Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Advanced Drug Delivery Reviews 57 (2005) 1539 – 1550

www.elsevier.com/locate/addr

Biofilms and antibiotic therapy: Is there a role for combating


bacterial resistance by the use of novel drug delivery systems?
Anthony W. Smith*
Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
Received 3 December 2004; accepted 11 April 2005
Available online 13 June 2005

Abstract

The conventional view of antibiotic resistance is one where bacteria exhibit significantly reduced susceptibility to
antimicrobials in laboratory tests by mechanisms such as altered drug uptake, altered drug target and drug inactivation. Whilst
these mechanisms undoubtedly make a major contribution to antibiotic failure in the clinic, the phenomenon of clinical failure
in spite of sensitivity in laboratory tests is also well recognised. It is in this context that attention has focussed on bacteria
growing as adherent biofilms, not only as the mode of growth of device-related infections associated for example with artificial
joints and venous catheters, but also with other chronic infections such as those occurring in the respiratory tract. Growth as a
biofilm almost always leads to a significant decrease in susceptibility to antimicrobial agents compared with cultures grown in
suspension and, whilst there is no generally agreed mechanism for the resistance of biofilm bacteria, it is largely phenotypic.
That is, when biofilm bacteria are grown in conventional laboratory suspension culture they become susceptible to anti-
microbials. A number of elements in the process of biofilm formation have been studied as targets for novel drug delivery
technologies. These include surface modification of devices to reduce bacterial attachment and biofilm development as well as
incorporation of antimicrobials—again to prevent colonisation. Electrical approaches have been used either to release
antimicrobials from device surfaces or to drive antimicrobials through the biofilm. Other technologies not specifically focussed
on biofilms include aerosolized delivery of antibiotics to the lung and formulation into liposome and polymer-based vehicles.
Liposomal systems have been widely studied, either to target antibiotics to the surface of bacterial biofilms, or by virtue of their
property of being taken up cells of the reticuloendothelial system, to target antibiotics towards intracellular bacteria. Many
polymer-based carrier systems have also been proposed, including those based on biodegradable polymers such as poly(lactide-
co-glycolide) as well as thermoreversible hydrogels. Their contribution to the prevention or resolution of infection is reviewed.
D 2005 Elsevier B.V. All rights reserved.

Keywords: Biofilms; Phenotypic resistance; Material modification; Polyurethanes; Iontophoresis; Bio-electric effect; Pulsed electromagnetic
fields; Ultrasound; Photodynamic enhancement; Liposomes; Pegylated liposomes; Biodegradable microspheres; Poly(lactide-co-glycolide);
Hydroxyapatite; Halloysite; Electrospun fibrous scaffolds; Thermoreversible gels; Infection responsive systems; Aerosols

* Tel.: +44 1225 383101; fax: +44 1225 386114.


E-mail address: a.w.smith@bath.ac.uk.

0169-409X/$ - see front matter D 2005 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2005.04.007
1540 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1540
2. Biofilms in infection. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1540
2.1. Biofilm resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1541
3. A role for drug delivery systems? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1542
3.1. Anti-biofilm approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1542
3.1.1. Material modification to prevent biofilm formation . . . . . . . . . . . . . . . . . . . . . . . 1542
3.1.2. Electrical enhancement of antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . 1543
3.1.3. Ultrasound enhancement of antimicrobial transport . . . . . . . . . . . . . . . . . . . . . . . 1543
3.1.4. Photodynamic approaches to biofilm treatment . . . . . . . . . . . . . . . . . . . . . . . . . 1544
3.2. Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1544
3.2.1. Liposomal delivery to biofilms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1544
3.2.2. Liposomal delivery in intracellular infection . . . . . . . . . . . . . . . . . . . . . . . . . . 1544
3.2.3. Stealth liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1545
3.3. Polymer-based antimicrobial delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1545
3.3.1. Implantable matrices, microparticles, fibrous scaffolds and thermoreversible gels.. . . . . . . 1545
3.3.2. Delivery on demand—an infection-responsive system . . . . . . . . . . . . . . . . . . . . . 1547
3.4. Aerosol delivery to the lung . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1547
4. Discussion and future aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1547
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1547
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1547

1. Introduction ena such as biofouling, it is now clear that microbial


biofilms are largely responsible for the recalcitrance of
In addressing the question of whether use of novel many infections to conventional antimicrobial therapy
drug delivery systems can overcome antibiotic resis- [1,2]. A microbial biofilm is broadly defined as ad-
tance, it is important to view resistance in the clinical herent microorganisms within a polymeric matrix,
context. It may not be an all-or-nothing response and typically comprising exopolysaccharide that develops
decreases in susceptibility do not necessarily mean into a complex community [3]. The composition is
clinical failure if sufficient antibiotic can be targeted often heterogeneous with water channels occurring
to the infection. It is in this scenario that novel drug between glycocalyx-enclosed microorganisms in
delivery systems may have some benefit. Unfortunate- stalk- or mushroom-like structures. The structure is
ly, a different scenario typically prevails in the clinic also a dynamic one and may include single or multiple
where treatment fails in spite of antibiotic sensitivity microbial species [4]. Cases of biofilm infection in-
in laboratory tests. In other words, clinical failure is clude the well-known examples of device-related
often due not to infections with bacteria harbouring infections such as those associated with artificial
mechanisms resulting in high-level antibiotic resis- joints, prosthetic heart valves and catheters. Even
tance, but rather to bacteria that are phenotypically with the use of perioperative antimicrobial prophylax-
resistant in vivo. is and a laminar air-flow surgical environment, the
risk of intraoperative infection is still around 1% for
hip and shoulder replacement and 2% after knee
2. Biofilms in infection replacement [5]. With more than 200,000 hip replace-
ments and 200,000 knee replacements each year in the
There is now widespread recognition of the contri- United States alone, the healthcare costs are high.
bution of biofilms to human infection. Previously Recent surveys also indicate that catheter-associated
thought to be the concern only of industrial and bacteremia following catheter-related infection, is by
environmental microbiologists interested in phenom- far the leading cause of nosocomial bloodstream in-
A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550 1541

fection in intensive care units [6]. Many chronic biofilm resistance [9,10,17–19] and a number of
infections, not related to medical devices, are now delivery strategies have been used in an attempt to
being attributed either to bacteria not growing and drive agents through biofilms. However, given that
being relatively dormant or growing slowly as bio- in many cases biofilms consist of stacks of cells
masses or adherent biofilms on mucosal surfaces. with aqueous channels flowing in between, impene-
trability seems highly unlikely [20], a finding con-
2.1. Biofilm resistance firmed with Klebsiella pneumoniae biofilm [21].
Commonly, the entire biofilm is coated with a com-
Growth as a biofilm almost always leads to a large plex hydrophilic polymer, the glycoclayx [22] that is
increase in resistance to antimicrobial agents com- typically anionic in nature. Where the antimicrobial
pared with cultures grown in suspension (planktonic) agent reacts chemically with exopolymer or is
in conventional liquid media, with up to 1000-fold adsorbed to it, then the net effect is that of having
decreases in susceptibility reported [7,8]. Currently, the appearance of a penetration barrier. There will
there is no generally agreed mechanism to account for be a similar effect if antimicrobials adsorb onto
the broad resistance of microbial biofilms [2,9–12]. cells, perhaps dead ones, in the outer parts of the
However, the general resistance of microbial biofilms biofilm.
is largely phenotypic. When biofilm organisms are As reviewed elsewhere, resistance can be due to
grown in conventional laboratory planktonic culture production of inactivating enzymes and there is evi-
they become susceptible to antimicrobials. Thus, the dence that the relatively large amounts of antibiotic-
resistance properties of biofilm bacteria mirror the inactivating enzymes such as h-lactamases which ac-
common finding of clinical failure of antibiotic ther- cumulate within the glycocalyx produce concentration
apy in spite of antibiotic susceptibility in laboratory gradients can protect underlying cells [23,24]. In con-
tests. Eradication of infection by antibiotic treatment trast, in other systems, enzyme inactivation appears
requires elimination of all the bacteria, typically not to contribute to the resistance of biofilm bacteria
assisted by the host defences. Specifically, biofilm [21].
resistance can be determined by the susceptibility of Efflux has also been reviewed elsewhere and the
the most resistant cell. There is significant heteroge- contribution of efflux to antimicrobial resistance of
neity within biofilms and it is not the case that all cells biofilms is uncertain. In one study, none of the four
within a biofilm are always highly resistant [9,13]. multidrug efflux pump systems present in the ge-
But the most resistant members of a biofilm popula- nome of Pseudomonas aeruginosa contributed to
tion are typically orders of magnitude more resistant biofilm resistance [25]. Recent work has highlighted
than similar members of a planktonic population. the contributions of oxygen deprivation and anaer-
Subculture often fails to show the existence of resis- obic growth to antibiotic resistance. There is evi-
tant mutants, but evidence is accumulating for the dence of P. aeruginosa growing anaerobically
contribution of persister cells to biofilm tolerance to within airway mucus of cystic fibrosis patients
antimicrobials [14]. Recent work now offers a mech- [26] and microelectrode studies on colony biofilms
anistic explanation for Escherichia coli persisters that indicated that oxygen only penetrated to approxi-
neither grow nor die in the presence of bactericidal mately 25% of the depth of the biofilm [27]. More-
agents and so are multidrug tolerant [15]. over, large anoxic regions developed as the biofilm
A full review of biofilm resistance is beyond the aged and regions of active protein synthesis oc-
scope of this work and it is noteworthy that to date curred only in a narrow band at the air interface
compelling evidence that any of the well characterised [28]. When challenged with antimicrobials, 4-h-old
mechanisms are uniquely responsible is lacking colony biofilms growing in the presence of air were
[11,16]. The focus here will be a brief overview of susceptible, however similar aged biofilms grown
drug penetration into the biofilm, antibiotic inactivat- anaerobically were much less susceptible [27]. The
ing enzymes, efflux and oxygen deprivation. authors calculated that oxygen limitation could ex-
Many papers have investigated the possible lack plain 70% or more of the protection of old biofilm
of antimicrobial penetration as an explanation of cells.
1542 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

3. A role for drug delivery systems? the failure of a mutant of Staphylococcus aureus
bearing an increased negative charge due to the lack
A number of the key elements in the infectious of d-alanine esters in its teichoic acids, to colonise
process and formation of biofilm have been proposed glass or polystyrene surfaces [30]. The observation
for application of novel technologies and drug deliv- that surfaces immediately become coated with a con-
ery systems—prevention of colonisation and biofilm ditioning film can thwart coating-based strategies to
formation, accumulation at the biofilm surface and reduce infection resulting from haematogenous seed-
drug penetration into the biofilm (Fig. 1). Recent ing of medical devices such as intravascular catheters.
developments will be reviewed here. Given the in- Put simply, the coating becomes coated. However,
creasing use of relatively invasive medical and surgi- given that many catheter-related infections are due
cal procedures, the material properties of medical to skin organisms acquired at the time of catheter
devices have received much attention as have strate- insertion, then anti-colonisation strategies are still
gies to target antimicrobials to device-related infec- worth exploring. For example, surfaces containing
tions. Dental plaque and oral hygiene is another immobilised long-chain N-alklyated polyvinylpyri-
common therapeutic target and there is now a consid- dines and structurally unrelated N-alklyated polyethy-
erable body of work using carrier systems to target lenimines were lethal to S. aureus, S. epidermidis, P.
antibiotics against intracellular infections. aeruginosa and E. coli. The structure–activity analy-
sis revealed that for surfaces to be bactericidal, the
3.1. Anti-biofilm approaches immobilised long polymeric chains have to be hydro-
phobic, but not excessively so, and positively charged
3.1.1. Material modification to prevent biofilm [31]. There are many instances where broad-spectrum
formation antimicrobials have been incorporated into the device
An unadulterated surface does not exist. Any sur- [32–36]. These are then eluted with the aim of pre-
face, synthetic or otherwise, is coated with consti- venting biofilm formation by killing early colonising
tuents of the local environment: first water and bacteria. As noted by others, such a strategy is not
electrolytes, then organic material. This conditioning without its problems [37]. Sufficient antibiotic must
film often exists before the arrival of any microorgan- be incorporated for the duser-lifetimeT of the device
isms. Firstly, there is a weak and reversible contact and such incorporation must not damage the proper-
between microbe and conditioning film resulting from ties of the material, for example, lubrication, lifetime
Brownian motion, gravitation, diffusion, or microbial and host compatibility. There is also the nagging
motility and involving electrostatic interactions [29]. concern that low levels of antimicrobials could favour
This interaction is a function of the microbial cell acquisition of antibiotic resistant organisms [38]. Wor-
surface and the nature of the adherent surface. The ryingly, in staphylococcal species commonly associ-
importance of electrostatic charge is highlighted by ated with device-related infections, sub-inhibitory

Carrier systems to Enhancement of drug


target antimicrobials to penetration into biofilm
biofilm surface

Development into
Planktonic cells
glycocalyx-enclosed
attach to surface
biofilm

Surface modification Adsorption of Antimicrobial


to reduce adhesion/ antimicrobials impregnated matrices
biofilm development onto surface

Fig. 1. Anti-biofilm strategies.


A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550 1543

concentrations of tetracycline and quinupristin–dalfo- 3.1.2. Electrical enhancement of antimicrobial


pristin enhanced biofilm development by increasing activity
expression of the intercellular adhesion ica locus [39]. Electro-chemical approaches have been proposed
Alternative approaches to antibiotic incorporation both as a means to prevent biofilm formation and also
have also been used. For example, rifampin and to enhance the activity of antimicrobials against estab-
amoxicillin have been adsorbed on polyurethanes lished biofilms—the bioelectric effect. For example,
exhibiting acidic or basic properties, the binding af- an iontophoretic approach has been taken where a
finity increasing with the introduction of polymer low-current power source is used to drive the release
side-chain functional groups and with matrix hydro- of antimicrobial silver ions from the device [44]. The
philicity. The zone of inhibition from amoxicillin- bioelectric effect refers to the concurrent application
coated polymers lasted only a few hours, whereas of antibiotics and a weak electric field to kill bacteria.
that from the rifampin-coated polymer lasted several With application of direct current electric fields be-
months [40]. Another approach has been to incorpo- tween 1.5 and 20 V/cm, the concentrations of anti-
rate drugs into the backbone of polymers as a main biotics needed to be effective against biofilm bacteria
chain monomer. Biodegradable polyurethanes have fell from approximately 5000 times to 4 times greater
been synthesised with the fluoroquinolone antibiotic than those necessary for planktonic bacteria in the
ciprofloxacin that is released upon the action of the absence of electricity [45]. Electrolytic generation of
macrophage-derived enzyme cholesterol esterase. oxygen may be partly responsible for this bioelectric
Since this enzyme recognises hydrophobic moieties, effect, at least with the aminoglycoside antibiotic
drug release could be achieved by formulating poly- tobramycin against P. aeruginosa biofilm [46]. En-
urethanes synthesised with 1,12 diisocyanatodode- hanced antibiotic activity has also been shown using
cane with long hydrophobic monomers immediately pulsed electromagnetic fields (PEMF). Here, PEMF
adjacent to the ciprofloxacin [41]. An alternative ap- enhanced the activity of gentamicin against 5-day
proach has been development of a liposomal hydrogel biofilms of S. epidermidis, although there was no
that reduces bacterial adhesion to silicone catheter significant effect when gentamicin was replaced
material. Here, liposomes containing ciprofloxacin with vancomycin [47].
are sequestered within a poly(ethylene glycol)-gelatin
hydrogel. Bacterial adhesion was completely inhibited 3.1.3. Ultrasound enhancement of antimicrobial
on catheter surfaces throughout a 7-day adhesion transport
assay [42]. Emergence of multi-drug resistant patho- Ultrasound has been used to enhance antibiotic
gens is always a risk when antibiotics are used both in transport through biofilms. For example, pulsed ultra-
prophylaxis and long-term therapy as well as in con- sound over 24 h enhanced the killing by gentamicin of
ventional acute therapy. Hence, there is interest in E. coli biofilms on polyethylene discs implanted sub-
inhibiting biofilm formation on polymer surfaces cutaneously into rabbits without damage to the skin
using novel compounds that are not otherwise used. [48]. Similar results have been reported for enhanced
One such compound is usnic acid, a secondary lichen vancomycin activity against S. epidermidis biofilms in
metabolite that possesses antimicrobial activity vivo, although longer treatment times were required
against a number of gram-positive bacteria, including [49]. Low-frequency ultrasound (70 kHz) of low
S. aureus, Enterococcus faecalis and E. faecium. acoustic intensity increased the growth rate of S.
When loaded into a modified polyurethane and placed epidermidis, P. aeruginosa and E. coli grown on
in infection-simulating flow cells, confocal laser scan- polyethylene surfaces and it was hypothesized that
ning microscopy indicated that whilst adhesion of S. ultrasound increases the transport of oxygen and nutri-
aureus occurred, usnic acid inhibited development of ents to the cells [50]. Such increased growth would
biofilm [43]. P. aeruginosa biofilm did form on usnic- likely enhance susceptibility to antibiotics. Using a
acid-coated polymer, although its morphology dif- colony biofilm model where diffusion of antibiotic
fered from the biofilm formed on untreated polymer, through the biofilm could be measured, insonation
leading the authors to suggest that signalling pathways of E. coli or P. aeruginosa biofilms significantly
within the biofilm may have been perturbed. increased transport of gentamicin across the biofilm,
1544 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

whereas in the absence of ultrasound diffusion either decylammonium bromide (DDAB) or anionic
did not occur or did so at a much slower rate [51]. liposomes substituting DMPC with phosphatidylino-
sitol (PI), they noted that each bacterium in the bio-
3.1.4. Photodynamic approaches to biofilm treatment film adsorbed independently and that the extent of
Photodynamic approaches have been proposed as a adsorption of anionic liposomes was smaller. Interest-
means to overcome resistance and to break down ingly, when targeting mixed biofilms of Streptococcus
biofilms [52]. Photosensitising drugs produce reactive sanguis and S. salivarius with liposomes loaded with
oxygen species that are difficult for the microorgan- the bactericide triclosan, anionic liposomes were most
ism to defend against. Necessarily, applications are effective against S. sanguis, but relatively ineffective
limited to those sites where light can reach. For this against S. salivarius [65]. An additional approach has
reason, the focus has been on pathogens associated been to load antibacterials into liposomes adsorbed on
with the skin and with the oral cavity. For example, the surface of zinc citrate particles, used in toothpaste
biofilms of the oral pathogen Actinomyces viscosus formulations, to produce solid supported vesicles
have been exposed to laser light at 666 nm in the (SSVs) containing either triclosan or aqueous-soluble
presence of methylene blue. Confocal microscopy penicillin-G. Anionic liposomes were prepared by
revealed that a single photomechanical wave in- incorporation of PI into DMPC liposomes and cation-
creased the penetration of methylene blue by 75% ic liposomes were prepared by incorporation of
and enhanced the photodestruction of the biofilm DDAB and cholesterol into DMPC. Whilst zinc cit-
[53]. Similar findings have been obtained using rate is itself antibacterial, it was noted that particles
multi-species biofilms of oral bacteria irradiated with and empty liposomes had an additional or synergistic
light from a helium/neon laser in the presence of effect, whereas particles and liposomally encapsulated
toluidine blue where greater than 95% of biofilm antimicrobials had an inhibitory effect on each other
bacteria were killed [54]. Comparisons of growth against S. oralis biofilms [63]. Other oral hygiene
phase and extracellular slime on the photodynamic approaches have included liposomal encapsulation
inactivation of S. epidermidis and S. aureus indicated of the enzyme glucose oxidase and horse radish per-
that slime production and stationary phase, both char- oxidase, that generate hydrogen peroxide and oxya-
acteristics of biofilm infections, were obstacles to this cids in the presence of their substrates. They were
therapy. However, use of polylysine-based cationic effective against S. gordonii biofilms in a manner
photosensitisers overcame some of the growth phase dependent upon liposome–biofilm and substrate–bio-
effects [55]. film incubation times [66].

3.2. Liposomes 3.2.2. Liposomal delivery in intracellular infection


The uptake of liposomes by cells of the reticulo-
Liposomes are attractive as drug delivery/targeting endothelial system has been exploited to target anti-
vehicles by virtue of their compatibility with biolog- biotics to those intracellular sites where parasitic
ical constituents and the range and extent of pay loads bacteria reside, and by virtue of sustained release
that they can carry. In the context of antibiotics and properties, extend the half-life of the drug in the
treatment of infection, they have been studied for their body. Ciprofloxacin has been incorporated with high
ability to concentrate agents at biofilm interfaces efficiency into DSPC/cholesterol liposomes and ex-
[56,57] and also to be taken up into cells harbouring amined in a mouse model of Francisella tularensis
intracellular pathogens [58–62]. [67]. Intravenous injection of liposome-encapsulated
ciprofloxacin resulted in increased drug retention in
3.2.1. Liposomal delivery to biofilms the lungs, liver and spleen compared with that of free
Jones and colleagues have reported extensive stud- drug. Aerosolized liposomal ciprofloxacin gave com-
ies of the interaction between liposomes and bacterial plete protection against a lethal pulmonary infection
biofilms [56,57,63–66]. For example, using either of F. tularensis, whereas free ciprofloxacin was inef-
cationic liposomes comprising dimyristoylphosphati- fective [67]. Caution should be exercised in extrapo-
dylcholine (DMPC), cholesterol and dimethlydiocta- lating data as it is clear that liposomal efficacy is
A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550 1545

dependent on the infecting organism. Liposome-en- infections such as tuberculosis and brucellosis. Small
capsulated ciprofloxacin, delivered intravenously, has biodegradable microspheres are useful alternatives to
been compared with free drug in a rat model of S. liposomes for targeting drugs to the monocyte–mac-
pneumoniae pneumonia and, whilst serum and lung rophage system. They tend not to suffer from the same
lavage levels were higher (peak and area under curve), difficulties of low encapsulation efficiency and stabil-
survival rates were similar [68]. ity on storage typically exhibited by liposomal for-
An interesting development of the liposomal con- mulations. Biodegradable microspheres prepared from
cept has been the use of pH-sensitive liposomes in a poly(lactide) (PLA) and its co-polymers with glyco-
murine salmonellosis model [69]. Here, gentamicin lide (PLGA) can release encapsulated drugs in a
encapsulated in liposomes including a pH-sensitive controlled way, depending on the method of microen-
lipid fusion between unsaturated PE and N-succinyl- capsulation and the physico-chemical properties of the
dioleyl-PE gave 153- and 437-fold greater drug levels polymer and drug.
in the liver and spleen, respectively, compared with
free drug. Overall, liposomal delivery was associated 3.3.1. Implantable matrices, microparticles, fibrous
with 10,000-fold greater activity than that of free scaffolds and thermoreversible gels
drug. Prevention and treatment of osteomyelitis, particu-
larly associated with orthopaedic implant surgery,
3.2.3. Stealth liposomes have been the focus of many studies. Systems
Somewhat counter-intuitively, stealth approaches implanted at the same time as the prosthesis may be
have been adopted for delivering antibiotics. Pegy- either non-biodegradable or biodegradable. Poly-
lated long-circulating liposomes loaded with gentami- methylmethacrylate beads impregnated with gentami-
cin were superior to free gentamicin in a rat model of cin have been available for about 20 years, however
K. pneumoniae unilateral pneumonia/septicaemic since they are non-biodegradable they have to be
[70]. Studies in vitro have also confirmed that pegyla- removed, usually about 4 weeks after insertion [77].
tion of liposomes reduced their affinity for S. aureus A number of osteoconductive/biodegradable alterna-
biofilms [64]. Here, they found that liposomes pre- tives have been studied, including calcium phosphates
pared from the phospholipids DMPC, dipalmitoyl PC such as hydroxyapatite whose chemical composition
and distearoyl PC containing DDAB (cationic) or PI is similar to the bone mineral phase. Studies with
(anionic) and variable amounts of dipalmitoylpho- ciprofloxacin incorporated into hydroxyapatite and
sphatidylethanolamine bonded to PEG of molecular poly(d,l-lactide) formulations implanted in the
mass 2000 (DPPE-PEG-2000), exhibited decreasing femur of rabbits, indicated that therapeutic bone levels
electrophoretic mobilities and zeta potentials with were achieved over 6 weeks with release enhanced by
increasing DPPE-PEG-2000 incorporation. The ad- erosion—disintegration and bone ingrowth into the
sorption of liposomes to S. aureus biofilms followed implant [78]. Other studies using the glycopeptide
the Langmuir isotherm and both surface coverage and antibiotic teicoplanin, effective against S. aureus, in-
the magnitude of the Gibbs energy of adsorption dicate that it too can be effective over several weeks
decreased with the extent of pegylation [64]. when incorporated into microspheres prepared from
PLGA (75:25) (Mr 136,000) polymer [79,80]. Other
3.3. Polymer-based antimicrobial delivery systems materials studied against implant-related osteomyelitis
include poly(3-hydroxybutyrate-co-3-hydroxyvale-
Many reviews have highlighted the use of biode- rate) (PHBV). The release behaviour of sulbactam:
gradable polyesters as effective drug carriers includ- cefoperazone from rods comprising 7%, 14% or 22%
ing nano- or microparticles, hydrogels, micelles and (mol) 3-hydroxyvalerate were representative of typi-
fibrous scaffolds [71–76]. Inevitably, there are advan- cal monolithic devices where a rapid early release
tages and disadvantages associated with each delivery phase is followed by a slower and prolonged phase.
system, however these experimental approaches have With PHBV 22 rods, this extended phase lasted for up
been investigated in a number of infections, including to 2 months, making it a promising controlled release
periodontitis and osteomyelitis, as well as intracellular vehicle since treatment of device-related infections is
1546 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

typically up to 6 weeks [81]. Release studies of anti- injectable collagen sponge, resulting in local drug
biotics adsorbed onto the surface of hydroxyapatite delivery combined with the tissue regeneration prop-
cylinders having a bimodal pore size distribution also erties of collagen [85]. Several other matrices have
have a prolonged duration of release, attributed to the been used, including the aluminosilcate material hal-
small pores, combined with favourable osteoconduc- loysite. Whilst chemically similar to kaolin, halloysite
tion properties into the large pores [82]. A number of has a tubular structure that can be loaded with drug.
matrices have been formed using poly(d,l-lactide-co- Moreover, surface charge neutralisation using cationic
glycolide) (PLGA), including discs [83] and electro- polymers can place an additional level of control on
spun nanofibrous scaffolds [84]. In an effort to drug release. In one study of tetracycline incorpora-
achieve the ideal drug release pattern of no lag time tion into halloysite for the treatment of periodontitis,
and zero-order release, lactide monomer or glycolide an initial burst of tetracycline release was followed by
monomer have been incorporated into PLGA discs a dramatic reduction in release. Coating with the
loaded with gentamicin. The idea here is that channels cationic polymer chitosan reduced the dburstT release
will form in the disc following dissolution of the from 45% to 30% and the total release over 9 days
monomer to aid the release of gentamicin. Discs con- from 88% to 78% [86]. A key concern was delivery of
taining 10% monomers showed nearly zero-order re- the halloysite to the gingival pocket and its subse-
lease kinetics for more than 1 month [83]. Evidence quent retention. The polyoxyethylene–polyoxypropy-
for the channel-forming properties of the monomers lene copolymer poloxamer 407 was used for its
came from the water uptake by the discs. After 7 days, thermoreversible properties of being liquid below 20
the amount of water absorbed by the control discs was 8C and forming a hydrogel at higher temperatures if
20%, compared with 60% in monomer-containing sufficiently concentrated, that is also biodegradable
discs. and relatively non-toxic. The ideal here is that the
Fibrous scaffolds are currently receiving attention delivery system should be a liquid at room tempera-
both as a means to prevent post-surgery adhesions and ture, so avoiding the need for refrigeration, and a
also to release drugs in a site-specific manner. Whilst device-retaining gel at the temperature within the
surgical implantation is required, they could be used gingival crevice. In this particular study, the transition
where surgery is already indicated and the drug re- temperature range was narrowed by the addition of
lease profile can be controlled by varying the scaf- polyethylene glycols, however this caused the storage
fold’s morphology, porosity and composition. In a modulus (solidity) of the gel to drop as well [86]. This
recent study, cefoxitin has been incorporated into was overcome by the addition of 1% octyl cyanoac-
PLGA scaffolds [84]. Here, PLGA controls the rate rylate (OCA), a powerful tissue adhesive that poly-
of degradation whilst high molecular weight poly(lac- merises at neutral pH. For storage, the pH of the
tide) (PLA) confers mechanical strength to the scaf- system was held at 4 by addition of glacial acetic
fold. An amphiphilic diblock co-polymer comprising acid, and upon application to the gingival crevice
poly(ethylene glycol)-b-poly(lactide) was added to the the natural buffer capacity of the tissue slowly brought
polymer solution to encapsulate the hydrophilic the pH of the formulation up to neutral, allowing the
cefoxitin sodium, which otherwise has poor solubility OCA to polymerise. Thermoreversible polymers have
in the PLGA solvent N,N-dimethyl formamide. The also been studied as delivery matrices in their own
drug/polymer solution was electrospun in a spinneret right [87–89]. For example, pluronic F-127 has been
at 23–27 kV. As the concentration of cefoxitin in- used to deliver vancomycin to treat methicillin-resis-
creased, the scaffolds changed from a bead and string tant S. aureus otitis media [90]. Based on the sol–gel
morphology, attributed to insufficient stretching of the phase transition measurements, a 25% w/v pluronic
polymer jet, to a fine fibrous structure of diameter solution was loaded with vancomycin and injected
260 F 90 nm. The spinning process did not affect the through a 26 gauge needle. It changed to a gel at 36
cefoxitin and following an initial burst, prolonged to 37 8C that consisted of large populations of the
release was measured for up to 1 week. micelles and aqueous channels from which the van-
Other applications of PLGA have included incor- comycin was released. Bioabsorption studies followed
poration of antibiotic-loaded microparticles into an over 50 days indicated that the gel gradually disap-
A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550 1547

peared leaving open spaces that initially showed some ultrasound used in some nebulisers. The resultant
inflammatory exudate, but by day 50 normal tissue aerosol properties will depend not only on the phy-
was observed without any inflammation, fibrosis, or sical process used to nebulise the drug, but also on
open spaces from the gel. the intrinsic device characteristics and its perfor-
mance with a particular drug [98].
3.3.2. Delivery on demand—an infection-responsive
system
An interesting approach to antibiotic delivery has 4. Discussion and future aspects
been the development of systems that are responsive
to microbial infection. The rationale here is to develop It is clear from this brief review of the current
systems that release antibiotics only during infection, literature that the full spectrum of modern drug deliv-
recognising that prophylactic or prolonged use of ery approaches is being applied to antimicrobial ther-
antibiotics can favour selection of resistant variants apy, but the prospects for a revolution in treatment
and also lead to renal and liver toxicity with some seem some way distant. The problem is not with the
agents such as the aminoglycosides. One such ap- drug delivery system; rather it is their payload. The
proach has been based on the observation that currently available antibiotics have a limited range of
wound fluid from S. aureus infection showed high cellular targets and were developed on the basis of
levels of thrombin-like activity. An insoluble poly- their activity against fast-growing planktonic cells in
mer–drug conjugate was prepared in which gentami- conventional laboratory culture. It seems likely that
cin was bound to poly(vinyl alcohol) through a drug delivery systems for the treatment of infection
thrombin-sensitive peptide linker [91]. The conjugate will only come of age when there are drugs that are
released gentamicin when it was incubated with worth carrying. We are beginning to understand the
thrombin and leucine aminopeptidase together, but physiology of biofilm bacteria at the genomic and
not with either component alone. Gentamicin was proteomic levels and we must look to these studies
also released upon incubation with S. aureus wound to yield new targets and to stimulate the design of
fluid and the conjugate reduced the bacterial number drugs that are worth carrying.
in an animal model of infection.

3.4. Aerosol delivery to the lung Acknowledgements

Aerosolization offers an attractive approach to Work in the author’s laboratory is supported by the
deliver antimicrobials directly into the respiratory BBSRC, the Lord Dowding fund for Humane Re-
tract for treatment and prophylaxis of pulmonary search and the Department of Health.
infections [92,93]. Aerosolized solutions of amino-
glycosides, particularly tobramycin, are used in
patients with cystic fibrosis, where high endobron- References
chial concentrations are achieved that may overcome
bacterial resistance as defined by standard suscepti- [1] C.A. Fux, P. Stoodley, L. Hall-Stoodley, J.W. Costerton, Bac-
bility testing protocols [94–96]. Other applications of terial biofilms: a diagnostic and therapeutic challenge, Expert
Rev. Anti-Infect. Ther. 1 (2003) 667 – 683.
aerosol technology include aerosolized antibiotics in
[2] P.S. Stewart, J.W. Costerton, Antibiotic resistance of bacteria
mechanically ventilated patients [97]. In this in- in biofilms, Lancet 358 (2001) 135 – 138.
stance, an efficient method was achieved that deli- [3] L. Hall-Stoodley, J.W. Costerton, P. Stoodley, Bacterial bio-
vers the aerosol beyond the endotracheal tube and films: from the natural environment to infectious diseases, Nat.
drug levels in pulmonary secretions were several Rev., Microbiol. 2 (2004) 95 – 108.
orders of magnitude higher than those following [4] P. Stoodley, K. Sauer, D.G. Davies, J.W. Costerton, Biofilm as
complex differentiated communities, Annu. Rev. Microbiol.
intravenous therapy. A number of questions have to 56 (2002) 187 – 209.
be addressed, notably whether the drug can with- [5] W. Zimmerli, A. Trampuz, P.E. Ochsner, Prosthetic-joint infec-
stand aerosol generation, such as the high-frequency tions, N. Engl. J. Med. 351 (2004) 1645 – 1654.
1548 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

[6] C. Brub-Buisson, New technologies and infection control [24] B. Giwercman, E.T. Jensen, N. Høiby, A. Kharazmi, J.W.
practices to prevent intravascular catheter-related infections, Costerton, Induction of h-lactamase production in Pseudomo-
Am. J. Respir. Crit. Care Med. 164 (2001) 1557 – 1558. nas aeruginosa biofilm, Antimicrob. Agents Chemother. 35
[7] P. Gilbert, P.J. Collier, M.R.W. Brown, Influence of growth (1991) 1008 – 1010.
rate on susceptibility to antimicrobial agents: biofilms, cell [25] T.R. De Kievit, M.D. Parkins, R.J. Gillis, R. Srikumar, H.
cycle, dormancy, and stringent response, Antimicrob. Agents Ceri, K. Poole, B.H. Iglewski, D.G. Storey, Multidrug efflux
Chemother. 34 (1990) 1865 – 1868. pumps: expression patterns and contribution to antibiotic re-
[8] J.W. Costerton, P.S. Stewart, E.P. Greenberg, Bacterial bio- sistance in Pseudomonas aeruginosa biofilms, Antimicrob.
films: a common cause of persistent infections, Science 284 Agents Chemother. 45 (2001) 1761 – 1770.
(1999) 1318 – 1322. [26] D.J. Hassett, J. Cuppoletti, B. Trapnell, S.V. Lymar, J.J. Rowe,
[9] K. Lewis, Riddle of biofilm resistance, Antimicrob. Agents S.S. Yoon, G.M. Hilliard, K. Parvatiyar, M.C. Kamani, D.J.
Chemother. 45 (2001) 999 – 1007. Wozniak, S.H. Hwang, T.R. McDermott, U.A. Ochsner, An-
[10] T.-F.C. Mah, G.A. O’Toole, Mechanisms of biofilm resistance aerobic metabolism and quorum sensing by Pseudomonas
to antimicrobial agents, Trends Microbiol. 9 (2001) 34 – 39. aeruginosa biofilms in chronically infected cystic fibrosis
[11] M.R.W. Brown, A.W. Smith, Antimicrobial agents and bio- airways: rethinking antibiotic treatment strategies and drug
films, in: M. Wilson, D. Devine (Eds.), Medical Implications targets, Adv. Drug Deliv. Rev. 54 (2002) 1425 – 1443.
of Biofilms, Cambridge University Press, Cambridge, 2003, [27] G. Borriello, E. Werner, F. Roe, A.M. Kim, G.D. Ehrlich, P.S.
pp. 36 – 55. Stewart, Oxygen limitation contributes to antibiotic tolerance
[12] P. Gilbert, T. Maira-Litrán, A.J. McBain, A.H. Rickard, F.W. of Pseudomonas aeruginosa biofilms, Antimicrob. Agents
Whyte, The physiology and collective recalcitrance of micro- Chemother. 48 (2004) 2659 – 2664.
bial biofilm communities, Adv. Microb. Physiol. 46 (2002) [28] E. Werner, F. Roe, A. Bugnicourt, M.J. Franklin, A. Heydorn,
202 – 256. S. Molin, B. Pitts, P.S. Stewart, Stratified growth in Pseudo-
[13] A. Brooun, S. Liu, K. Lewis, A dose–response study of monas aeruginosa biofilms, Appl. Environ. Microbiol. 70
antibiotic resistance in Pseudomonas aeruginosa biofilms, (2004) 6188 – 6196.
Antimicrob. Agents Chemother. 44 (2000) 640 – 646. [29] Y.H. An, R.B. Dickinson, R.J. Doyle, Mechanisms of bacterial
[14] I. Keren, N. Kaldalu, A. Spoering, Y. Wang, K. Lewis, Per- adhesion and pathogenesis of implant and tissue infections, in:
sister cells and tolerance to antimicrobials, FEMS Microbiol. Y.H. An, R.J. Friedman (Eds.), Handbook of Bacterial Adhe-
Lett. 230 (2004) 13 – 18. sion: Principles, Methods, and Applications, Humana Press
[15] I. Keren, D. Shah, A. Spoering, N. Kaldalu, K. Lewis, Inc., Totowa, NJ, 2000, pp. 1 – 27.
Specialized persister cells and the mechanism of multidrug [30] M. Gross, S.E. Cramton, F. Gotz, A. Peschel, Key role of
tolerance in Escherichia coli, J. Bacteriol. 186 (2004) teichoic acid net charge in Staphylococcus aureus colonization
8172 – 8180. of artificial surfaces, Infect. Immun. 69 (2001) 3423 – 3426.
[16] E. Drenkard, Antimicrobial resistance of Pseudomonas aeru- [31] J. Lin, S. Qui, K. Lewis, A.M. Klibanov, Bactericidal prop-
ginosa biofilms, Microbes Infect. 5 (2003) 1213 – 1219. erties of flat surfaces and nanoparticles derivatized with
[17] P. Gilbert, A.E. Hodgson, M.R.W. Brown, Influence of the alkylated polyethylenimines, Biotechnol. Prog. 18 (2002)
environment on the properties of microorganisms grown in 1082 – 1086.
association with surfaces, in: M.R.W. Brown, P. Gilbert (Eds.), [32] A. Bach, H. Eberhardt, A. Frick, H. Schmidt, B.W. Bottinger,
Microbiological Quality Assurance: A Guide Towards Rele- E. Martin, Efficacy of silver-coating central venous catheters
vance and Reproducibility of Inocula, CRC Press Inc., Boca in reducing bacterial colonization, Crit. Care Med. 27 (1999)
Raton, FL, 1995, pp. 61 – 82. 515 – 520.
[18] P.S. Stewart, Theoretical aspects of antibiotic diffusion into [33] G. Donelli, I. Francolini, A. Piozzi, R. Di Rosa, W. Marconi,
microbial biofilms, Antimicrob. Agents Chemother. 38 (1996) New polymer–antibiotic systems to inhibit bacterial biofilm
2125 – 2133. formation: a suitable approach to prevent central venous cath-
[19] K.D. Xu, G. McFeters, P.S. Stewart, Biofilm resistance to eter-associated infections, J. Chemother. 14 (2002) 501 – 507.
antimicrobial agents, Microbiology 146 (2000) 547 – 549. [34] J.M. Schierholz, L.J. Lucas, A. Rump, G. Pulverer, Efficacy
[20] W.W. Nichols, Biofilms, antibiotics and penetration, Rev. of silver-coated medical devices, J. Hosp. Infect. 40 (1998)
Med. Microbiol. 2 (1991) 177 – 181. 257 – 262.
[21] J.N. Anderl, M.J. Franklin, P.S. Stewart, Role of antibiotic [35] R.J. Sheretz, W.A. Carruth, A.A. Hampton, M.P. Byron, D.D.
penetration limitation in Klebsiella pneumoniae biofilm resis- Solomon, Efficacy of antibiotic-coated catheters in preventing
tance to ampicillin and ciprofloxacin, Antimicrob. Agents subcutaneous Staphylococcus aureus infection, J. Infect. Dis.
Chemother. 44 (2000) 1818 – 1824. 167 (1993) 98 – 106.
[22] I.W. Sutherland, The biofilm matrix—an immobilised but [36] S.E. Tebbs, T.S.J. Elliott, A novel antimicrobial central venous
dynamic environment, Trends Microbiol. 9 (2001) 222 – 227. catheter impregnated with benzalkonium chloride, J. Antimi-
[23] N. Bagge, O. Ciofu, L.T. Skovgaard, N. Høiby, Rapid devel- crob. Chemother. 31 (1993) 261 – 271.
opment in vitro and in vivo of resistance to ceftazidime in [37] G. Donelli, I. Francolini, Efficacy of antiadhesive, antibiotic
biofilm-growing Pseudomonas aeruginosa due to chromo- and antiseptic coatings in preventing catheter-related infec-
somal h-lactamase, APMIS 108 (2000) 589 – 600. tions: review, J. Chemother. 13 (2001) 595 – 606.
A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550 1549

[38] P.N. Danese, Antibiofilm approaches: prevention of catheter [52] M. Wainwright, K.B. Crossley, Photosensitising agents—cir-
colonization, Chem. Biol. 9 (2002) 873 – 880. cumventing resistance and breaking down biofilms: a review,
[39] S. Rachid, K. Ohlsen, W. Witte, J. Hacker, W. Ziebuhr, Effect Int. Biodeterior. Biodegrad. 53 (2004) 119 – 126.
of subinhibitory antibiotic concentrations on polysaccharide [53] N.S. Soukos, S.S. Socransky, S.E. Mulholland, S. Lee, A.G.
intercellular adhesin expression in biofilm-forming Staphylo- Doukas, Photomechanical drug delivery into bacterial bio-
coccus epidermidis, Antimicrob. Agents Chemother. 44 films, Pharm. Res. 17 (2000) 405 – 409.
(2000) 3357 – 3363. [54] J.F. O’Neill, C.K. Hope, M. Wilson, Oral bacteria in multi-
[40] A. Piozzi, I. Francolini, L. Occhiaperti, M. Venditti, W. Mar- species biofilms can be killed by red light in the presence of
coni, Antimicrobial activity of polyurethanes coated with anti- toluidine blue, Lasers Surg. Med. 31 (2002) 86 – 90.
biotics: a new approach to the realization of medical devices [55] F. Gad, T. Zahra, T. Hasan, M.R. Hamblin, Effects of growth
exempt from microbial colonization, Int. J. Pharm. 280 (2004) phase and extracellular slime on photodynamic inactivation of
173 – 183. gram-positive pathogenic bacteria, Antimicrob. Agents Che-
[41] G.L.Y. Woo, M.L. Yang, H.Q. Yin, F. Jaffer, M.W. Mittleman, mother. 48 (2004) 2173 – 2178.
J.P. Santerre, Biological characterization of a novel biodegrad- [56] M.N. Jones, Y.-H. Song, M. Kaszuba, M.D. Reboiras, The
able antimicrobial polymer synthesized with fluoroquinolones, interaction of phospholipid liposomes with bacteria and their
J. Biomed. Mater. Res. 59 (2002) 35 – 45. use in the delivery of bactericides, J. Drug Target. 5 (1997)
[42] V. DiTizio, G.W. Ferguson, M.W. Mittelman, A.E. Khoury, 25 – 34.
A.W. Bruce, F. DiCosmo, A liposomal hydrogel for the pre- [57] J.-H. Kim, M. Gias, M.N. Jones, The adsorption of cationic
vention of bacterial adhesion to catheters, Biomaterials 19 liposomes to Staphylococcus aureus biofilms, Colloids Surf.,
(1998) 1877 – 1884. A Physicochem. Eng. Asp. 149 (1999) 561 – 570.
[43] I. Francolini, P. Norris, A. Piozzi, G. Donelli, P. Stoodley, [58] S.D. Nightingale, S.L. Saletan, C.E. Swenson, A.J. Lawrence,
Usnic acid, a natural antimicrobial agent able to inhibit bac- D.A. Watson, F.G. Pilkiewicz, E.G. Silverman, S.X. Cal,
terial biofilm formation on polymer surfaces, Antimicrob. Liposome encapsulated gentamicin treatment of Mycobacteri-
Agents Chemother. 48 (2004) 4360 – 4365. um avium–Mycobacterium intracellulare complex bacteremia
[44] I. Raad, R. Hachem, A. Zermeno, L.C. Stephens, G.P. Bodey, in AIDS patients, Antimicrob. Agents Chemother. 37 (1993)
Silver iontophoretic catheter: a prototype of a long-term anti- 1869 – 1872.
infective vascular access device, J. Infect. Dis. 173 (1996) [59] L. Eduardo, M. Bermudez, M. Wu, L.S. Young, Intracellular
495 – 498. killing of Mycobacterium avium complex by rifapentine and
[45] J.W. Costerton, B. Ellis, K. Lam, F. Johnson, A.E. Khoury, liposome encapsulated amikacin, J. Infect. Dis. 156 (1987)
Mechanism of electrical enhancement of efficacy of antibiotics 510 – 513.
in killing biofilm bacteria, Antimicrob. Agents Chemother. 38 [60] H. Pinto-Alphandary, A. Andremont, P. Couvreur, Targeted
(1994) 2803 – 2809. delivery of antibiotics using liposomes and nanoparticles:
[46] P.S. Stewart, W. Wattanakaroon, L. Goodrum, S.M. Fortun, research and implications, Int. J. Antimicrob. Agents 13
B.R. McLeod, Electrolytic generation of oxygen partially (2000) 155 – 168.
explains electrical enhancement of tobramycin efficacy against [61] M.W. Fountain, S.J. Weiss, A.G. Fountain, A. Shen, R.P.
Pseudomonas aeruginosa biofilm, Antimicrob. Agents Che- Lenk, Treatment of Brucella canis and Brucella abortus in
mother. 43 (1999) 292 – 296. vitro and in vivo by stable plurilamellar vesicle-encapsulated
[47] S.A.W. Pickering, R. Bayston, T.F. Scanlin, Electromagnetic aminoglycosides, J. Infect. Dis. 152 (1985) 529 – 535.
augmentation of antibiotic efficacy in infection of orthopaedic [62] S. Majumdar, D. Flasher, D.S. Friend, P. Nassos, D. Yajko,
implants, J. Bone Jt. Surg., BR 85B (2003) 588 – 593. W.K. Hadley, N. Duzgunes, Efficacies of liposome-encapsu-
[48] A.M. Rediske, B.L. Roeder, J.L. Nelson, R.L. Robison, G.B. lated streptomycin and ciprofloxacin against Mycobacterium
Schaalje, R.A. Robison, Pulsed ultrasound enhances the avium–Mycobacterium intracellulare complex infections in
killing of Escherichia coli biofilms by aminoglycoside anti- human peripheral blood monocyte/macrophages, Antimicrob.
biotics in vivo, Antimicrob. Agents Chemother. 44 (2000) Agents Chemother. 36 (1992) 2808 – 2815.
771 – 772. [63] C. Catuogno, M.N. Jones, The antibacterial properties of solid
[49] J.C. Carmen, B.L. Roeder, J.L. Nelson, B.L. Beckstead, C.M. supported liposomes on Streptococcus oralis biofilms, Int. J.
Runyan, G.B. Schaalje, R.A. Robison, W.G. Pitt, Ultrasoni- Pharm. 257 (2003) 125 – 140.
cally enhanced vancomycin activity against Staphylococcus [64] K. Ahmed, P.W. Muiruri, G.H. Jones, M.J. Scott, M.N. Jones,
epidermidis biofilms in vivo, J. Biomater. Appl. 18 (2004) The effect of grafted poly(ethylene glycol) on the electropho-
237 – 245. retic properties of phospholipid liposomes and their adsorption
[50] W.G. Pitt, S.A. Ross, Ultrasound increases the rate of bacterial to bacterial biofilms, Colloids Surf., A Physicochem. Eng.
growth, Biotechnol. Prog. 19 (2003) 1038 – 1044. Asp. 194 (2001) 287 – 296.
[51] J.C. Carmen, J.L. Nelson, B.L. Beckstead, C.M. Runyan, R.A. [65] A.M. Robinson, M. Bannister, J.E. Creeth, M.N. Jones,
Robison, A.P. Schaap, W.G. Pitt, Ultrasonic-enhanced genta- The interaction of phospholipid liposomes with mixed
micin transport through colony biofilms of Pseudomonas bacterial biofilms and their use in the delivery of bacteri-
aeruginosa and Escherichia coli, J. Infect. Dis. 10 (2004) cide, Colloids Surf., A Physicochem. Eng. Asp. 186 (2001)
193 – 199. 43 – 53.
1550 A.W. Smith / Advanced Drug Delivery Reviews 57 (2005) 1539–1550

[66] K.J. Hill, M. Kaszuba, J.E. Creeth, M.N. Jones, Reactive with bimodal pore size distribution, J. Biomed. Mater. Res.
liposomes encapsulating a glucose oxidase–peroxidase system 70B (2004) 332 – 339.
with antibacterial activity, Biochim. Biophys. Acta, Bio- [83] J.Y. Yoo, J.M. Kim, G. Khang, M.S. Kim, S.H. Cho, H.B. Lee,
membr. 1326 (1997) 37 – 46. Y.S. Kim, Effect of lactide/glycolide monomers on release
[67] J.P. Wong, H. Yuang, K.L. Blasetti, G. Schnell, J. Conley, behaviours of gentamicin sulfate-loaded PLGA discs, Int. J.
L.N. Schofield, Liposome delivery of ciprofloxacin against Pharm. 276 (2004) 1 – 9.
Francisella tularensis infection, J. Control. Release 92 (2003) [84] K. Kim, Y.K. Luu, C. Chang, D. Fang, B.S. Hsiao, B. Chu, M.
265 – 273. Hadjiargyrou, Incorporation and controlled release of a hydro-
[68] M.H. Ellbogen, K.M. Olsen, M.J. Gentry-Nielsen, L.C. Pre- philic antibiotic using poly(lactide-co-glycolide)-based elec-
heim, Efficacy of liposome-encapsulated ciprofloxacin com- trospun nanofibrous scaffolds, J. Control. Release 98 (2004)
pared with ciprofloxacin and ceftriaxone in a rat model of 47 – 56.
pneumococcal pneumonia, J. Antimicrob. Chemother. 51 [85] M. Schlapp, W. Friess, Collagen/PLGA microparticle con-
(2003) 83 – 91. trolled composites for local delivery of gentamicin, J.
[69] C. Cordeiro, D.J. Wiseman, P. Lutwyche, M. Uh, J.C. Pharm. Sci. 92 (2003) 2145 – 2151.
Evans, B.B. Finlay, M.S. Webb, Antibacterial efficacy of [86] H.M. Kelly, P.B. Deasy, E. Ziaka, N. Claffey, Formulation and
gentamicin encapsulated in pH-sensitive liposomes against preliminary in vivo dog studies of a novel drug delivery
an in vivo Salmonella enterica serovar typhimurium intra- system for the treatment of periodontitis, Int. J. Pharm. 274
cellular infection model, Antimicrob. Agents Chemother. 44 (2004) 167 – 183.
(2000) 533 – 539. [87] S. Miyazaki, T. Tobiyama, M. Takada, D. Attwood, Percuta-
[70] I.A.J.M. Bakker-Woudenberg, R.M. Schiffelers, M.T. ten neous absorption of indomethacin from pluronic F 127 gels in
Kate, G. Storm, L. Guo, P. Working, Targeting of antibiotics rats, J. Pharm. Pharmacol. 47 (1995) 455 – 457.
in bacterial infection using pegylated long-circulating lipo- [88] E. Esposito, V. Carotta, A. Scabbia, L.D. Trombelli, P. Antona,
somes, J. Liposome Res. 10 (2000) 513 – 521. E. Menegatti, Comparative analysis of tetracycline-containing
[71] S. Freiberg, X.X. Zhu, Polymer microspheres for controlled dental gels: poloxamer- and monoglyceride-based formula-
drug release, Int. J. Pharm. 282 (2004) 1 – 18. tions, Int. J. Pharm. 142 (1996) 9 – 23.
[72] N.K. Varde, D.W. Pack, Microspheres for controlled drug [89] M.L. Veyries, G. Couarraze, S. Geiger, F. Agnely, L. Massias,
delivery, Expert Opin. Biol. Ther. 4 (2004) 35 – 51. B. Kunzli, Controlled release of vancomycin from poloxamer
[73] M.A. Moses, H. Brem, R. Langer, Advancing the field of drug 407 gels, Int. J. Pharm. 192 (1999) 183 – 193.
delivery: taking aim at cancer, Cancer Cell 4 (2003) 337 – 341. [90] S.H. Lee, J.E. Lee, W.Y. Baek, J.O. Lim, Regional delivery of
[74] V.R. Sinha, A. Trehan, Biodegradable microspheres for protein vancomycin using pluronic F-127 to inhibit methicillin resis-
delivery, J. Control. Release 90 (2003) 261 – 280. tance Staphylococcus aureus (MRSA) growth in chronic otitis
[75] Z. Zhao, J. Wang, H.Q. Mao, K.W. Leong, Polyphosphoesters media in vitro and in vivo, J. Control. Release 96 (2004) 1 – 7.
in drug and gene delivery, Adv. Drug Deliv. Rev. 55 (2003) [91] M. Tanihara, Y. Suzuki, Y. Nishimura, K. Suzuki, Y. Kaki-
483 – 499. maru, Y. Fukunisi, A novel microbial infection-responsive
[76] B.E. Rabinow, Nanosuspensions in drug delivery, Nat. Rev., drug release system, J. Pharm. Sci. 88 (1999) 510 – 514.
Drug Discov. 3 (2004) 785 – 796. [92] T.G. O’Riordan, Inhaled antimicrobial therapy: from cystic
[77] K. Kanellalopoulou, E.J. Giamarellos-Bourboulis, Carrier sys- fibrosis to the flu, Respir. Care 45 (2004) 275 – 276.
tems for the local delivery of antibiotics in bone infections, [93] M.E. Klepser, Role of nebulized antibiotics for the treatment
Drugs 59 (2000) 1223 – 1232. of respiratory infections, Curr. Opin. Infect. Dis. 17 (2004)
[78] C. Castro, E. Sánchez, A. Delgado, I. Soriano, P. Núñez, M. 109 – 112.
Baro, A. Perera, C. Évora, Ciprofloxacin implants for bone [94] A.L. Smith, Inhaled antibiotic therapy: what drug? what dose?
infection. In vitro–in vivo characterization, J. Control. Release what regimen? what formulation? J. Cyst. Fibros. 1 (2002)
93 (2003) 341 – 354. S189 – S193.
[79] I. Yenice, S. Çalis, B. Atilla, H.S. Kas, M. Ozalp, M. Ekizoglu, [95] S.P. Conway, K.G. Brownlee, M. Denton, D.G. Peckham,
H. Bilgili, A.A. Hincal, In vitro/in vivo evaluation of the Antibiotic treatment of multidrug-resistant organisms in cystic
efficiency of teicoplanin-loaded biodegradable microparticles fibrosis, Am. J. Respir. Medicine 2 (2003) 321 – 332.
formulated for implantation to infected bone defects, J. Micro- [96] B.W. Ramsey, M.S. Pepe, J.M. Quan, K.L. Otto, A.B. Mon-
encapsul 20 (2003) 705 – 717. tgomery, J. Williams-Warren, M. Vasiljev-K, D. Borowitz,
[80] I. Yenice, S. Çalis, H.S. Kas, M. Ozalp, M. Ekizoglu, A.A. C.M. Bowman, B.C. Marshall, S. Marshall, A.L. Smith, Inter-
Hincal, Biodegradable implantable teicoplanin beads for the mittent administration of inhaled tobramycin in patients with
treatment of bone infections, Int. J. Pharm. 242 (2002) cystic fibrosis, N. Engl. J. Med. 340 (1999) 23 – 30.
271 – 275. [97] G.C. Smaldone, Aerosolized antibiotics in mechanically ven-
[81] I. Gursel, F. Yagmurlu, F. Korkusuz, V. Hasirci, In vitro tilated patients, Respir. Care 49 (2004) 635 – 639.
antibiotic release from poly(3hydroxybutyrate-co-3-hydroxy- [98] P. Diot, P.F. Dequin, B. Rivoire, F. Gagnadoux, F. Faurisson,
valerate) rods, J. Microencapsul 19 (2002) 153 – 164. E. Diot, E. Boissinot, A. La Pape, L. Palmer, E. Lemarie,
[82] M. Hasegawa, A. Sudo, V.S. Komlev, S.M. Barinov, A. Aerosols and infectious diseases, J. Aerosol Med. 14 (2001)
Uchida, High release of antibiotic from a novel hydroxyapatite 55 – 64.

You might also like