marjanovic, N., Ferencic, Z., et al.
(2006). Anti-inammatory activity of azithromycin attenuates the effects of lipopolysaccharide administration in mice. Eur J Pharmacol 539, 131138.
Ivetic Tkalcevic, V., Cuzic, S., Kramaric, M. D., Parnham, M. J., & Erakovic Haber, V. (2012).
Topical azithromycin and clarithromycin inhibit acute and chronic skin inammation
in sensitized mice, with apparent selectivity for Th2-mediated processes in delayedtype hypersensitivity. Inammation 35, 192205.

241

242

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

Iwamoto, S., Azuma, E., Kumamoto, T., Hirayama, M., Yoshida, T., Ito, M., et al. (2013). Efcacy of azithromycin in preventing lethal graft-versus-host disease. Clin Exp
Immunol 171, 338345.
Iwamoto, S., Kumamoto, T., Azuma, E., Hirayama, M., Ito, M., Amano, K., et al. (2011). The
effect of azithromycin on the maturation and function of murine bone marrowderived dendritic cells. Clin Exp Immunol 166, 385392.
Jacobs, M. R., & Johnson, C. E. (2003). Macrolide resistance: an increasing concern for
treatment failure in children. Pediatr Infect Dis J 22, S131S138.
Jacobs, R. F., Maples, H. D., Aranda, J. V., Espinoza, G. M., Knirsch, C., Chandra, R., et al.
(2005). Pharmacokinetics of intravenously administered azithromycin in pediatric
patients. Pediatr Infect Dis J 24, 3439.
Jenkins, S. G., Brown, S. D., & Farrell, D. J. (2008). Trends in antibacterial resistance among
Streptococcus pneumoniae isolated in the USA: update from PROTEKT US Years 14.
Ann Clin Microbiol Antimicrob 7, 1.
Ju, J. S., & Weihl, C. C. (2010). Inclusion body myopathy, Paget's disease of the bone and
fronto-temporal dementia: a disorder of autophagy. Hum Mol Genet 19, R38R45.
Kannan, S., & Mattoo, T. K. (2001). Diffuse crescentic glomerulonephritis in bacterial endocarditis. Pediatr Nephrol 16, 423428.
Kanoh, S., & Rubin, B. K. (2010). Mechanisms of action and clinical application of
macrolides as immunomodulatory medications. Clin Microbiol Rev 23, 590615.
Karhu, J., Ala-Kokko, T. I., Ohtonen, P., & Syrjala, H. (2013). Severe community-acquired
pneumonia treated with beta-lactam-respiratory quinolone vs. beta-lactammacrolide combination. Acta Anaesthesiol Scand 57, 587593.
Karpov, O. I. (1999). The clinical and economic efcacies of short courses of azithromycin
in acute sinusitis. Antibiot Khimioter 44, 2832.
Kent, J. R., Almond, M. K., & Dhillon, S. (2001). Azithromycin: an assessment of its pharmacokinetics and therapeutic potential in CAPD. Perit Dial Int 21, 372377.
Kezerashvili, A., Khattak, H., Barsky, A., Nazari, R., & Fisher, J. D. (2007). Azithromycin as a
cause of QT-interval prolongation and torsade de pointes in the absence of other
known precipitating factors. J Interv Card Electrophysiol 18, 243246.
Khair, O. A., Devalia, J. L., Abdelaziz, M. M., Sapsford, R. J., & Davies, R. J. (1995). Effect of
erythromycin on Haemophilus inuenzae endotoxin-induced release of IL-6, IL-8
and sICAM-1 by cultured human bronchial epithelial cells. Eur Respir J 8, 14511457.
Khan, A. A., Slifer, T. R., Araujo, F. G., & Remington, J. S. (1999). Effect of clarithromycin and
azithromycin on production of cytokines by human monocytes. Int J Antimicrob
Agents 11, 121132.
Kita, E., Sawaki, M., Nishikawa, F., Mikasa, K., Yagyu, Y., Takeuchi, S., et al. (1990). Enhanced interleukin production after long-term administration of erythromycin stearate. Pharmacology 41, 177183.
Kneyber, M. C., van Woensel, J. B., Uijtendaal, E., Uiterwaal, C. S., & Kimpen, J. L.Dutch
Antibiotics in, R. S. V. T. R. G. (2008). Azithromycin does not improve disease course
in hospitalized infants with respiratory syncytial virus (RSV) lower respiratory tract
disease: a randomized equivalence trial. Pediatr Pulmonol 43, 142149.
Kobayashi, H., Ohgaki, N., & Takeda, H. (1993). Therapeutic possibilities for diffuse
panbronchiolitis. Int J Antimicrob Agents 3(Suppl. 1), S81S86.
Kobayashi, Y., Wada, H., Rossios, C., Takagi, D., Higaki, M., Mikura, S., et al. (2013). A novel
macrolide solithromycin exerts superior anti-inammatory effect via NF-kappaB inhibition. J Pharmacol Exp Ther 345, 7684.
Kobrehel, G., Radobolja, G., Tamburasev, Z., Djokic, S. (1982). 11-Aza-10-deoxo-10dihydroerythromycin A and derivatives thereof as well as a process for their preparation. U.S. Patent 4,328,334.
Koch, C. C., Esteban, D. J., Chin, A. C., Olson, M. E., Read, R. R., Ceri, H., et al. (2000). Apoptosis,
oxidative metabolism and interleukin-8 production in human neutrophils exposed to
azithromycin: effects of Streptococcus pneumoniae. J Antimicrob Chemother 46, 1926.
Kohler, T., Dumas, J. L., & Van Delden, C. (2007). Ribosome protection prevents
azithromycin-mediated quorum-sensing modulation and stationary-phase killing of
Pseudomonas aeruginosa. Antimicrob Agents Chemother 51, 42434248.
Kolumbic Lakos, A., Skerk, V., Malekovic, G., Dujnic Spoljarevic, T., Kovacic, D., Pasini, M.,
et al. (2011). A switch therapy protocol with intravenous azithromycin and ciprooxacin combination for severe, relapsing chronic bacterial prostatitis: a prospective
non-comparative pilot study. J Chemother 23, 350353.
Konno, S., Adachi, M., Asano, K., Kawazoe, T., Okamoto, K., & Takahashi, T. (1992). Inuences of roxithromycin on cell-mediated immune responses. Life Sci 51, L107L112.
Kornfeld, S. (1992). Structure and function of the mannose 6-phosphate/insulinlike
growth factor II receptors. Annu Rev Biochem 61, 307330.
Kosol, S., Schrank, E., Krajacic, M. B., Wagner, G. E., Meyer, N. H., Gobl, C., et al. (2012).
Probing the interactions of macrolide antibiotics with membrane-mimetics by NMR
spectroscopy. J Med Chem 55, 56325636.
Kotsaki, A., & Giamarellos-Bourboulis, E. J. (2012). Emerging drugs for the treatment of
sepsis. Expert Opin Emerg Drugs 17, 379391.
Kudoh, S. (2004). Applying lessons learned in the treatment of diffuse panbronchiolitis to
other chronic inammatory diseases. Am J Med 117(Suppl. 9A), 12S19S.
Kudoh, S., Azuma, A., Yamamoto, M., Izumi, T., & Ando, M. (1998). Improvement of survival in patients with diffuse panbronchiolitis treated with low-dose erythromycin. Am J
Respir Crit Care Med 157, 18291832.
Lakos, A. K., Pangercic, A., Gasparic, M., Kukuruzovic, M. M., Kovacic, D., & Barsic, B. (2012).
Safety and effectiveness of azithromycin in the treatment of respiratory infections in
children. Curr Med Res Opin 28, 155162.
Lau, C. Y., & Qureshi, A. K. (2002). Azithromycin versus doxycycline for genital
chlamydial infections: a meta-analysis of randomized clinical trials. Sex
Transm Dis 29, 497502.
Leclercq, R. (2002). Mechanisms of resistance to macrolides and lincosamides: nature of
the resistance elements and their clinical implications. Clin Infect Dis 34, 482492.
Legssyer, R., Huaux, F., Lebacq, J., Delos, M., Marbaix, E., Lebecque, P., et al. (2006).
Azithromycin reduces spontaneous and induced inammation in DeltaF508 cystic brosis mice. Respir Res 7, 134.

Li, H., Liu, D. H., Chen, L. L., Zhao, Q., Yu, Y. Z., Ding, J. J., et al. (2013). Adverse effects of
long-term azithromycin use in patients with chronic lung diseases: a meta-analysis.
Antimicrob Agents Chemother 58, 511517.
Li, H., Zhou, Y., Fan, F., Zhang, Y., Li, X., Yu, H., et al. (2011). Effect of azithromycin on patients with diffuse panbronchiolitis: retrospective study of 51 cases. Intern Med 50,
16631669.
Lin, H. C., Wang, C. H., Liu, C. Y., Yu, C. T., & Kuo, H. P. (2000). Erythromycin inhibits beta2integrins (CD11b/CD18) expression, interleukin-8 release and intracellular oxidative
metabolism in neutrophils. Respir Med 94, 654660.
Lin, S. J., Yan, D. C., Lee, W. I., Kuo, M. L., Hsiao, H. S., & Lee, P. Y. (2012). Effect of
azithromycin on natural killer cell function. Int Immunopharmacol 13, 814.
Liu, P., Fang, A. F., LaBadie, R. R., Crownover, P. H., & Arguedas, A. G. (2011). Comparison of
azithromycin pharmacokinetics following single oral doses of extended-release and
immediate-release formulations in children with acute otitis media. Antimicrob
Agents Chemother 55, 50225026.
Lode, H., Borner, K., Koeppe, P., & Schaberg, T. (1996). Azithromycinreview of key chemical,
pharmacokinetic and microbiological features. J Antimicrob Chemother 37(Suppl. C), 18.
Lucchi, M., Damle, B., Fang, A., de Caprariis, P. J., Mussi, A., Sanchez, S. P., et al. (2008).
Pharmacokinetics of azithromycin in serum, bronchial washings, alveolar macrophages and lung tissue following a single oral dose of extended or immediate release
formulations of azithromycin. J Antimicrob Chemother 61, 884891.
Luchs, J. (2010). Azithromycin in DuraSite for the treatment of blepharitis. Clin
Ophthalmol 4, 681688.
Luke, D. R., & Foulds, G. (1997). Disposition of oral azithromycin in humans. Clin
Pharmacol Ther 61, 641648.
Luo, L., Perelman, J. M., Kolosov, V. P., & Zhou, X. D. (2011). Inhibition of airway mucous
hypersecretion by azithromycin through matrix metalloproteinase 9. Zhonghua Yi
Xue Za Zhi 91, 689693.
Lutz, L., Pereira, D. C., Paiva, R. M., Zavascki, A. P., & Barth, A. L. (2012). Macrolides
decrease the minimal inhibitory concentration of anti-pseudomonal agents
against Pseudomonas aeruginosa from cystic brosis patients in biolm. BMC
Microbiol 12, 196.
Magri, V., Marras, E., Skerk, V., Markotic, A., Restelli, A., Garlaschi, M. C., et al. (2010). Eradication of Chlamydia trachomatis parallels symptom regression in chronic bacterial
prostatitis patients treated with a uoroquinolone-macrolide combination.
Andrologia 42, 366375.
Magri, V., Trinchieri, A., Pozzi, G., Restelli, A., Garlaschi, M. C., Torresani, E., et al. (2007).
Efcacy of repeated cycles of combination therapy for the eradication of infecting organisms in chronic bacterial prostatitis. Int J Antimicrob Agents 29, 549556.
Malhotra-Kumar, S., Lammens, C., Coenen, S., Van Herck, K., & Goossens, H. (2007). Effect
of azithromycin and clarithromycin therapy on pharyngeal carriage of macrolideresistant streptococci in healthy volunteers: a randomised, double-blind, placebocontrolled study. Lancet 369, 482490.
Mann, J. M., Sha, K. K., Kline, G., Breuer, F. U., & Miller, A. (2005). World Trade Center dyspnea: bronchiolitis obliterans with functional improvement: a case report. Am J Ind
Med 48, 225229.
Marjanovic, N., Bosnar, M., Michielin, F., Wille, D. R., Anic-Milic, T., Culic, O., et al. (2011).
Macrolide antibiotics broadly and distinctively inhibit cytokine and chemokine production by COPD sputum cells in vitro. Pharmacol Res 63, 389397.
Marple, B. F., Roberts, C. S., Frytak, J. R., Schabert, V. F., Wegner, J. C., Bhattacharyya, H.,
et al. (2010). Azithromycin extended release vs amoxicillin/clavulanate: symptom
resolution in acute sinusitis. Am J Otolaryngol 31, 18.
Martin-Loeches, I., Lisboa, T., Rodriguez, A., Putensen, C., Annane, D., Garnacho-Montero, J.,
et al. (2010). Combination antibiotic therapy with macrolides improves survival in
intubated patients with community-acquired pneumonia. Intensive Care Med 36,
612620.
Massei, F., Gori, L., Macchia, P., & Maggiore, G. (2005). The expanded spectrum of
bartonellosis in children. Infect Dis Clin North Am 19, 691711.
Matijasic, M., Munic Kos, V., Nujic, K., Cuzic, S., Padovan, J., Kragol, G., et al. (2012). Fluorescently labeled macrolides as a tool for monitoring cellular and tissue distribution
of azithromycin. Pharmacol Res 66, 332342.
Matsumura, Y., Mitani, A., Suga, T., Kamiya, Y., Kikuchi, T., Tanaka, S., et al. (2011).
Azithromycin may inhibit interleukin-8 through suppression of Rac1 and a nuclear
factor-kappa B pathway in KB cells stimulated with lipopolysaccharide. J Periodontol
82, 16231631.
Matzneller, P., Krasniqi, S., Kinzig, M., Sorgel, F., Huttner, S., Lackner, E., et al. (2013).
Blood, tissue, and intracellular concentrations of azithromycin during and after end
of therapy. Antimicrob Agents Chemother 57, 17361742.
Mazzei, T., Surrenti, C., Novelli, A., Crispo, A., Fallani, S., Carla, V., et al. (1993). Pharmacokinetics of azithromycin in patients with impaired hepatic function. J Antimicrob
Chemother 31(Suppl. E), 5763.
McDonald, P. J., & Pruul, H. (1991). Phagocyte uptake and transport of azithromycin. Eur J
Clin Microbiol Infect Dis 10, 828833.
Mena, L. A., Mroczkowski, T. F., Nsuami, M., & Martin, D. H. (2009). A randomized comparison of azithromycin and doxycycline for the treatment of Mycoplasma genitaliumpositive urethritis in men. Clin Infect Dis 48, 16491654.
Mencarelli, A., Distrutti, E., Renga, B., Cipriani, S., Palladino, G., Booth, C., et al. (2011). Development of non-antibiotic macrolide that corrects inammation-driven immune
dysfunction in models of inammatory bowel diseases and arthritis. Eur J Pharmacol
665, 2939.
Mergenhagen, K. A., Olbrych, P. M., Mattappallil, A., Krajewski, M. P., & Ott, M. C. (2013).
Effect of azithromycin on anticoagulation-related outcomes in geriatric patients receiving warfarin. Clin Ther 35, 425430.
Mertens, V., Blondeau, K., Pauwels, A., Farre, R., Vanaudenaerde, B., Vos, R., et al. (2009).
Azithromycin reduces gastroesophageal reux and aspiration in lung transplant recipients. Dig Dis Sci 54, 972979.

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245


Metersky, M. L., Ma, A., Houck, P. M., & Bratzler, D. W. (2007). Antibiotics for bacteremic
pneumonia: improved outcomes with macrolides but not uoroquinolones. Chest
131, 466473.
Meyer, M., Huaux, F., Gavilanes, X., van den Brule, S., Lebecque, P., Lo Re, S., et al. (2009).
Azithromycin reduces exaggerated cytokine production by M1 alveolar macrophages
in cystic brosis. Am J Respir Cell Mol Biol 41, 590602.
Milberg, P., Eckardt, L., Bruns, H. J., Biertz, J., Ramtin, S., Reinsch, N., et al. (2002). Divergent
proarrhythmic potential of macrolide antibiotics despite similar QT prolongation: fast
phase 3 repolarization prevents early afterdepolarizations and torsade de pointes. J
Pharmacol Exp Ther 303, 218225.
Miller, S. A., Selzman, C. H., Shames, B. D., Barton, H. A., Johnson, S. M., & Harken, A. H.
(2000). Chlamydia pneumoniae activates nuclear factor kappaB and activator protein
1 in human vascular smooth muscle and induces cellular proliferation. J Surg Res 90,
7681.
Millrose, M., Kruse, M., Flick, B., & Stahlmann, R. (2009). Effects of macrolides on proinammatory epitopes on endothelial cells in vitro. Arch Toxicol 83, 469476.
Min, J. Y., & Jang, Y. J. (2012). Macrolide therapy in respiratory viral infections. Mediators
Inamm 2012, 649570.
Miossec-Bartoli, C., Pilatre, L., Peyron, P., N'Diaye, E. N., Collart-Dutilleul, V., MaridonneauParini, I., et al. (1999). The new ketolide HMR3647 accumulates in the azurophil
granules of human polymorphonuclear cells. Antimicrob Agents Chemother 43,
24572462.
Miyazaki, M., Zaitsu, M., Honjo, K., Ishii, E., & Hamasaki, Y. (2003). Macrolide antibiotics
inhibit prostaglandin E2 synthesis and mRNA expression of prostaglandin synthetic
enzymes in human leukocytes. Prostaglandins Leukot Essent Fatty Acids 69, 229235.
Mizunoe, S., Kadota, J., Tokimatsu, I., Kishi, K., Nagai, H., & Nasu, M. (2004). Clarithromycin
and azithromycin induce apoptosis of activated lymphocytes via down-regulation of
Bcl-xL. Int Immunopharmacol 4, 12011207.
Montenez, J. P., Van Bambeke, F., Piret, J., Brasseur, R., Tulkens, P. M., & Mingeot-Leclercq,
M. P. (1999). Interactions of macrolide antibiotics (Erythromycin A, roxithromycin,
erythromycylamine [Dirithromycin], and azithromycin) with phospholipids:
computer-aided conformational analysis and studies on acellular and cell culture
models. Toxicol Appl Pharmacol 156, 129140.
Montenez, J. P., Van Bambeke, F., Piret, J., Schanck, A., Brasseur, R., Tulkens, P. M., et al.
(1996). Interaction of the macrolide azithromycin with phospholipids. II. Biophysical
and computer-aided conformational studies. Eur J Pharmacol 314, 215227.
Morinaga, Y., Yanagihara, K., Miyashita, N., Seki, M., Izumikawa, K., Kakeya, H., et al.
(2009). Azithromycin, clarithromycin and telithromycin inhibit MUC5AC induction
by Chlamydophila pneumoniae in airway epithelial cells. Pulm Pharmacol Ther 22,
580586.
Moriya, S., Che, X. F., Komatsu, S., Abe, A., Kawaguchi, T., Gotoh, A., et al. (2013). Macrolide
antibiotics block autophagy ux and sensitize to bortezomib via endoplasmic reticulum stress-mediated CHOP induction in myeloma cells. Int J Oncol 42, 15411550.
Mtairag, E. M., Abdelghaffar, H., Douhet, C., & Labro, M. T. (1995). Role of extracellular calcium in in vitro uptake and intraphagocytic location of macrolides. Antimicrob Agents
Chemother 39, 16761682.
Mulholland, S., Gavranich, J. B., Gillies, M. B., & Chang, A. B. (2012). Antibiotics for
community-acquired lower respiratory tract infections secondary to Mycoplasma
pneumoniae in children. Cochrane Database Syst Rev 9, CD004875.
Munic, V., Banjanac, M., Kostrun, S., Nujic, K., Bosnar, M., Marjanovic, N., et al. (2011). Intensity of macrolide anti-inammatory activity in J774A.1 cells positively correlates
with cellular accumulation and phospholipidosis. Pharmacol Res 64, 298307.
Munic, V., Kelneric, Z., Mikac, L., & Erakovic Haber, V. (2010). Differences in assessment of
macrolide interaction with human MDR1 (ABCB1, P-gp) using rhodamine-123 efux,
ATPase activity and cellular accumulation assays. Eur J Pharm Sci 41, 8695.
Murphy, B. S., Sundareshan, V., Cory, T. J., Hayes, D., Jr., Anstead, M. I., & Feola, D. J. (2008).
Azithromycin alters macrophage phenotype. J Antimicrob Chemother 61, 554560.
Murphy, D. M., Forrest, I. A., Corris, P. A., Johnson, G. E., Small, T., Jones, D., et al. (2008).
Azithromycin attenuates effects of lipopolysaccharide on lung allograft bronchial epithelial cells. J Heart Lung Transplant 27, 12101216.
Murray, J. J., Emparanza, P., Lesinskas, E., Tawadrous, M., & Breen, J. D. (2005). Efcacy and
safety of a novel, single-dose azithromycin microsphere formulation versus 10 days
of levooxacin for the treatment of acute bacterial sinusitis in adults. Otolaryngol
Head Neck Surg 133, 194200.
Muto, C., Liu, P., Chiba, K., & Suwa, T. (2011). Pharmacokineticpharmacodynamic analysis of azithromycin extended release in Japanese patients with common respiratory
tract infectious disease. J Antimicrob Chemother 66, 165174.
Naidoo, R. V., & Bryant, P. A. (2009). Not every cough in bronchiolitis season is bronchiolitis. BMJ Case Rep 2009.
Navarro-Xavier, R. A., Newson, J., Silveira, V. L., Farrow, S. N., Gilroy, D. W., & Bystrom, J.
(2010). A new strategy for the identication of novel molecules with targeted
proresolution of inammation properties. J Immunol 184, 15161525.
Nie, Y. C., Wu, H., Li, P. B., Xie, L. M., Luo, Y. L., Shen, J. G., et al. (2012). Naringin attenuates
EGF-induced MUC5AC secretion in A549 cells by suppressing the cooperative activities of MAPKs-AP-1 and IKKs-IkappaB-NF-kappaB signaling pathways. Eur J
Pharmacol 690, 207213.
Nightingale, C. H. (1997). Pharmacokinetics and pharmacodynamics of newer macrolides.
Pediatr Infect Dis J 16, 438443.
Nujic, K., Banjanac, M., Munic, V., Polancec, D., & Erakovic Haber, V. (2012). Impairment of
lysosomal functions by azithromycin and chloroquine contributes to antiinammatory phenotype. Cell Immunol 279, 7886.
Nujic, K., Smith, M., Lee, M., Belamaric, D., Tomaskovic, L., Alihodzic, S., et al. (2012).
Valosin containing protein (VCP) interacts with macrolide antibiotics without mediating their anti-inammatory activities. Eur J Pharmacol 677, 163172.
Panpanich, R., Lerttrakarnnon, P., & Laopaiboon, M. (2008). Azithromycin for acute lower
respiratory tract infections. Cochrane Database Syst Rev, CD001954.

Parnham, M. J. (2005). Antibiotics, inammation and its resolution: an overview. In B. K.


Rubin, & J. Tamaoka (Eds.), Antibiotics as Anti-Inammatory and Immunomodulatory
Agents (pp. 2747). Basel: Birkhuser Verlag.
Parnham, M. J., Culic, O., Erakovic, V., Munic, V., Popovic-Grle, S., Barisic, K., et al. (2005).
Modulation of neutrophil and inammation markers in chronic obstructive pulmonary disease by short-term azithromycin treatment. Eur J Pharmacol 517, 132143.
Pascual, A., Conejo, M. C., Garcia, I., & Perea, E. J. (1995). Factors affecting the intracellular
accumulation and activity of azithromycin. J Antimicrob Chemother 35, 8593.
Peeters, T., Matthijs, G., Depoortere, I., Cachet, T., Hoogmartens, J., & Vantrappen, G.
(1989). Erythromycin is a motilin receptor agonist. Am J Physiol 257, G470G474.
Pene Dumitrescu, T., Anic-Milic, T., Oreskovic, K., Padovan, J., Brouwer, K. L., Zuo, P., et al.
(2013). Development of a population pharmacokinetic model to describe
azithromycin whole-blood and plasma concentrations over time in healthy subjects.
Antimicrob Agents Chemother 57, 31943201.
Perletti, G., Skerk, V., Magri, V., Markotic, A., Mazzoli, S., Parnham, M. J., et al. (2011).
Macrolides for the treatment of chronic bacterial prostatitis: an effective application
of their unique pharmacokinetic and pharmacodynamic prole (Review). Mol Med
Rep 4, 10351044.
Persson, H. L., Vainikka, L. K., Sege, M., Wennerstrom, U., Dam-Larsen, S., & Persson, J.
(2012). Leaky lysosomes in lung transplant macrophages: azithromycin prevents oxidative damage. Respir Res 13, 83.
Piacentini, G. L., Peroni, D. G., Bodini, A., Pigozzi, R., Costella, S., Loiacono, A., et al. (2007).
Azithromycin reduces bronchial hyperresponsiveness and neutrophilic airway inammation in asthmatic children: a preliminary report. Allergy Asthma Proc 28,
194198.
Pinto, L. A., Pitrez, P. M., Luisi, F., de Mello, P. P., Gerhardt, M., Ferlini, R., et al. (2012).
Azithromycin therapy in hospitalized infants with acute bronchiolitis is not associated with better clinical outcomes: a randomized, double-blinded, and placebocontrolled clinical trial. J Pediatr 161, 11041108.
Pitsouni, E., Iavazzo, C., Athanasiou, S., & Falagas, M. E. (2007). Single-dose azithromycin
versus erythromycin or amoxicillin for Chlamydia trachomatis infection during pregnancy: a meta-analysis of randomised controlled trials. Int J Antimicrob Agents 30,
213221.
Plesko, S., Banic, M., Plecko, V., Anic, B., Brkic, T., Renata, H., et al. (2010). Effect of
azithromycin on acute inammatory lesions and colonic bacterial load in a murine
model of experimental colitis. Dig Dis Sci 55, 22112218.
Polancec, D. S., Munic Kos, V., Banjanac, M., Vrancic, M., Cuzic, S., Belamaric, D., et al.
(2012). Azithromycin drives in vitro GM-CSF/IL-4-induced differentiation of human
blood monocytes toward dendritic-like cells with regulatory properties. J Leukoc
Biol 91, 229243.
Pukander, J., & Rautianen, M. (1996). Penetration of azithromycin into middle ear effusions in acute and secretory otitis media in children. J Antimicrob Chemother
37(Suppl. C), 5361.
Raines, D. A., Yusuf, A., Jabak, M. H., Ahmed, W. S., Karcioglu, Z. A., & El-Yazigi, A. (1998).
Simultaneous high-performance liquid chromatography analysis of azithromycin and
two of its metabolites in human tears and plasma. Ther Drug Monit 20, 680684.
Ramsey, P. S., Vaules, M. B., Vasdev, G. M., Andrews, W. W., & Ramin, K. D. (2003). Maternal and transplacental pharmacokinetics of azithromycin. Am J Obstet Gynecol 188,
714718.
Randow, F., & Munz, C. (2012). Autophagy in the regulation of pathogen replication and
adaptive immunity. Trends Immunol 33, 475487.
Ray, W. A., Murray, K. T., Hall, K., Arbogast, P. G., & Stein, C. M. (2012). Azithromycin and
the risk of cardiovascular death. N Engl J Med 366, 18811890.
Renna, M., Schaffner, C., Brown, K., Shang, S., Tamayo, M. H., Hegyi, K., et al. (2011).
Azithromycin blocks autophagy and may predispose cystic brosis patients to mycobacterial infection. J Clin Invest 121, 35543563.
Restrepo, M. I., Mortensen, E. M., Waterer, G. W., Wunderink, R. G., Coalson, J. J., &
Anzueto, A. (2009). Impact of macrolide therapy on mortality for patients with severe
sepsis due to pneumonia. Eur Respir J 33, 153159.
Ribeiro, C. M., Hurd, H., Wu, Y., Martino, M. E., Jones, L., Brighton, B., et al. (2009).
Azithromycin treatment alters gene expression in inammatory, lipid metabolism,
and cell cycle pathways in well-differentiated human airway epithelia. PLoS One 4,
e5806.
Rodvold, K. A., Danziger, L. H., & Gotfried, M. H. (2003). Steady-state plasma and
bronchopulmonary concentrations of intravenous levooxacin and azithromycin in
healthy adults. Antimicrob Agents Chemother 47, 24502457.
Rohof, W. O., Bennink, R. J., de Ruigh, A. A., Hirsch, D. P., Zwinderman, A. H., &
Boeckxstaens, G. E. (2012). Effect of azithromycin on acid reux, hiatus hernia and
proximal acid pocket in the postprandial period. Gut 61, 16701677.
Saiman, L., Anstead, M., Mayer-Hamblett, N., Lands, L. C., Kloster, M., Hocevar-Trnka, J.,
et al. (2010). Effect of azithromycin on pulmonary function in patients with cystic brosis uninfected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA
303, 17071715.
Saiman, L., Marshall, B. C., Mayer-Hamblett, N., Burns, J. L., Quittner, A. L., Cibene, D. A.,
et al. (2003). Azithromycin in patients with cystic brosis chronically infected with
Pseudomonas aeruginosa: a randomized controlled trial. JAMA 290, 17491756.
Saint-Criq, V., Rufn, M., Rebeyrol, C., Guillot, L., Jacquot, J., Clement, A., et al. (2012).
Azithromycin fails to reduce inammation in cystic brosis airway epithelial cells.
Eur J Pharmacol 674, 16.
Sakito, O., Kadota, J., Kohno, S., Abe, K., Shirai, R., & Hara, K. (1996). Interleukin 1 beta,
tumor necrosis factor alpha, and interleukin 8 in bronchoalveolar lavage uid of patients with diffuse panbronchiolitis: a potential mechanism of macrolide therapy.
Respiration 63, 4248.
Salman, S., Rogerson, S. J., Kose, K., Grifn, S., Gomorai, S., Baiwog, F., et al. (2010). Pharmacokinetic properties of azithromycin in pregnancy. Antimicrob Agents Chemother
54, 360366.

243

244

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245

Sato, Y., Kaneko, K., & Inoue, M. (2007). Macrolide antibiotics promote the LPS-induced
upregulation of prostaglandin E receptor EP2 and thus attenuate macrolide suppression of IL-6 production. Prostaglandins Leukot Essent Fatty Acids 76, 181188.
Scaglione, F., Demartini, G., Dugnani, S., Arcidiacono, M. M., Pintucci, J. P., & Fraschini, F.
(1999). Interpretation of middle ear uid concentrations of antibiotics: comparison
between ceftibuten, cexime and azithromycin. Br J Clin Pharmacol 47, 267271.
Serisier, D. J., & Martin, M. L. (2011). Long-term, low-dose erythromycin in bronchiectasis
subjects with frequent infective exacerbations. Respir Med 105, 946949.
Shakeri-Nejad, K., & Stahlmann, R. (2006). Drug interactions during therapy with three
major groups of antimicrobial agents. Expert Opin Pharmacother 7, 639651.
Shepard, R. M., & Falkner, F. C. (1990). Pharmacokinetics of azithromycin in rats and dogs.
J Antimicrob Chemother 25(Suppl. A), 4960.
Shinkai, M., Henke, M. O., & Rubin, B. K. (2008). Macrolide antibiotics as immunomodulatory medications: proposed mechanisms of action. Pharmacol Ther 117, 393405.
Shinkai, M., Tamaoki, J., Kobayashi, H., Kanoh, S., Motoyoshi, K., Kute, T., et al. (2006).
Clarithromycin delays progression of bronchial epithelial cells from G1 phase to S
phase and delays cell growth via extracellular signal-regulated protein kinase suppression. Antimicrob Agents Chemother 50, 17381744.
Shorr, A. F., Zilberberg, M. D., Kan, J., Hoffman, J., Micek, S. T., & Kollef, M. H. (2013).
Azithromycin and survival in Streptococcus pneumoniae pneumonia: a retrospective
study. BMJ Open 3.
Singlas, E. (1995). Clinical pharmacokinetics of azithromycin. Pathol Biol (Paris) 43,
505511.
Skalsky, K., Yahav, D., Lador, A., Eliakim-Raz, N., Leibovici, L., & Paul, M. (2013). Macrolides
vs. quinolones for community-acquired pneumonia: meta-analysis of randomized
controlled trials. Clin Microbiol Infect 19, 370378.
Skerk, V., Krhen, I., Lisic, M., Begovac, J., Roglic, S., Skerk, V., et al. (2004). Comparative randomized pilot study of azithromycin and doxycycline efcacy in the treatment of
prostate infection caused by Chlamydia trachomatis. Int J Antimicrob Agents 24,
188191.
Skerk, V., Marekovic, I., Markovinovic, L., Begovac, J., Skerk, V., Barsic, N., et al. (2006).
Comparative randomized pilot study of azithromycin and doxycycline efcacy and
tolerability in the treatment of prostate infection caused by Ureaplasma urealyticum.
Chemotherapy 52, 911.
Skerk, V., Schonwald, S., Krhen, I., Banaszak, A., Begovac, J., Strugar, J., et al. (2003). Comparative analysis of azithromycin and ciprooxacin in the treatment of chronic prostatitis caused by Chlamydia trachomatis. Int J Antimicrob Agents 21, 457462.
Skerk, V., Schonwald, S., Krhen, I., Markovinovic, L., Barsic, B., Marekovic, I., et al. (2002).
Comparative analysis of azithromycin and clarithromycin efcacy and tolerability in
the treatment of chronic prostatitis caused by Chlamydia trachomatis. J Chemother
14, 384389.
Southern, K. W., Barker, P. M., Solis-Moya, A., & Patel, L. (2011). Macrolide antibiotics for
cystic brosis. Cochrane Database Syst Rev, CD002203.
Spagnolo, P., Fabbri, L. M., & Bush, A. (2013). Long-term macrolide treatment for chronic
respiratory disease. Eur Respir J 42, 239251.
Spyridaki, A., Raftogiannis, M., Antonopoulou, A., Tsaganos, T., Routsi, C., Baziaka, F., et al.
(2012). Effect of clarithromycin in inammatory markers of patients with ventilatorassociated pneumonia and sepsis caused by Gram-negative bacteria: results from a
randomized clinical study. Antimicrob Agents Chemother 56, 38193825.
Srivastava, P., Vardhan, H., Bhengraj, A. R., Jha, R., Singh, L. C., Salhan, S., et al. (2011).
Azithromycin treatment modulates the extracellular signal-regulated kinase mediated pathway and inhibits inammatory cytokines and chemokines in epithelial cells
from infertile women with recurrent Chlamydia trachomatis infection. DNA Cell Biol
30, 545554.
Stamatiou, R., Boukas, K., Paraskeva, E., Molyvdas, P. A., & Hatziefthimiou, A. (2010).
Azithromycin reduces the viability of human bronchial smooth muscle cells.
J Antibiot (Tokyo) 63, 7175.
Stamatiou, R., Paraskeva, E., Boukas, K., Gourgoulianis, K. I., Molyvdas, P. A., &
Hatziefthimiou, A. A. (2009). Azithromycin has an antiproliferative and autophagic
effect on airway smooth muscle cells. Eur Respir J 34, 721730.
Stamm, W. E., Hicks, C. B., Martin, D. H., Leone, P., Hook, E. W., III, Cooper, R. H., et al.
(1995). Azithromycin for empirical treatment of the nongonococcal urethritis syndrome in men. A randomized double-blind study. JAMA 274, 545549.
Stepanic, V., Kostrun, S., Malnar, I., Hlevnjak, M., Butkovic, K., Caleta, I., et al. (2011).
Modeling cellular pharmacokinetics of 14- and 15-membered macrolides with physicochemical properties. J Med Chem 54, 719733.
Stover, D. E., & Mangino, D. (2005). Macrolides: a treatment alternative for bronchiolitis
obliterans organizing pneumonia? Chest 128, 36113617.
Sugie, M., Asakura, E., Zhao, Y. L., Torita, S., Nadai, M., Baba, K., et al. (2004). Possible involvement of the drug transporters P glycoprotein and multidrug resistanceassociated protein Mrp2 in disposition of azithromycin. Antimicrob Agents
Chemother 48, 809814.
Sugiyama, K., Shirai, R., Mukae, H., Ishimoto, H., Nagata, T., Sakamoto, N., et al. (2007). Differing effects of clarithromycin and azithromycin on cytokine production by murine
dendritic cells. Clin Exp Immunol 147, 540546.
Suresh Babu, K., Kastelik, J., & Morjaria, J. B. (2013). Role of long term antibiotics in chronic
respiratory diseases. Respir Med 107, 800815.
Sutcliffe, J. A., & Leclerq, R. (2002). Mechanisms of resistance to macrolides, lincosamides,
and ketolides. In W. Schnfeld, & H. A. Kirst (Eds.), Macrolide Antibiotics
(pp. 281317). Basel: Birkhuser Verlag.
Svanstrom, H., Pasternak, B., & Hviid, A. (2013). Use of azithromycin and death from cardiovascular causes. N Engl J Med 368, 17041712.
Tabbara, K. F., al-Kharashi, S. A., al-Mansouri, S. M., al-Omar, O. M., Cooper, H., el-Asrar, A. M.,
et al. (1998). Ocular levels of azithromycin 116, 16251628.
Tait-Kamradt, A., Davies, T., Cronan, M., Jacobs, M. R., Appelbaum, P. C., & Sutcliffe, J.
(2000). Mutations in 23S rRNA and ribosomal protein L4 account for resistance in

pneumococcal strains selected in vitro by macrolide passage. Antimicrob Agents


Chemother 44, 21182125.
Tamaoki, J., Kadota, J., & Takizawa, H. (2004). Clinical implications of the immunomodulatory effects of macrolides. Am J Med 117(Suppl. 9A), 5S11S.
Tateda, K., Comte, R., Pechere, J. C., Kohler, T., Yamaguchi, K., & Van Delden, C. (2001).
Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrob
Agents Chemother 45, 19301933.
Terao, H., Asano, K., Kanai, K., Kyo, Y., Watanabe, S., Hisamitsu, T., et al. (2003). Suppressive activity of macrolide antibiotics on nitric oxide production by lipopolysaccharide
stimulation in mice. Mediators Inamm 12, 195202.
Tessmer, A., Welte, T., Martus, P., Schnoor, M., Marre, R., & Suttorp, N. (2009). Impact of
intravenous {beta}-lactam/macrolide versus {beta}-lactam monotherapy on mortality in hospitalized patients with community-acquired pneumonia. J Antimicrob
Chemother 63, 10251033.
Thorpe, E. M., Jr., Stamm, W. E., Hook, E. W., III, Gall, S. A., Jones, R. B., Henry, K., et al.
(1996). Chlamydial cervicitis and urethritis: single dose treatment compared with
doxycycline for seven days in community based practises. Genitourin Med 72, 9397.
Tomaskovic, L., Komac, M., Makaruha Stegic, O., Munic, V., Ralic, J., Stanic, B., et al. (2013).
Macrolactonolides: a novel class of anti-inammatory compounds. Bioorg Med Chem
21, 321332.
Tomazic, J., Kotnik, V., & Wraber, B. (1993). In vivo administration of azithromycin affects
lymphocyte activity in vitro. Antimicrob Agents Chemother 37, 17861789.
Tong, J., Liu, Z. C., & Wang, D. X. (2011). Azithromycin acts as an immunomodulatory
agent to suppress the expression of TREM-1 in Bacillus pyocyaneus-induced sepsis.
Immunol Lett 138, 137143.
Tresse, E., Salomons, F. A., Vesa, J., Bott, L. C., Kimonis, V., Yao, T. P., et al. (2010). VCP/p97 is
essential for maturation of ubiquitin-containing autophagosomes and this function is
impaired by mutations that cause IBMPFD. Autophagy 6, 217227.
Tsai, W. C., Hershenson, M. B., Zhou, Y., & Sajjan, U. (2009). Azithromycin increases survival and reduces lung inammation in cystic brosis mice. Inamm Res 58, 491501.
Tsai, W. C., Rodriguez, M. L., Young, K. S., Deng, J. C., Thannickal, V. J., Tateda, K., et al.
(2004). Azithromycin blocks neutrophil recruitment in Pseudomonas endobronchial
infection. Am J Respir Crit Care Med 170, 13311339.
Tsai, W. C., & Standiford, T. J. (2004). Immunomodulatory effects of macrolides in the
lung: lessons from in-vitro and in-vivo models. Curr Pharm Des 10, 30813093.
Tyteca, D., Schanck, A., Dufrene, Y. F., Deleu, M., Courtoy, P. J., Tulkens, P. M., et al. (2003).
The macrolide antibiotic azithromycin interacts with lipids and affects membrane organization and uidity: studies on LangmuirBlodgett monolayers, liposomes and
J774 macrophages. J Membr Biol 192, 203215.
Tyteca, D., Van Der Smissen, P., Mettlen, M., Van Bambeke, F., Tulkens, P. M., MingeotLeclercq, M. P., et al. (2002). Azithromycin, a lysosomotropic antibiotic, has distinct
effects on uid-phase and receptor-mediated endocytosis, but does not impair
phagocytosis in J774 macrophages. Exp Cell Res 281, 86100.
Tyteca, D., Van Der Smissen, P., Van Bambeke, F., Leys, K., Tulkens, P. M., Courtoy, P. J., et al.
(2001). Azithromycin, a lysosomotropic antibiotic, impairs uid-phase pinocytosis in
cultured broblasts. Eur J Cell Biol 80, 466478.
Valery, P. C., Morris, P. S., Byrnes, C. A., Grimwood, K., Torzillo, P. J., Bauert, P. A., et al. (2013).
Long-term azithromycin for Indigenous children with non-cystic-brosis bronchiectasis or chronic suppurative lung disease (Bronchiectasis Intervention Study):
a multicentre, double-blind, randomised controlled trial. Lancet Respir Med 1, 610620.
Van Bambeke, F., Montenez, J. P., Piret, J., Tulkens, P. M., Courtoy, P. J., & Mingeot-Leclercq,
M. P. (1996). Interaction of the macrolide azithromycin with phospholipids. I. Inhibition of lysosomal phospholipase A1 activity. Eur J Pharmacol 314, 203214.
van Delden, C., Kohler, T., Brunner-Ferber, F., Francois, B., Carlet, J., & Pechere, J. C. (2012).
Azithromycin to prevent Pseudomonas aeruginosa ventilator-associated pneumonia
by inhibition of quorum sensing: a randomized controlled trial. Intensive Care Med
38, 11181125.
van Eijk, A. M., & Terlouw, D. J. (2011). Azithromycin for treating uncomplicated malaria.
Cochrane Database Syst Rev, CD006688.
Vanaudenaerde, B. M., Meyts, I., Vos, R., Geudens, N., De Wever, W., Verbeken, E. K., et al.
(2008). A dichotomy in bronchiolitis obliterans syndrome after lung transplantation
revealed by azithromycin therapy. Eur Respir J 32, 832843.
Vanaudenaerde, B. M., Vos, R., Meyts, I., De Vleeschauwer, S. I., Verleden, S. E., WidyastutiWillems, A., et al. (2008). Macrolide therapy targets a specic phenotype in respiratory medicine: from clinical experience to basic science and back. Inamm Allergy
Drug Targets 7, 279287.
Vanaudenaerde, B. M., Wuyts, W. A., Geudens, N., Dupont, L. J., Schoofs, K., Smeets, S., et al.
(2007). Macrolides inhibit IL17-induced IL8 and 8-isoprostane release from human
airway smooth muscle cells. Am J Transplant 7, 7682.
Verleden, G. M., Vanaudenaerde, B. M., Dupont, L. J., & Van Raemdonck, D. E. (2006).
Azithromycin reduces airway neutrophilia and interleukin-8 in patients with bronchiolitis obliterans syndrome. Am J Respir Crit Care Med 174, 566570.
Verleden, S. E., Vandooren, J., Vos, R., Willems, S., Dupont, L. J., Verleden, G. M., et al.
(2011). Azithromycin decreases MMP-9 expression in the airways of lung transplant
recipients. Transpl Immunol 25, 159162.
Verleden, S. E., Vos, R., Mertens, V., Willems-Widyastuti, A., De Vleeschauwer, S. I., Dupont, L. J.,
et al. (2011). Heterogeneity of chronic lung allograft dysfunction: insights from protein
expression in broncho alveolar lavage. J Heart Lung Transplant 30, 667673.
Videler, W. J., Badia, L., Harvey, R. J., Gane, S., Georgalas, C., van der Meulen, F. W., et al.
(2011). Lack of efcacy of long-term, low-dose azithromycin in chronic
rhinosinusitis: a randomized controlled trial. Allergy 66, 14571468.
Villarino, N., & Martin-Jimenez, T. (2013). Pharmacokinetics of macrolides in foals. J Vet
Pharmacol Ther 36, 113.
Vos, R., Vanaudenaerde, B. M., Ottevaere, A., Verleden, S. E., De Vleeschauwer, S. I.,
Willems-Widyastuti, A., et al. (2010). Long-term azithromycin therapy for bronchiolitis obliterans syndrome: divide and conquer? J Heart Lung Transplant 29, 13581368.

M.J. Parnham et al. / Pharmacology & Therapeutics 143 (2014) 225245


Vos, R., Vanaudenaerde, B. M., Verleden, S. E., De Vleeschauwer, S. I., Willems-Widyastuti,
A., Van Raemdonck, D. E., et al. (2011). A randomised controlled trial of azithromycin
to prevent chronic rejection after lung transplantation. Eur Respir J 37, 164172.
Vrancic, M., Banjanac, M., Nujic, K., Bosnar, M., Murati, T., Munic, V., et al. (2012).
Azithromycin distinctively modulates classical activation of human monocytes
in vitro. Br J Pharmacol 165, 13481360.
Wang, L., Kitaichi, K., Hui, C. S., Takagi, K., Takagi, K., Sakai, M., et al. (2000). Reversal of
anticancer drug resistance by macrolide antibiotics in vitro and in vivo. Clin Exp
Pharmacol Physiol 27, 587593.
Wang, P. L. (2010). Roles of oral bacteria in cardiovascular diseasesfrom molecular
mechanisms to clinical cases: treatment of periodontal disease regarded as biolm
infection: systemic administration of azithromycin. J Pharmacol Sci 113, 126133.
Wierzbowski, A. K., Hoban, D. J., Hisanaga, T., DeCorby, M., & Zhanel, G. G. (2006). The use
of macrolides in treatment of upper respiratory tract infections. Curr Allergy Asthma
Rep 6, 171181.
Wildfeuer, A., Reisert, I., & Laufen, H. (1993). Uptake and subcellular distribution of
azithromycin in human phagocytic cells. Demonstration of the antibiotic in neutrophil polymorphonuclear leucocytes and monocytes by autoradiography and electron
microscopy. Arzneimittelforschung 43, 484486.
Willems-Widyastuti, A., Vanaudenaerde, B. M., Vos, R., Dilisen, E., Verleden, S. E., De
Vleeschauwer, S. I., et al. (2013). Azithromycin attenuates broblast growth factors
induced vascular endothelial growth factor via p38(MAPK) signaling in human airway smooth muscle cells. Cell Biochem Biophys 67, 331339.
Willenborg, M., Schmidt, C. K., Braun, P., Landgrebe, J., von Figura, K., Saftig, P., et al.
(2005). Mannose 6-phosphate receptors, Niemann-Pick C2 protein, and lysosomal
cholesterol accumulation. J Lipid Res 46, 25592569.
Wilms, E. B., Touw, D. J., & Heijerman, H. G. (2006). Pharmacokinetics of azithromycin in
plasma, blood, polymorphonuclear neutrophils and sputum during long-term therapy in patients with cystic brosis. Ther Drug Monit 28, 219225.
Wilson, B. Z., Anzueto, A., Restrepo, M. I., Pugh, M. J., & Mortensen, E. M. (2012). Comparison of two guideline-concordant antimicrobial combinations in elderly patients hospitalized with severe community-acquired pneumonia. Crit Care Med 40, 23102314.

Wong, C., Jayaram, L., Karalus, N., Eaton, T., Tong, C., Hockey, H., et al. (2012).
Azithromycin for prevention of exacerbations in non-cystic brosis bronchiectasis
(EMBRACE): a randomised, double-blind, placebo-controlled trial. Lancet 380,
660667.
Wuyts, W. A., Willems, S., Vos, R., Vanaudenaerde, B. M., De Vleeschauwer, S. I., Rinaldi, M.,
et al. (2010). Azithromycin reduces pulmonary brosis in a bleomycin mouse model.
Exp Lung Res 36, 602614.
Yamada, K., Yanagihara, K., Kaku, N., Harada, Y., Migiyama, Y., Nagaoka, K., et al. (2013).
Azithromycin attenuates lung inammation in a mouse model of ventilatorassociated pneumonia by multidrug-resistant Acinetobacter baumannii. Antimicrob
Agents Chemother 57, 38833888.
Yamamoto, H., Naito, Y., Okano, M., Kanazawa, T., Takematsu, H., & Kozutsumi, Y.
(2011). Sphingosylphosphorylcholine and lysosulfatide have inverse regulatory
functions in monocytic cell differentiation into macrophages. Arch Biochem
Biophys 506, 8391.
Yamauchi, K., Shibata, Y., Kimura, T., Abe, S., Inoue, S., Osaka, D., et al. (2009).
Azithromycin suppresses interleukin-12p40 expression in lipopolysaccharide and
interferon-gamma stimulated macrophages. Int J Biol Sci 5, 667678.
Yamaya, M., Azuma, A., Takizawa, H., Kadota, J., Tamaoki, J., & Kudoh, S. (2012).
Macrolide effects on the prevention of COPD exacerbations. Eur Respir J 40,
485494.
Yanagihara, K., Izumikawa, K., Higa, F., Tateyama, M., Tokimatsu, I., Hiramatsu, K., et al.
(2009). Efcacy of azithromycin in the treatment of community-acquired pneumonia, including patients with macrolide-resistant Streptococcus pneumoniae infection.
Intern Med 48, 527535.
Yang, M., Dong, B. R., Lu, J., Lin, X., & Wu, H. M. (2013). Macrolides for diffuse
panbronchiolitis. Cochrane Database Syst Rev 2, CD007716.
Yasuda, K., Lan, L. B., Sanglard, D., Furuya, K., Schuetz, J. D., & Schuetz, E. G. (2002). Interaction of cytochrome P450 3A inhibitors with P-glycoprotein. J Pharmacol Exp Ther
303, 323332.
Yeung, T., & Grinstein, S. (2007). Lipid signaling and the modulation of surface charge
during phagocytosis. Immunol Rev 219, 1736.

245

Potrebbero piacerti anche