Sei sulla pagina 1di 14

Advanced Drug Delivery Reviews 78 (2014) 14–27

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Therapeutic strategies to combat antibiotic resistance☆


Benjamin D. Brooks a, Amanda E. Brooks b,c,⁎
a
Wasatch Microfluidics, Salt Lake City, UT 84112, USA
b
Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
c
Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND58108, USA

a r t i c l e i n f o a b s t r a c t

Available online 28 October 2014 With multidrug resistant bacteria on the rise, new antibiotic approaches are required. Although a number of new
small molecule antibiotics are currently in the development pipeline with many more in preclinical develop-
Keywords: ment, the clinical options and practices for infection control must be expanded. Biologics and non-antibiotic
Antibiotics adjuvants offer this opportunity for expansion. Nevertheless, to avoid known mechanisms of resistance, intelligent
Antimicrobial polymers combination approaches for multiple simultaneous and complimentary therapies must be designed. Combination
Biofilm
approaches should extend beyond biologically active molecules to include smart controlled delivery strategies.
Quorum sensing
Combination therapy
Infection control must integrate antimicrobial stewardship, new antibiotic molecules, biologics, and delivery strat-
Nanoparticle delivery egies into effective combination therapies designed to 1) fight the infection, 2) avoid resistance, and 3) protect the
natural microbiome. This review explores these developing strategies in the context of circumventing current
mechanisms of resistance.
© 2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2. Antibiotic resistance: a global health threat . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.1. Resistance on the rise . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
2.2. Classes of antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.3. Mechanisms of resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.4. Antibiotic stewardship . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3. Addressing antibiotic resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.1. New drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.1.1. Quorum sensing inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3.1.2. Biologics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4. Dual drug delivery approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
4.1. Combination approaches that target different pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.1.1. Combinations with non-antibiotic adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.2. Combination approaches that act on the same pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.3. Combination approaches that act on the same target . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.4. Combination approaches to address polymicrobial infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.5. Resistance and synergistic drug combinations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.6. Pitfalls of combination drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5. Antibiotic delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.1. Systemic versus local antibiotic delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.1.1. Antimicrobial polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5.2. A unique local delivery system—nanoparticle/liposome delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5.2.1. Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5.2.2. Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Emergence of multidrug resistance bacteria: Important role of macromolecules as a new drug targeting mi-
crobial membranes”.
⁎ Corresponding author at: 1401 Albrecht Blvd, Fargo, ND, 58108, USA. Tel.: +1 701 231 7906.
E-mail address: Amanda.e.brooks@ndsu.edu (A.E. Brooks).

http://dx.doi.org/10.1016/j.addr.2014.10.027
0169-409X/© 2014 Elsevier B.V. All rights reserved.
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 15

5.3. Impact of local drug delivery pressure on the development of resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21


6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

1. Introduction strategic fight against resistance, and protecting the delicate symbiotic
host-microbial balance (Fig. 2).
The growing tide of bacterial resistance imposes a tremendous
economic and social burden on the healthcare system worldwide and 2. Antibiotic resistance: a global health threat
unfortunately, the development of new antibiotics is vastly outpaced
by current therapies that are losing their efficacy (Fig. 1). This review 2.1. Resistance on the rise
covers both current antibiotic therapies in the context of rising multi-
drug resistance, but also emerging therapeutic alternatives seeking to The emergence of multidrug resistance pathogens is an increasingly
address antibiotic resistance and prolong the effective life of an antibiot- significant global economic and healthcare crisis [1,2]. Listed by the
ic. Combination therapies, alternate delivery systems, and alternative World Health Organization as one of the top 3 threats to global public
antimicrobial molecules in the pipeline will be discussed. Ultimately, health [3], more than 2 million Americans suffer from an antibiotic
fighting multidrug resistance necessitates a paradigm shift in therapy, resistant infection at a direct cost of over $20 billion [4] with over
including mounting a local antimicrobial attack when possible, a 23,000 dying annually [2]. Analogous worldwide statistics are staggering,

Fig. 1. A) Antibiotic resistance is increasing while the number of antibiotics is decreasing (modified from [40]). B) At most there is roughly 60 years between antibiotic discovery to the first
incidence of resistance to that same antibiotic (modified from [11]).
16 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

Fig. 2. Conceptual diagram outlining the three criteria (on the left) necessary for any strategy designed to circumvent the four recognized modes of antibiotic resistance (on the right).

prompting intense multidisciplinary efforts by scientific and clinical life cycle [28–30]. Within these broad classifications, bacteria can mod-
communities to develop innovative products and tools to address the ify the antibiotic by: 1) inactivation or enzymatic modification [31–33],
threat (Fig. 1). The CDC has recently classified emergent resistant species 2) alteration of the antibiotic target [34], or 3) changes in cell perme-
as urgent, serious, or concerning with a generalized picture of the cycle ability and efflux [35–37]. These mechanisms are pictorially described
of resistance shown in Fig. 3 [2]. Antibiotic surveillance programs, such in Fig. 3. However, for an in depth description of antibiotic resistance
as the SENTRY program, have monitored worldwide Methicillin Resis- mechanisms, the reader is referred to several excellent articles and
tant Staphylococcus aureus (MRSA) statistics (MRSA in hospitals be- reviews as cited above.
tween 1997 and 1999 was 22.4% in Australia, 66.8% in Japan, 34.9% in
Latin America, 40.4% in South America, 32.4% in the USA and 26.3% in 2.4. Antibiotic stewardship
Europe [5–7]) and noted with horror the necessity of elevated antibiotic
concentrations to effectively kill many common human pathogens Considering the rising inventory of multidrug resistant microbes, an-
(e.g., Pseudomonas aeruginosa, Klebsiella pneumoniae and Acinetobacter tibiotic stewardship, as defined by a number of preventative measures,
baumannii, Enterobacter and Escherichia coli) (i.e., MIC creep) [8,9], is not just a formal and practical strategy, but must now be implement-
prompting concerns of reverting to a pre-antibiotic era [10]. ed out of necessity [38]. Recently, the Transatlantic Taskforce on Antimi-
crobial Resistance (TATFAR) outlined the most pressing needs to fight
2.2. Classes of antibiotics antimicrobial resistance. These include (1) appropriate therapeutic use
in human and veterinary medicine, (2) prevention of drug-resistant in-
Resistance has developed to virtually every class of antibiotics in fections, and (3) strategies for improving the pipeline of new antimicro-
current use. Development of bacterial resistance to a given antibiotic bial drugs [39]. The Infectious Diseases Society of America has mirrored
is anticipated to evolve within an average of 50 years after initial use these recommendations along with providing additional surveillance
[11]. Figs. 4 and 5 graphically depict current drug mechanisms of action. measures [40]. While each of these recommendations is commonly ac-
Resistance to certain antibiotics (e.g. tetracyclines, etc.), often develop cepted as necessary for infection control, several barriers to antibiotic
in at least one bacterial species within a year of drug FDA approval stewardship programs remain, including lack of clinician participation
[12] with clinically significant levels of resistance appearing within [41], an absence of formal diagnostic standards, and non-uniform
months to years [13,14]. The prevalence of bacterial multidrug resis- reporting guidelines [38]. Nevertheless, appropriate antibiotic use is crit-
tance now vastly outpaces the advent of new antibiotic classes and ical as are prescreening microbiological tests with appropriate antibiotic
alternatives (Fig. 1) [8]. Since a 2009 report on antibiotic resistance follow-up [7] and stringent hand-washing guidelines and enforcement.
from the Infectious Disease Society of America, only 2 new antibiotics The use of combinations, particularly those with non-antibiotic adju-
(telavancin in 2009 and ceftaroline fosamil in 2010) have been intro- vants, offers a more effective long-term solution to address multidrug
duced to the market (Fig. 1). resistant variants via de novo drug delivery. Regardless, each strategy re-
quires a major change in antibiotic-prescribing patterns [42]. Ultimately,
2.3. Mechanisms of resistance antibiotic resistance is not just a medical crisis, but must encompass a
worldwide societal change at all levels to combat the evolution of antibi-
Drug discovery and development are locked in a co-evolutionary otic resistance.
battle with natural bacterial compounds [15]. Extensive studies on anti-
biotic resistance mechanisms have not only provided valuable insight 3. Addressing antibiotic resistance
into the underlying evolutionary processes that govern the develop-
ment and spread of antibiotic resistance [16,17], but have also allowed 3.1. New drugs
antibiotic resistance to be broadly classified as occurring via either:
1) innate resistance [18], 2) acquired resistance (e.g., by horizontal gene While new approaches to avoid drug resistance are researched and
transfer [19–21] or elevated mutation rates [22–24]), or 3) adaptive re- developed continually, antibiotic discovery is not keeping pace with
sistance (environmentally induced genetic changes [25]) [26,27], which rates of drug resistance (Fig. 1). Combating the complex issue of antibiotic
includes the conversion from a planktonic life cycle to a sessile biofilm resistance must go beyond the mechanistic development of new
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 17

Fig. 3. Graphic depicting the behavioral and molecular mechanisms of resistance. Modified from the Center for Disease Control [2].

pharmaceuticals and incorporate a multidisciplinary culture of change. redundancy in bacterial communication and kill persisters [45]. In an ef-
Thus, the third aim arising out of TATFAR seeks to provide (1) incentives fort to identify novel inhibitors, natural and small molecule libraries have
to stimulate the development of new antibacterial drugs in human been explored, identifying several promising candidates (e.g., HT61, C30
medicine, (2) research to support the development of new antibacte- furanone [46–49], etc.). Although some molecules have been tested in
rials, and (3) publication of funding opportunities to EU and US research preclinical animal models [50–52], clinical application is far from certain.
communities [39,40]. Despite these recommendations, the develop- The quorum inhibitor, FS3, was evaluated in a rat model and showed
ment of new antimicrobials or modifications of the current arsenal good efficacy and synergy with daptomycin [50]. An agent that does
may not be able to effectively address the trends in resistance. Instead, not impose life or death selective pressure but instead disrupts bacterial
antibiotic resistance may better be addressed with “smart” delivery sys- communication and virulence may circumvent the majority of the
tems and innovative combinations of biologically inspired molecules known resistance mechanisms [53]. Unfortunately, despite this promise,
(e.g., quorum sensing inhibitors, biosurfactants, bacteriophage, enzymes, increasing evidence suggests that quorum quenching may provoke a re-
etc.). Table 2 summarizes some of the more traditional drugs currently in sistant phenotype [54,55], although the mechanism is not clear.
the development pipeline while the sections below explore some of the
more nontraditional drugs being researched. 3.1.2. Biologics

3.1.1. Quorum sensing inhibitors 3.1.2.1. Antimicrobial peptides. Four main classes of antimicrobial pep-
Considering its role as a master switch, interrupting quorum sensing tides (AMPs) exist in nature including (1) anionic peptides, (2) cationic
with quorum quenchers (QQ) or quorum sensing inhibitors (QSI) is a fea- peptides with high molar percents of proline, arginine, phenylalanine,
sible alternative to antibiotics [9,43]. Three general approaches are ap- glycine, or tryptophan, (3) anionic and cationic peptides that contain
plied to quench or suppress microbial quorum sensing: 1) disruption of cysteine and form disulfide bonds, and (4) linear cationic alpha-helical
autoinducer synthesis, 2) inhibition of ligand/receptor interactions, and peptides [56,57]. The mechanism of AMPs includes cell membrane dam-
3) destruction of the autoinducer via enzymatic cleavage and degrada- age, inhibition of cell wall synthesis formation of pores, and disruption of
tion [9,44]. A combination of approaches may be necessary to overcome the membrane bilayer [58], and inhibition of nucleic acid and protein
18 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

synthesis [56]. These antimicrobial peptides, 12 to 50 amino acids in


length, are potent (at micromolar concentrations), broad-spectrum anti-
biotics and function as an integral component of the innate immune sys-
tem. Unfortunately, these drugs are expensive, often antigenic [59], and
have limited stability. Some of these disadvantages can be circumvented
by covalently grafting the AMP to a biomaterial surface [60,61]. Thus, due
to their role in the innate immune system and minimal risk of antibiotic
resistance [62,63], this class of natural antibiotics provides an excellent re-
source for future drug development [64]. Current AMPs approved by the
FDA include bacitracin, colistin, and polymyxin B; however, additional
synthetic molecules are being designed to mimic the architecture and
charge of natural AMPs [65–69].

3.1.2.2. Bacteriophage. Bacteriophage, a virus specifically designed to


infect bacteria through recognition of a cell surface receptor, pre-
sents one of the most attractive alternatives to combating antimicro-
bial resistance. Unlike more traditional antibiotic strategies,
bacteriophage titer and its subsequent efficacy change in direct pro-
portion to bacterial populations, and increase to combat the patho-
gen re-emergence [70]. Bacteriophages have been reported
effective in a variety of gram-negative (e.g., E. coli, P. aeruginosa,
A. baumannii, K. pneumoniae, Vibrio vulnificus, Salmonella spp) and
gram-positive bacteria (e.g., Enterococcus faecium and S. aureus)
[71–75]); however, bacteriophage use is not recommended without
reservation. Although not a complete litany of their disadvantages, of
primary concern is the integration of phage DNA into the bacterial
genome, the failure of bacteriophage therapy due to restrictive
specificity, and the development of bacterial resistance based on al-
teration of the bacterial cell surface receptor [70,76]. A cocktail
containing several different phages, especially when used in
conjunction with a traditional antibiotic [77] may circumvent these Fig. 5. Three of the main antibiotic resistance strategies used by bacteria (modified from
disadvantages. Alternatively, using bacteriophage components [11]).
(e.g., virolysin, antimicrobial peptides, etc.) may circumvent many,
if not most, of these pitfalls [70], serving as a significant source of po-
tent new antimicrobials. Furthermore, using a modified phage coat 4. Dual drug delivery approaches
to display an antigenic peptide, in conjunction with its natural bacte-
rial targeting specificity, may potentially provoke the host immune In the face of rising concerns over the development of antibiotic
response at the site of infection [78,79]. resistance, single drug clinical treatment strategies may not be adequate

Fig. 4. Summary of the different mechanisms of antibiotic action and examples of the different drugs that act via each mechanism. Modification of Fig. 20-14 from Brock Biology of Micro-
organisms [193].
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 19

to combat the problem. Several strategies exist to address resistance 4.1.1.2. Inhibitor adjuvants. Using a combination therapy capable of
beyond simply filling of the new drug pipeline. Based on the scientific delivering a bactericidal antibiotic while concurrently inhibiting the
literature and clinically proven recommendations and treatments [80, mechanism of resistance is a very effective strategy to overcome certain
81], combination approaches are more effective to combat multidrug resistance mechanisms [97,98]. This approach is particularly useful in
resistance [82,83]. Over 7 decades of experience has proven that drug cases of potential enzymatic disruption of the antibiotic, such as in the
combinations are key tools in the clinical arsenal against viruses [84] case of Augmentin, which pairs a β-lactam antibiotic (amoxicillin)
(e.g., Stribild, a 4-drug cocktail for HIV), cancers [85] and microbes with a β-lactamase inhibitor (e.g., clavulanic acid). Alternatively, com-
(e.g., Menhibrix, a combination vaccine for meningococcal disease and pounds that inhibit efflux pumps have also been used in several antibi-
type B influenza) [86,87]. In fact, among these classes, bacteria seem otic combinations to suppress a resistant phenotype. Reserpine, an MDR
to be particularly hard hit by combinatorial approaches. Additionally, a pump inhibitor, used in conjunction with ciprofloxacin has been ob-
combinatorial approach can effectively prolong the effective life- served to suppress resistance in both S. aureus and Streptococcus
time of a drug [12] in a landscape of antibiotic resistance as well as pneumoniae [99]; whereas, celecoxib, a nonsteroidal anti-inflammatory
restore efficacy to a drug which bacteria have developed resistance drug (NSAID) that inhibits the MDR1 efflux pump, enhances the sensi-
(e.g., antibiotics with nanoparticles) [88]. Regardless of the specific tivity of S. aureus to a variety of antibiotics, including ampicillin,
cocktail, strategies can be divided into three categories based on the kannamycin, chloramphenicol, and ciprofloxacin [100]. Most efflux
drug target: 1) antibiotic targets in different pathways (e.g., isoniazid, pump inhibitors increase sensitivity by allowing the intracellular accu-
rifampicin, ethambutol, and pyrazinamide for the treatment of M. mulation of drugs, providing a complimentary mechanism to prolong
tuberculosis infections), 2) different targets in the same pathway the life of a drug and stifle multidrug resistance [101]. Using an inhibitor
(e.g., sulfamethoxazole and trimethoprin), and 3) the same target as a non-antibiotic adjuvant has two important distinctions: 1) unlike an
with multiple mechanisms (e.g., streptogramins and virginamycin antiseptic adjuvant, which itself has antimicrobial potential, inhibitors
[81,89]) [90] (Fig. 4). circumvent or repress a mechanism of resistance but do not themselves
have antimicrobial potential [81] and 2) the complementary mechanism
4.1. Combination approaches that target different pathways of an inhibitor adjuvant directed at antibiotic resistance not only
enhances the efficacy of its antimicrobial delivery partner but does
Dual drug delivery approaches that combine antibiotics targeting not necessarily provoke a new mechanism of resistance [90]. These
different pathways are the most studied and successful approaches for two distinctions indicate a great deal of promise in this approach.
creating multidrug cocktails to combat antibiotic resistance [90]. To fur-
ther enhance efficacy while minimizing necessary drug concentrations, 4.1.1.3. Natural and biological adjuvants. One of the more novel
coupling a multidrug combinatorial approach with local drug delivery approaches to combination therapy has been the use of natural and
may prove the most efficacious dosing regimen to prolong and restore biological adjuvants. The pairing of an antibiotic with a bacteriophage
the efficacy of the current antibiotic arsenal [91]. Regardless of the ad- adjuvant, or a bacteriophage component adjuvant may prove more
ministration route, many effective combinations that target different effective than either component delivered individually and is now avail-
pathways are not strictly confined to antibiotics but extend to combina- able in Georgia (PhagoBioDerm uses a combination of a lytic phage
tions with non-antibiotic adjuvants. cocktail and ciprofloxacin in a biodegradable polymer matrix [102])
[70,103]. Alternative approaches to using biological compounds as syn-
4.1.1. Combinations with non-antibiotic adjuvants ergistic adjuvants have led to the use of the plant-derived thymol, in com-
Often the combination of an antibiotic with a non-antibiotic bination with vancomycin and EDTA [90], and the use of biosurfactants
adjuvant—defined as any compound used to assist in the amelioration, such as sophorolipid, which has no inherent antimicrobial activity [104].
prevention, or cure of disease—is the most effective way to combat multi- The use of plant-derived compounds and biosurfactants is considered
drug resistance due to the diversity of mechanisms. Such combinations very safe. Biosurfactants have been approved by the FDA for use in phar-
(e.g., minocycline, rifampin, and chlorhexidine [92]), are designed to com- maceuticals and food [104]. Using biological adjuvants takes advantage of
bat the redundancy of biological systems. Three common adjuvants that natural processes to enhance antimicrobial efficacy and provides a fea-
have had some clinical success are antiseptics inhibitors, and other bio- sible alternative to combat antimicrobial resistance. Antibodies (IgG
logical (e.g., bacteriophage) or natural (e.g., plant-derived) moieties. classes) have also been proposed for use as an adjuvant to promote a
Additionally, several known classes of compounds, e.g., antihistamines, host immune response and circumvent antibiotic resistance develop-
tranquilizers, anti-hypertensives, antispasmodics, and anti-inflammatory ment since the selective pressure is not placed directly on the bacteria
drugs, are now being explored as antibiotic adjuvants [93]. [105].

4.1.1.1. Antiseptic adjuvants. One of the most clinically familiar classes of 4.2. Combination approaches that act on the same pathway
non-antibiotic adjuvants is the antiseptics or biocides [94]. Chlorhexi-
dine, a bisbiguanide, is the most commonly used antiseptic. Also de- A combination strategy can also target different molecules in the
signed to kill or inhibit the proliferation of microorganisms, antiseptics same pathway. Although this presents a less diversified strategy than
are reported to be more indiscriminate in their mode of actions, having targeting different pathways, if the proper pathway is chosen, this can
multiple targets [95]. Efficacy is thought to derive from an ability to per- prove a very effective strategy. Two parameters confine the choice of
meate and disrupt the membrane as well as inactivate ATPase [95]. This pathway. First, the targeted pathway must represent an absolute surviv-
favors combination approaches that exploit the complimentary phar- al requirement, such as a requirement for folate to synthesize dTMP, a
macodynamics of antibiotics. One of the most clinically successful com- precursor for DNA synthesis. Second, the pathway cannot be redundant,
bination approaches is an antibiotic/antiseptic combination catheter as redundancy predisposes the strategy to resistance [89]. Targeting two
coating, which has proven effective against a variety of pathogens [83]. steps in the same pathway presents a more risky strategy in the face of
Alternatively, local delivery of an antibiotic/antiseptic combination via rising antibiotic resistance, yet it is still more effective than monothera-
a chitosan sponge has also been explored as an effective treatment pies in many cases [87,106,107].
[91]. Despite these successful strategies, concerns about resistance aris-
ing locally due to selective pressures are not completely mitigated and 4.3. Combination approaches that act on the same target
has been observed in vitro with combinations of chlorhexidine or silver
sulfadiazine (antiseptics) and minocycline or rifampicin (antibiotics) Although targeting two different molecules in the same bacterial
[96]. pathway is a less diversified approach than targeting two completely
20 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

different pathways, it is more diversified than using a combination ther- requisite dosing and concentration [88]; moreover, individual drug
apy where both mono therapies inhibit the same molecule. The classic mechanisms of action must be weighed so as not to introduce antagonis-
example revolves around the host of antibiotics that target the bacterial tic interactions, such as that which has been observed between certain
ribosome. A case study of synercid, a semisynthetic two-drug combina- DNA synthesis inhibitors and protein synthesis inhibitors [120]. Ulti-
tion in which the components bind to adjacent regions in the 50S ribo- mately, a better mechanistic understanding of antibiotic combinations
somal subunits proves the utility of this approach, as they are 10–100 in addition to more creative combinations, such as pairing an antibiotic
fold more effective than either drug individually [89]. Using a combina- with non-antibiotic adjuvant or a quorum sensing inhibitor, is necessary.
tion of two drugs that target the ribosome may prove effective because
of the critical and conserved nature of the bacterial ribosome. Thus, once 5. Antibiotic delivery
again it is clear how proper target selection is critical in this strategy.
5.1. Systemic versus local antibiotic delivery
4.4. Combination approaches to address polymicrobial infections
Traditionally, systemic antibiotic administration has been the foun-
In addition to combinations effective against a single species of path- dation of clinical therapies to address the ever-present infectious on-
ogen, combination therapies are also critical for the treatment of poly- slaught. Unfortunately, poor penetration to ischemic or post-operative
microbial infections [108]. Between 4 and 27%, with another study tissue, inappropriate prescribing patterns, systemic toxicity, and poor
suggesting numbers as high as 37% [109], of peri-prosthetic joint infec- patient compliance, have predominated the conversation and limited
tions result from the presence of more than one pathogen [110,111]; the usefulness of certain antibiotics. Furthermore, systemic administra-
thus, the ability to target multiple pathogens concurrently and quickly tion is often not effective, as it does not provide local tissue concentra-
is emerging as a valuable tool in fighting infections. Importantly, combi- tions sufficient to kill bacteria prior to incurring serious side effects, such
nation therapies need not be limited to only two component drug cock- as renal and liver damage. Sub-therapeutic or sub-inhibitory antibiotic
tails. Triple antibiotic combinations such as a combination of β-lactam, a concentrations are known to inadvertently exacerbate infectious compli-
glycopeptide, and an aminoglycoside have proven much more effective cation and promote antibiotic resistance [83,121]. Local delivery of cur-
against MRSA strains when compared to two antibiotic combinations rent antibiotics and other antimicrobial biologics (e.g., antimicrobial
[112]. Furthermore, combinations composed of antibiotics with varying peptides (AMPs), anti-quorum sensors, bacteriophage, etc.) may preserve
mechanisms of action (i.e., protein synthesis inhibitors (macrolides, and extend their efficacy in the evolutionary race between antimicrobial
aminoglycosides, tetracyclines, lincosamides, and chloramphenicol), development and bacterial resistance. In fact systemic toxicity, and to a
DNA synthesis inhibitors (fluoroquinolones and quinolones), folic acid lesser extent, antibiotic resistance is rarely seen for local applications of
synthesis inhibitors (sulfonamides and diaminopyrimidines), cell wall the same drugs, achieving locally higher concentrations and overcoming
synthesis inhibitors, polypeptide antibiotics, preservatives, and analge- the reducing effects of lowered bacterial metabolism [91,122–124].
sics) may prove particularly effective for polymicrobial infections due Thus, device integrated, local delivery strategies to mitigate the unaccept-
to their varied modes of action [112]. able consequences of systemic antibiotic delivery (e.g., development of
multi-drug resistant bacteria, systemic toxicity, and rising healthcare
4.5. Resistance and synergistic drug combinations costs, etc.) are urgently needed to keep pace with the rising demand for
medical devices.
Although it initially appears that using a drug combination would The concept of locally and sustainably delivering an anti-infective
serve to circumvent many resistance mechanisms, a drug cocktail may agent is not new. Vancomycin, tobramycin, amoxicillin, gentamicin,
in fact promote the evolution of drug resistance, despite multiple selec- cefamandol, caphalothin, and carbenicillin have all been incorporated
tive pressures [87,113–115]. This may be related to the fact that resis- into commercially available local release systems [125–127]. As early
tance to certain antibiotics in vitro, particularly aminoglycosides, as the 1940s, efforts were made in dentistry and orthopedics to endow
enhances the sensitivity to other antimicrobial agents, as indicated by implantable materials (e.g., cements and resins) with biological activi-
an increase in the minimum inhibitory concentration [116]. Drug cock- ty [128,129]; however, it quickly became apparent that the efficacy of
tails can also increase the danger of super-infection due to the co- these systems in locally combating a pathogenic infection was largely
evolution of multi-drug resistant variants and sensitization to other an- dependent on the pharmacokinetics of the system [83]. Ideally, local an-
tibiotics in the same class as the treatment [108]. In order to overcome tibiotic release kinetics should exhibit an initial high dose, well above
this threat, designing effective drug combinations is an important the minimal inhibitory concentration (MIC) of the offending bacteria
approach to reducing the emergence of antibiotic resistant bacteria but below the toxic threshold of the surrounding host cells, followed
while increasing the efficacy of the treatment and prolonging the utility by a sustained release of antibiotic sufficient to address the later emer-
of individual agents. gence of any latent bacteria. The duration and level of sustained release
are dependent on the host tissue matrix, the type and mechanism of the
4.6. Pitfalls of combination drug delivery antibiotic, and the clinically established critical postoperative period
[127]. Unfortunately, many local antibiotic delivery technologies have
All contemplated drug combinations must carefully consider the suffered from early burst release with subsequent sub-therapeutic activ-
impact of drug–drug interactions, compound ratios, and dosing regimens ity, inadvertently evoking bacterial antibiotic resistance. This quandary
on the adsorption, metabolism and excretion of each drug individually is epitomized by aminoglycoside delivery from (poly)methyl methacry-
[98,117,118]. The relatively simplistic approach of concurrently adminis- late (PMMA) for the treatment of osteomyelitis. Once the drug has
tering two or more synergistic (i.e., the efficacy of the drugs together is leached through small pores to a sustained sub-inhibitory level, PMMA
greater than the effect of either drug individually) antibiotics may not acts as a foreign body and nidus of infection, promoting the evolution
only alter the pharmacokinetics of drug delivery based on drug–drug of multidrug resistant bacteria [130,131]. Consequently, degradable
interactions but may also prove toxic to the bacteria as well as to the polymers have been sought in order to achieve sustainable high local
host cells and valuable symbiotic microbes in the host microbiome. concentrations while still avoiding the pitfalls of systemic administra-
Sequential synergistic drug dosing may avoid some of these common pit- tion [30,132–134]. Many different natural and synthetic biodegradable
falls as can hybrids (i.e., a single antibiotic with two distinct functions), polymers (e.g., collagen [135,136], chitosan [123,124,137,138], silk
such as the lantibiotic, nisin [14,119]. Notably, certain drug combinations [139], hydroxyapatite [140], fibrin clots [141,142], polyurethane [143],
can actually mitigate the toxicity of individual drugs in the cocktail polycaprolactone [144,145], PLGA [146] etc.) have been explored for
(e.g., metallic nanoparticles and antibiotics), often due to decreasing local antibiotic release.
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 21

5.1.1. Antimicrobial polymers are unique in their varied administration routes, drug elution kinetics
Unlike many classic antimicrobial compounds that are not capable of and intracellular delivery capability.
adequately addressing surface bound bacteria, antimicrobial polymers
were designed for just such a scenario [147–150]. These biologically ac- 5.2.1. Nanoparticles
tive polymers fill a critical niche in the development of new biocompat- Nanoparticle drug delivery systems can claim most of the same
ible, active medical devices. Such antimicrobial devices may effectively advantages of liposome delivery systems (e.g., cellular penetration,
ease the burden on systemic antibiotics, essentially promoting antibiot- surface modification for targeting to increase therapeutic efficacy,
ic stewardship, prolonging the life of conventional antibiotic therapies, etc.); however, their polymer composition provides additional stability
and curbing the evolution of antibiotic resistant microbes. Historically, not seen in liposome drug preparations to increase their resident circu-
cationic poly-amino acids and polyelectrolytes have been the molecules lation time and protect their drug payload from degradation [172].
of choice for development of antibiotic polymers; however, recent Nanotechnology-based drug delivery systems are an emerging approach
efforts have expanded this narrow focus to include polymers with anti- to improve: 1) the therapeutic index thereby decreasing the dose and
microbial chemical modifications, polymers containing antimicrobial frequency of administration, 2) intracellular delivery to mitigate the de-
organic compounds and those that incorporate antibacterial inorganic velopment of multidrug resistant bacteria [173], and 3) targeted organ
compounds. Integrating an antibiotic agent into a polymer matrix accumulation to limit systemic side effects [174,175] (Fig. 6). Metal ion
serves to minimize antibiotic toxicity, increase antibiotic stability, and nanoparticles, especially silver compounds, have an established history
improve antibiotic half-life and efficacy [150–152]. of use in medicine and pharmaceutics and have been explored as adju-
vants and carriers for antibiotic delivery [176,177]. Recent efforts have
5.1.1.1. Polymers with intrinsic antimicrobial activity. Certain polymers attempted to couple gold nanoparticles with a variety of antibiotics
can be exploited based on their inherent antibiotic activity, which (e.g., ciprofloxacin and other fluoroquinolones [178], vancomycin [179],
often includes chemical and other structural elements. Among those gentamicin [180], etc.) with inconsistent results. Gu et al. found enhanced
most studied are polymers mimicking natural peptides (e.g., peptoids activity against vancomycin resistant gram-negative bacteria when
(oligo-N-substitutes glycines [69,153]), etc.), cyclic peptides [154], hal- decorating the surface of gold nanoparticles with vanomycin [179]; how-
ogen polymers (fluorine or chlorine-containing polymers, polymeric ever, Burygin et al. showed no enhanced activity of gentamicin gold
N-halamines [155], phenylene ethynylenes [156], and organometallic nanoparticles over that of the unbound drug [180]. These inconsistencies
polymers. This group can also include polymers with cationic groups may be attributable to the characteristics of the antibiotics used as well as
(e.g., biguanide, quaternary ammonium salts, quarternary pyridinium the experimental conditions and the size of the nanoparticles, all param-
salts, and phosphonium salts) [157,158] or cationic conjugates eters known to affect efficacy. Although not a factor in this particular case,
(e.g., polyelectrolytes, polysiloxanes, polyoxazolines, polyionenes, etc.). it is important to note that nanoparticle drug delivery is generally more
active against gram-negative species [90]. Nevertheless, despite the po-
tential for nanoparticle drug delivery systems and clinical success, partic-
5.1.1.2. Polymers with antimicrobial chemical modifications. Since 1965,
ularly with metallic nanoparticles, there are relatively few marketed
polymers have been covalently modified to endow them with bacteria
nanoparticle based drug delivery systems [181]. This may be a conse-
resistant and bactericidal properties [159]. Traditionally, these chemical
quence of the complicated characterization and analyses necessary for
modifications have fallen into 3 primary categories: 1) covalently at-
nanoparticle technologies and their limited targeting efficiency and
tached small molecules on a polymer backbone [160,161], 2) attached
short half-life (for a review of these issues please see [182]).
antimicrobial peptides to a biologically inactive polymer [162,163], or
3) grafted antimicrobial polymer on a biologically inactive polymer
5.2.2. Liposomes
[164,165].
Intravenous injection is the most popular route for liposome drug
delivery and provides some advantage over traditional systemic deliv-
5.1.1.3. Polymers containing antimicrobial organic compounds. Nonco-
ery systems. Both targeted delivery to the lungs via inhalation [172]
valently linked antimicrobial agents can be released from a polymer
and local application in the eye [183] highlight the multiple advantages
matrix offering a very versatile strategy to combat device-associated in-
of liposomes to: 1) deliver biologics and hydrophilic compounds, 2) sta-
fections. Despite being one of the most common approaches to create an
bilize antibiotic compounds, 3) reduce toxicity and increase therapeutic
antimicrobial polymer, inadequate release kinetics, governed by the
index, and 4) target delivery [184,185]. Many of these advantages are
complexities of polymer/drug/solvent interactions, may inadvertently
thought to stem from their membrane-like structure that allows them
promote the evolution of bacterial resistance [131,144,145,166]. This
to fuse with the cellular membrane and deliver a drug payload directly
classification can also include physical blends or mixtures of antimicro-
to the cytoplasmic compartment, potentially saturating the bacterial
bial and non-antimicrobial polymers [167], but blending distinct com-
drug-efflux pumps and circumventing a primary mechanism of antibi-
pounds poses its own problems with homogeneity and consistency.
otic resistance [172]. Unfortunately, lipid drug delivery systems also
have limited encapsulation efficiency [175], a relatively short half-life
5.1.1.4. Polymers incorporating antimicrobial inorganic compounds. Inor- due to opsonization, filtration, and the hydrolytic instability of lipid
ganic compounds such as metals or metallic oxides can be embedded ester bonds, aggregation and fusion of liposomes, and temperature sen-
in a polymer matrix analogous to organic antibiotic agents. The history sitivity [186]. These limitations can lead to inadequate delivery partially
of antimicrobial silver, in its various forms (ions, salts, nanoparticles, due to drug leakage in circulation. Liposome stability can be improved
etc.) is deeply entwined with that of polymers, offering some of the with a variety of modifications (e.g., incorporation of hydrophilic long
most fundamental studies of antimicrobial polymers [168]. Other suc- chain polymers in the lipid bilayer, cholesterol, anionic charged groups,
cessful strategies use zinc oxide [169], gold, and titanium oxide [170]. targeting ligands, etc.) to create stealth liposomes [187]. An extensive
Regardless, almost all of these strategies employ the use of nanoparti- discussion of liposome antibiotic encapsulation and delivery optimi-
cles [171]. zation is outside the scope of this review but has been previously ad-
dressed in several reviews [175,185].
5.2. A unique local delivery system—nanoparticle/liposome delivery
5.3. Impact of local drug delivery pressure on the development of resistance
Local drug delivery systems take a variety of forms from topical
applications to coatings on biomedical implants to bulk implantable de- Local drug release systems can deliver antibiotics without incurring
vices; however, nanoparticle and liposome based drug delivery systems the same toxicity or provoking the same level of antibiotic resistance
22 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

Fig. 6. Nanoparticle mechanism of action as an antibiotic or antibiotic carrier (modified from [11]).

that plagues systemic drug administration; however, the presence of any resistance, must be to protect the natural flora of the host, as there is a
antibiotic, regardless of its route of administration, exerts some degree of relatively clear connection between protecting the host microbiome
evolutionary pressure for bacteria to develop a multidrug resistant phe- and the prevention of multidrug resistant bacterial colonization [188].
notype. This concern is particularly voiced by epidemiologists when Systemic antibiotic treatment threatens this symbiotic relationship
using antibiotic-carrier biomaterials prophylactically as there is evidence and allows opportunistic infections to flourish unchecked. Local deliv-
that prophylactic antibiotic use may contribute to the problem [122]. This ery might best achieve this goal.
view is slightly tempered by the regional prevalence of multidrug resis-
tant (MDR) species, highlighting the necessity for epidemiological sur-
veillance and clinical cognizance of regional microbiome. Furthermore, 6. Conclusions
it is important to note that both in vitro and in vivo evidence has identified
the presence of heterogeneous populations in large bacterial loads, some Infection control has long been a product of an evolutionary arms
of which are capable of surviving the initial antibiotic burst despite their race. Despite clinical evidence that small molecule monotherapy ap-
lack of antibiotic resistance genes. Shortly after the first widespread use proaches are limited in this landscape of resistance, an evaluation of
of antibiotics and local antibiotic delivery systems in humans, pre- the infection control candidates in the development pipeline indicates
existing resistant variants were recognized following exposure to lethal that research remains focused on discovery and development of new
antibiotic doses [15], indicating that otherwise healthy individuals harbor drugs. Unfortunately, as demonstrated in Tables 1 and 2, antibiotic
a small number of resident bacteria intrinsically resistant to antibiotics biotherapeutics, new combination therapies, and drug delivery mecha-
[42]. The more prevalent, antibiotic-susceptible host flora is able to keep nisms still lag behind the development of new small molecule drugs that
this small number of resistant microbes in check in the absence of antibi- are often an extension of existing drug classes. No longer are systemic
otic selective pressure. However, regardless of the route of administration, monotherapy approaches adequate in a world where resistance outpaces
emergence and proliferation of the resistant population occur upon new drug development. Thus, instead of fighting microbial evolution, in-
antibiotic administration when the normal flora decreases [42]. It is fection control efforts may be better served by targeting mechanisms of
imperative that one of the main criteria in emerging antimicrobial tech- resistance based on biological inspiration (e.g., furanones deployed by
nologies, designed to circumvent the overwhelming wave of antibiotic red sea algae to interrupt quorum sensing, etc.).
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 23

Table 1
New antimicrobial drugs in the development pipeline. Importantly, only small molecules and biologics in phase trials are included [194–196]. This table does not capture all candidates in
pre-clinical testing and development.

New antibiotic candidate pipeline [195–197]

Phase I Phase II Phase III

Candidate (company) Family Candidate (company) Family Candidate (company) Family

BAL30072 (Basilea) Monosulfactam GSK-2696266 (Shionogi/GSK) Cephem Omadacycline (Paratek) Tetracycline


XF-73 (Destiny Pharma) Porphyrin SMT-19969 (Summit) Bibenzo[d]-imidazole Eravacycline (Tetraphase) Tetracycline
NVB302 (Novacta) Lantibiotic LFF-571 (Novartis) Solithromycin (Campra) Erythromycin
POL7080 (Polyphor) Protegrin I Auriclosene (Novabay) Chlorotaurine Tedizolid phosphate (Trius Pharma) Oxazolidinone
LCB01-0371 (LegoChem) Oxazolidinone Sarecycline (Warner Chilcott) Tetracycline Delamanid (Otsuka) Nitroimidazole
MRX-I (MicuRx) Oxazolidinone BC-3781 (Forest) Pleuromutilin Perchlozone (USC Pharmasyntez) Thiosemicarbazone
ACHN-975 (Achaogen) Plazomicin (Achaogen) Aminoglycoside SQ109 (Sequlla/Infectex) Ethambutol
GSK-214094 (GSK) GSK1322322 (GSK) Actinonin Finafloxacin (MerLion) Fluoroquinolone
KPI-10 (Kalidex) Fluoroquinolone DPK-060 (Pergamum) Kinigongen Delafloxacin (Rib-X) Fluoroquinolone
DS-8587 (Daiichi) Fluoroquinolone PA-824 (Global Alliance TB Drug Avarofloxacin (Furiex) Fluoroquinolone
KRP-AM1977 (Kyorin) Quinolone Development) Nitroimidazole Zabofloxacin (Dong Wha) Fluoroquinolone
Radezolid (Rib-X) Oxazolidinone Nemonoxacin (TaiGen) Quinolone
Sutezolid (Pfizer) Oxazolidinone Ozenoxacin (Grupo Ferner) Quinolone
Posizolid (AstraZeneca) Oxazolidinone
Cadazolid (Actelion) Oxazolidinone
AFN-1252 (Affinium)
CG400549 (Crystal Genomics) Triclosan)
WCK-771 (Wockhardt) Fluoroquinolone
WCK-2349 (Wockhardt) Fluoroquinolone

Biologics
Brilacidin (PolyMedix) Defensin Dalbavancin (Durate Therapeutics) Glycopeptide
LTX-109 (Lytix Bio) Peptide Oritavancin (The Medicines Company) Glycopeptide
LL-37 (Pergamum) Cathelicidin Surotomycin (Cubist) Lipopeptide

Although use of biologics in infection control is still in its infancy, its socially, and economically will be dire [190]. Simple antibiotic steward-
potential for combating multi-drug resistance cannot be ignored. Small ship is no longer an option in the face of rising drug resistance. Clinical
molecules will always have a place in infection control; however, effec- treatments and practices for infection control must evolve in response
tive candidate development may be more intelligently pursued based to epidemiological trends in multi-drug resistant bacteria and the devel-
on biological inspiration. Drug development tools necessary to overcome opment of new treatment strategies. Hopefully, repeated emphasis will
the pitfalls of biologics, including limited in vitro stability, restricted de- promote adoption of new research strategies for infection treatments,
livery options, limited high-throughput, discovery and development including adherence to three criteria: (1) invention of efficacious new
screening tools, imperfect pharmacokinetics and relatively unexplored drugs, (2) prevention of resistance, and (3) protection of the natural
pharmacodynamics, are not as advanced as those available for small host microbiome (Fig. 2). The best strategy to meet these criteria includes
molecule development. Fortunately, limitless potential exists in the the development of new combination approaches coupled with local and
combinations of biologics, biologically inspired molecules, and drug smart delivery technologies (e.g. targeted liposomes and nanoparticles
delivery technologies. Thus, a paradigm shift akin to that necessary for and infection-responsive polymer controlled delivery [191,192]). This
complex viruses and cancers is essential to design a targeted attack paradigm shift requires a significantly more sophisticated approach to
that eradicates susceptible microbes, contains and squelches microbial antibiotic discovery and development. Even with this paradigm shift,
resistance, and protects the host flora from the therapeutic. Such a strat- humans and microbes are likely deadlocked in an evolutionary arms
egy may arise from combinations of traditional antibiotics, new adju- race; without this paradigm shift, the results are more tenuous.
vants, and sustainable localized delivery approaches. Using advanced
bioinformatics to predict successful combination delivery and novel tar-
gets may pose significant advantages over current costly development Conflict of interest statement
models that often fail in late phase trials [189].
Solutions to antibiotic resistance are not trivial to implement, with AEB discloses a financial interest in a corporate entity, Elute Inc., that
consequences affecting everyone. While solutions have been proposed, has licensed technology related to that discussed in this publication
with some even being launched to address the problem, action taken to from the University of Utah for commercial use. Elute Inc. partially
date is merely token. Antibiotics remain indispensable in all health sys- funded this work although they had no input in the design or analysis
tems, and the consequences of a lack of a coordinated effort medically, of that presented in this work.

Table 2
New combination antimicrobial therapies currently in phase trials. Other additional combination therapies not captured here are currently in preclinical development.

New antibiotic combinatory delivery pipeline [195–197]

Phase I Phase II Phase III

Candidate (company) Family Candidate (company) Family Candidate (company) Family

ATM-AVI aztreonam & Monobactam/ CXL Ceftaroline & Cephalosporin/ CXA-201—ceftolozane Cephalosporin/
avibactam (AstraZeneca) β-lactamase inhibitor avibactam (AstraZeneca) β-lactamase inhibitor & tazobactam (Cubist) β-lactamase inhibitor
Carbavance biapenem Carbapenem/ Imipenem cilastatin Carbapenem/ CAZ104 ceftazidime Cephalosporin/
&RPX7009 (Rempex) β-lactamase inhibitor & MK-7655 (Merck) β-lactamase inhibitor & avibactam (AstraZeneca) β-lactamase inhibitor
24 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

Acknowledgments [25] M.A. Sánchez-Romero, J. Casadesús, Contribution of phenotypic heterogeneity to


adaptive antibiotic resistance, Proc. Natl. Acad. Sci. U. S. A. (2013), http://dx.doi.
org/10.1073/pnas.1316084111.
This is a vast and expanding field and we apologize to those whose [26] L. Fernández, R.E.W. Hancock, Adaptive and mutational resistance: role of porins
work may have been overlooked in preparing this manuscript. Thank and efflux pumps in drug resistance, Clin. Microbiol. Rev. 25 (2012) 661–681,
http://dx.doi.org/10.1128/CMR. 00043-12.
you to Dr. David Grainger (University of Utah) for technical editing. [27] D.I. Andersson, D. Hughes, Antibiotic resistance and its cost: is it possible to reverse
resistance? Nat. Rev. Microbiol. 8 (2010) 260–271, http://dx.doi.org/10.1038/
References nrmicro2319.
[28] N. Høiby, T. Bjarnsholt, M. Givskov, S. Molin, O. Ciofu, Antibiotic resistance of bac-
[1] A. Huttner, S. Harbarth, J. Carlet, S. Cosgrove, H. Goossens, A. Holmes, et al., Antimi- terial biofilms, Int. J. Antimicrob. Agents 35 (2010) 322–332, http://dx.doi.org/10.
crobial resistance: a global view from the 2013 World Healthcare-Associated Infec- 1016/j.ijantimicag.2009.12.011.
tions Forum, Antimicrob. Resist. Infect. Control. 2 (2013) 31, http://dx.doi.org/10. [29] P.S. Stewart, J.W. Costerton, Antibiotic resistance of bacteria in biofilms, Lancet 358
1186/2047-2994-2-31. (2001) 135–138.
[2] T. Frieden, Antibiotic Resistance Threats in the United States, 2013, CDC, 2013. [30] J. Davies, D. Davies, Origins and evolution of antibiotic resistance, Microbiol. Mol.
(http://www.cdc.gov/drugresistance/threat-report-2013/pdf/ar-threats-2013- Biol. Rev. 74 (2010) 417–433, http://dx.doi.org/10.1128/MMBR. 00016-10.
508.pdf). [31] G.D. Wright, Bacterial resistance to antibiotics: enzymatic degradation and modifi-
[3] M. Bassetti, F. Ginocchio, M. Mikulska, New treatment options against gram- cation, Adv. Drug Deliv. Rev. 57 (2005) 1451–1470, http://dx.doi.org/10.1016/j.
negative organisms, Crit. Care 15 (2011) 215, http://dx.doi.org/10.1186/cc9997. addr.2005.04.002.
[4] R.R. Roberts, B. Hota, I. Ahmad, R.D. Scott, S.D. Foster, F. Abbasi, et al., Hospital and [32] M.S. Ramirez, M.E. Tolmasky, Aminoglycoside modifying enzymes, drug resist,
societal costs of antimicrobial-resistant infections in a Chicago teaching hospital: Updat. Rev. Comment. Antimicrob. Anticancer Chemother. 13 (2010) 151–171,
implications for antibiotic stewardship, Clin. Infect. Dis. 49 (2009) 1175–1184, http://dx.doi.org/10.1016/j.drup.2010.08.003.
http://dx.doi.org/10.1086/605630. [33] G.S. Tillotson, N. Theriault, New and alternative approaches to tackling antibiotic
[5] J.M. Bell, J.D. Turnidge, High prevalence of oxacillin-resistant staphylococcus aureus resistance, F1000prime Rep52013. 51, http://dx.doi.org/10.12703/P5-51.
isolates from hospitalized patients in Asia-Pacific and South Africa: results from [34] D.N. Wilson, Ribosome-targeting antibiotics and mechanisms of bacterial resistance,
SENTRY antimicrobial surveillance program, 1998–1999, Antimicrob. Agents Nat. Rev. Microbiol. 12 (2013) 35–48, http://dx.doi.org/10.1038/nrmicro3155.
Chemother. 46 (2002) 879–881, http://dx.doi.org/10.1128/AAC. 46.3.879-881.2002. [35] X.-Z. Li, H. Nikaido, Efflux-mediated drug resistance in bacteria: an update, Drugs
[6] D.J. Diekema, M.A. Pfaller, F.J. Schmitz, J. Smayevsky, J. Bell, R.N. Jones, et al., Survey 69 (2009) 1555.
of infections due to Staphylococcus species: frequency of occurrence and antimi- [36] K. Poole, A. Russell, P. Lambert, Mechanisms of antimicrobial resistance: opportuni-
crobial susceptibility of isolates collected in the United States, Canada, Latin ties for new targeted therapies, Adv. Drug Deliv. Rev. 57 (2005) 1443–1445, http://
America, Europe, and the Western Pacific region for the SENTRY Antimicrobial Sur- dx.doi.org/10.1016/j.addr.2005.05.001.
veillance Program, 1997-1999, Clin. Infect. Dis. 32 (Suppl. 2) (2001) S114–S132, [37] M.N. Alekshun, S.B. Levy, Molecular mechanisms of antibacterial multidrug resis-
http://dx.doi.org/10.1086/320184. tance, Cell 128 (2007) 1037–1050, http://dx.doi.org/10.1016/j.cell.2007.03.004.
[7] R.H. Deurenberg, C. Vink, S. Kalenic, A.W. Friedrich, C.A. Bruggeman, E.E. [38] A.M. Bal, I.M. Gould, Antibiotic stewardship: overcoming implementation barriers,
Stobberingh, The molecular evolution of methicillin-resistant Staphylococcus aureus, Curr. Opin. Infect. Dis. 24 (2011) 357–362, http://dx.doi.org/10.1097/QCO.
Clin. Microbiol. Infect. 13 (2007) 222–235, http://dx.doi.org/10.1111/j.1469-0691. 0b013e3283483262.
2006.01573.x. [39] Transatlantic Taskforce on Antimicrobial Resistance: recommendations for future
[8] H.W. Boucher, G.H. Talbot, J.S. Bradley, J.E. Edwards, D. Gilbert, L.B. Rice, et al., Bad collaboration between the US and EU, http://www.cdc.gov/drugresistance/pdf/
bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of tatfar-report.pdf (accessed April 10, 2014).
America, Clin. Infect. Dis. 48 (2009) 1–12, http://dx.doi.org/10.1086/595011. [40] Infectious Diseases Society of America (IDSA), Combating antimicrobial resistance:
[9] S.B. Tay, W.S. Yew, Development of quorum-based anti-virulence therapeutics policy recommendations to save lives, Clin. Infect. Dis. 52 (2011) S397–S428,
targeting Gram-negative bacterial pathogens, Int. J. Mol. Sci. 14 (2013) http://dx.doi.org/10.1093/cid/cir153.
16570–16599, http://dx.doi.org/10.3390/ijms140816570. [41] D.A. Goldmann, W.C. Huskins, Control of nosocomial antimicrobial-resistant bacte-
[10] A.J. Alanis, Resistance to antibiotics: are we in the post-antibiotic era? Arch. Med. ria: a strategic priority for hospitals worldwide, Clin. Infect. Dis. 24 (Suppl. 1)
Res. 36 (2005) 697–705, http://dx.doi.org/10.1016/j.arcmed.2005.06.009. (1997) S139–S145.
[11] D. Bhatt, M. Kavitha, C.K. Nisha, Y. Mahajan, Nanotechnology Solutions to Combat [42] R. Colgan, J.H. Powers, Appropriate antimicrobial prescribing: approaches that limit
Superbugs, Nanowerk, 2013. (http://www.nanowerk.com/spotlight/spotid= antibiotic resistance, Am. Fam. Physician 64 (2001) 999–1004.
32188.php). [43] B. LaSarre, M.J. Federle, Exploiting quorum sensing to confuse bacterial pathogens,
[12] Y. Hu, A. Shamaei-Tousi, Y. Liu, A. Coates, A new approach for the discovery of Microbiol. Mol. Biol. Rev. 77 (2013) 73–111, http://dx.doi.org/10.1128/MMBR.
antibiotics by targeting non-multiplying bacteria: a novel topical antibiotic for 00046-12.
staphylococcal infections, PLoS ONE 5 (2010), http://dx.doi.org/10.1371/journal. [44] M. Chen, Q. Yu, H. Sun, Novel strategies for the prevention and treatment of biofilm
pone.0011818. related infections, Int. J. Mol. Sci. 14 (2013) 18488–18501, http://dx.doi.org/10.
[13] J. Davies, Bacteria on the rampage, Nature 383 (1996) 219–220, http://dx.doi.org/ 3390/ijms140918488.
10.1038/383219a0. [45] H. Hirakawa, H. Tomita, Interference of bacterial cell-to-cell communication: a new
[14] C. Walsh, Molecular mechanisms that confer antibacterial drug resistance, Nature concept of antimicrobial chemotherapy breaks antibiotic resistance, Front.
406 (2000) 775–781, http://dx.doi.org/10.1038/35021219. Microbiol. 4 (2013) 114, http://dx.doi.org/10.3389/fmicb.2013.00114.
[15] A. Rodríguez-Rojas, J. Rodríguez-Beltrán, A. Couce, J. Blázquez, Antibiotics and [46] S.A. Al-Bataineh, R. Luginbuehl, M. Textor, M. Yan, Covalent immobilization of
antibiotic resistance: a bitter fight against evolution, Int. J. Med. Microbiol. 303 antibacterial furanones via photochemical activation of perfluorophenylazide,
(2013) 293–297, http://dx.doi.org/10.1016/j.ijmm.2013.02.004. Langmuir 25 (2009) 7432–7437, http://dx.doi.org/10.1021/la900334w.
[16] E. Toprak, A. Veres, J.-B. Michel, R. Chait, D.L. Hartl, R. Kishony, Evolutionary paths [47] J.K. Baveja, G. Li, R.E. Nordon, E.B.H. Hume, N. Kumar, M.D.P. Willcox, et al., Biolog-
to antibiotic resistance under dynamically sustained drug selection, Nat. Genet. 44 ical performance of a novel synthetic furanone-based antimicrobial, Biomaterials
(2012) 101–105, http://dx.doi.org/10.1038/ng.1034. 25 (2004) 5013–5021, http://dx.doi.org/10.1016/j.biomaterials.2004.02.007.
[17] H.H. Lee, M.N. Molla, C.R. Cantor, J.J. Collins, Bacterial charity work leads to [48] J. Pan, D. Ren, Structural effects on persister control by brominated furanones, Bioorg.
population-wide resistance, Nature 467 (2010) 82–85, http://dx.doi.org/10.1038/ Med. Chem. Lett. 23 (2013) 6559–6562, http://dx.doi.org/10.1016/j.bmcl.2013.10.070.
nature09354. [49] L. Yujie, X. Geng, Y. Huang, Y. Li, K. Yang, L. Ye, et al., The effect of brominated
[18] I. Vranakis, I. Goniotakis, A. Psaroulaki, V. Sandalakis, Y. Tselentis, K. Gevaert, et al., furanones on the formation of Staphylococcus aureus biofilm on PVC, Cell Biochem.
Proteome studies of bacterial antibiotic resistance mechanisms, J. Proteomics Biophys. 67 (2013) 1501–1505, http://dx.doi.org/10.1007/s12013-013-9652-2.
(2013), http://dx.doi.org/10.1016/j.jprot.2013.10.027. [50] O. Cirioni, F. Mocchegiani, I. Cacciatore, J. Vecchiet, C. Silvestri, L. Baldassarre, et al.,
[19] J.C. Kester, S.M. Fortune, Persisters and beyond: mechanisms of phenotypic drug Quorum sensing inhibitor FS3-coated vascular graft enhances daptomycin efficacy
resistance and drug tolerance in bacteria, Crit. Rev. Biochem. Mol. Biol. (2013), in a rat model of staphylococcal infection, Peptides 40 (2013) 77–81, http://dx.doi.
http://dx.doi.org/10.3109/10409238.2013.869543. org/10.1016/j.peptides.2012.12.002.
[20] P.C.Y. Woo, A.P.C. To, S.K.P. Lau, K.Y. Yuen, Facilitation of horizontal transfer of an- [51] L.D. Christensen, M. van Gennip, T.H. Jakobsen, M. Alhede, H.P. Hougen, N. Høiby,
timicrobial resistance by transformation of antibiotic-induced cell-wall-deficient et al., Synergistic antibacterial efficacy of early combination treatment with
bacteria, Med. Hypotheses 61 (2003) 503–508, http://dx.doi.org/10.1016/S0306- tobramycin and quorum-sensing inhibitors against Pseudomonas aeruginosa in an
9877(03)00205-6. intraperitoneal foreign-body infection mouse model, J. Antimicrob. Chemother.
[21] A. Norman, L.H. Hansen, S.J. Sørensen, Conjugative plasmids: vessels of the com- 67 (2012) 1198–1206, http://dx.doi.org/10.1093/jac/dks002.
munal gene pool, Philos. Trans. R. Soc. Lond. B Biol. Sci. 364 (2009) 2275–2289, [52] N. Balaban, A. Giacometti, O. Cirioni, Y. Gov, R. Ghiselli, F. Mocchegiani, et al., Use of
http://dx.doi.org/10.1098/rstb.2009.0037. the quorum-sensing inhibitor RNAIII-inhibiting peptide to prevent biofilm forma-
[22] J.L. Martinez, F. Baquero, Mutation frequencies and antibiotic resistance, tion in vivo by drug-resistant Staphylococcus epidermidis, J. Infect. Dis. 187 (2003)
Antimicrob. Agents Chemother. 44 (2000) 1771–1777. 625–630, http://dx.doi.org/10.1086/345879.
[23] R.T. Cirz, J.K. Chin, D.R. Andes, V. de Crecy-Lagard, W.A. Craig, F.E. Romesberg, Inhi- [53] A.K. Bhardwaj, K. Vinothkumar, N. Rajpara, Bacterial quorum sensing inhibitors:
bition of mutation and combating the evolution of antibiotic resistance, PLoS Biol. 3 attractive alternatives for control of infectious pathogens showing multiple drug
(2005), http://dx.doi.org/10.1371/journal.pbio.0030176. resistance, Recent Pat. Antiinfect. Drug Discov. 8 (2013) 68–83.
[24] M.A. Kohanski, M.A. DePristo, J.J. Collins, Sublethal antibiotic treatment leads to multi- [54] J.P. Gerdt, H.E. Blackwell, Competition studies confirm two major barriers that can
drug resistance via radical-induced mutagenesis, Mol. Cell 37 (2010) 311–320, http:// preclude the spread of resistance to quorum-sensing inhibitors in bacteria, ACS
dx.doi.org/10.1016/j.molcel.2010.01.003. Chem. Biol. (2014), http://dx.doi.org/10.1021/cb5004288.
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 25

[55] S. Scutera, M. Zucca, D. Savoia, Novel approaches for the design and discovery of [84] M. Dybul, A.S. Fauci, J.G. Bartlett, J.E. Kaplan, A.K. Pau, Panel on Clinical Practices for
quorum-sensing inhibitors, Expert Opin. Drug Discov. 9 (2014) 353–366, http:// Treatment of HIV, Guidelines for using antiretroviral agents among HIV-infected
dx.doi.org/10.1517/17460441.2014.894974. adults and adolescents, Ann. Intern. Med. 137 (2002) 381–433.
[56] F. Guilhelmelli, N. Vilela, P. Albuquerque, L. da, S. Derengowski, I. Silva-Pereira, C.M. [85] G.J. Peters, C.L. van der Wilt, C.J.A. van Moorsel, J.R. Kroep, A.M. Bergman, S.P.
Kyaw, Antibiotic development challenges: the various mechanisms of action of Ackland, Basis for effective combination cancer chemotherapy with antimetabo-
antimicrobial peptides and of bacterial resistance, Front. Microbiol. 4 (2013) 353, lites, Pharmacol. Ther. 87 (2000) 227–253, http://dx.doi.org/10.1016/S0163-
http://dx.doi.org/10.3389/fmicb.2013.00353. 7258(00)00086-3.
[57] J. Vizioli, M. Salzet, Antimicrobial peptides from animals: focus on invertebrates, [86] N. Phougat, S. Khatri, A. Singh, M. Dangi, M. Kumar, R. Dabur, et al., Combination
Trends Pharmacol. Sci. 23 (2002) 494–496. therapy: the propitious rationale for drug development, Comb. Chem. High
[58] K. Lohner, E.J. Prenner, Differential scanning calorimetry and X-ray diffraction stud- Throughput Screen. 17 (1) (2013) 53–67.
ies of the specificity of the interaction of antimicrobial peptides with membrane- [87] R. Pena-Miller, D. Laehnemann, G. Jansen, A. Fuentes-Hernandez, P. Rosenstiel, H.
mimetic systems, Biochim. Biophys. Acta 1462 (1999) 141–156. Schulenburg, et al., When the most potent combination of antibiotics selects for
[59] J.H. Fritz, S. Brunner, M.L. Birnstiel, M. Buschle, A. v Gabain, F. Mattner, et al., The the greatest bacterial load: the smile–frown transition, PLoS Biol. 11 (2013),
artificial antimicrobial peptide KLKLLLLLKLK induces predominantly a TH2-type http://dx.doi.org/10.1371/journal.pbio.1001540.
immune response to co-injected antigens, Vaccine 22 (2004) 3274–3284, http:// [88] A.M. Allahverdiyev, K.V. Kon, E.S. Abamor, M. Bagirova, M. Rafailovich, Coping with
dx.doi.org/10.1016/j.vaccine.2004.03.007. antibiotic resistance: combining nanoparticles with antibiotics and other antimi-
[60] F. Costa, I.F. Carvalho, R.C. Montelaro, P. Gomes, M.C.L. Martins, Covalent immobi- crobial agents, Expert Rev. Anti-Infect. Ther. 9 (2011) 1035–1052, http://dx.doi.
lization of antimicrobial peptides (AMPs) onto biomaterial surfaces, Acta Biomater. org/10.1586/eri.11.121.
7 (2011) 1431–1440, http://dx.doi.org/10.1016/j.actbio.2010.11.005. [89] M.A. Fischbach, Combination therapies for combating antimicrobial resistance, Curr.
[61] D. Alves, M. Olívia Pereira, Mini-review: antimicrobial peptides and enzymes as Opin. Microbiol. 14 (2011) 519–523, http://dx.doi.org/10.1016/j.mib.2011.08.003.
promising candidates to functionalize biomaterial surfaces, Biofouling (2014), [90] R. Hamoud, S. Zimmermann, J. Reichling, M. Wink, Synergistic interactions in two-
http://dx.doi.org/10.1080/08927014.2014.889120. drug and three-drug combinations (thymol, EDTA and vancomycin) against multi
[62] A. Peschel, H.-G. Sahl, The co-evolution of host cationic antimicrobial peptides and drug resistant bacteria including E. coli, Phytomedicine (2013), http://dx.doi.org/
microbial resistance, Nat. Rev. Microbiol. 4 (2006) 529–536, http://dx.doi.org/10. 10.1016/j.phymed.2013.10.016.
1038/nrmicro1441. [91] J.K. Smith, A.R. Moshref, J.A. Jennings, H.S. Courtney, W.O. Haggard, Chitosan
[63] F. Dorner, K. Lienkamp, CHAPTER 5. Polymer-based synthetic mimics of antimicro- sponges for local synergistic infection therapy: a pilot study, Clin. Orthop. 471
bial peptides (SMAMPs) &#8211, in: A. Muñoz-Bonilla, M. Cerrada, M. (2013) 3158–3164, http://dx.doi.org/10.1007/s11999-013-2988-5.
Fernández-García (Eds.), A New Class of Nature-Inspired Antimicrobial Agents [92] L.A. Sampath, S.M. Tambe, S.M. Modak, In vitro and in vivo efficacy of catheters im-
with Low Bacterial Resistance Formation Potential, RSC Polym. Chem. Ser., Royal pregnated with antiseptics or antibiotics: evaluation of the risk of bacterial resis-
Society of Chemistry, Cambridge, 2013, pp. 97–138 (http://ebook.rsc.org/?DOI= tance to the antimicrobials in the catheters •, Infect. Control Hosp. Epidemiol. 22
10.1039/9781782624998-00097 (accessed April 8, 2014)). (2001) 640–646, http://dx.doi.org/10.1086/501836.
[64] S. Lohan, G.S. Bisht, Recent approaches in design of peptidomimetics for antimicro- [93] L. Ejim, M.A. Farha, S.B. Falconer, J. Wildenhain, B.K. Coombes, M. Tyers, et al., Com-
bial drug discovery research, Mini-Rev. Med. Chem. 13 (2013) 1073–1088. binations of antibiotics and nonantibiotic drugs enhance antimicrobial efficacy,
[65] G.N. Tew, R.W. Scott, M.L. Klein, W.F. DeGrado, De novo design of antimicrobial Nat. Chem. Biol. 7 (2011) 348–350, http://dx.doi.org/10.1038/nchembio.559.
polymers, foldamers, and small molecules: from discovery to practical applica- [94] G. Müller, A. Kramer, Biocompatibility index of antiseptic agents by parallel assess-
tions, Acc. Chem. Res. 43 (2010) 30–39, http://dx.doi.org/10.1021/ar900036b. ment of antimicrobial activity and cellular cytotoxicity, J. Antimicrob. Chemother.
[66] G.N. Tew, D. Liu, B. Chen, R.J. Doerksen, J. Kaplan, P.J. Carroll, et al., De novo design 61 (2008) 1281–1287, http://dx.doi.org/10.1093/jac/dkn125.
of biomimetic antimicrobial polymers, Proc. Natl. Acad. Sci. 99 (2002) 5110–5114, [95] G. McDonnell, A.D. Russell, Antiseptics and disinfectants: activity, action, and resis-
http://dx.doi.org/10.1073/pnas.082046199. tance, Clin. Microbiol. Rev. 12 (1999) 147–179.
[67] H.D. Thaker, F. Sgolastra, D. Clements, R.W. Scott, G.N. Tew, Synthetic mimics of [96] K. Lewis, Persister cells and the riddle of biofilm survival, Biochem. Biokhim. 70
antimicrobial peptides from triaryl scaffolds, J. Med. Chem. 54 (2011) 2241–2254, (2005) 267–274.
http://dx.doi.org/10.1021/jm101410t. [97] S.M. Drawz, K.M. Papp-Wallace, R.A. Bonomo, New β-lactamase inhibitors: a ther-
[68] R.E.W. Hancock, H.-G. Sahl, Antimicrobial and host-defense peptides as new anti- apeutic renaissance in an MDR world, Antimicrob. Agents Chemother. 58 (2014)
infective therapeutic strategies, Nat. Biotechnol. 24 (2006) 1551–1557, http://dx. 1835–1846, http://dx.doi.org/10.1128/AAC. 00826-13.
doi.org/10.1038/nbt1267. [98] T. Roemer, T. Schneider, M.G. Pinho, Auxiliary factors: a chink in the armor of
[69] G.J. Gabriel, A. Som, A.E. Madkour, T. Eren, G.N. Tew, Infectious disease: connecting MRSA resistance to β-lactam antibiotics, Curr. Opin. Microbiol. 16 (2013)
innate immunity to biocidal polymers, Mater. Sci. Eng. R. Rep. 57 (2007) 28–64, 538–548, http://dx.doi.org/10.1016/j.mib.2013.06.012.
http://dx.doi.org/10.1016/j.mser.2007.03.002. [99] O. Lomovskaya, M.S. Warren, A. Lee, J. Galazzo, R. Fronko, M. Lee, et al., Identifica-
[70] M. Sabouri Ghannad, A. Mohammadi, Bacteriophage: time to re-evaluate the tion and characterization of inhibitors of multidrug resistance efflux pumps in
potential of phage therapy as a promising agent to control multidrug-resistant Pseudomonas aeruginosa: novel agents for combination therapy, Antimicrob.
bacteria, Iran. J. Basic Med. Sci. 15 (2012) 693–701. Agents Chemother. 45 (2001) 105–116, http://dx.doi.org/10.1128/AAC. 45.1.105-
[71] J. Bradbury, “My enemy's enemy is my friend”. Using phages to fight bacteria, Lancet 116.2001.
363 (2004) 624–625. [100] A.M. Kalle, A. Rizvi, Inhibition of bacterial multidrug resistance by celecoxib, a
[72] B. Burrowes, D.R. Harper, J. Anderson, M. McConville, M.C. Enright, Bacteriophage cyclooxygenase-2 inhibitor, Antimicrob. Agents Chemother. 55 (2011) 439–442,
therapy: potential uses in the control of antibiotic-resistant pathogens, Expert http://dx.doi.org/10.1128/AAC. 00735-10.
Rev. Anti-Infect. Ther. 9 (2011) 775–785, http://dx.doi.org/10.1586/eri.11.90. [101] C. Kourtesi, A.R. Ball, Y.-Y. Huang, S.M. Jachak, D.M.A. Vera, P. Khondkar, et al., Mi-
[73] M.D. Mathur, S. Vidhani, P.L. Mehndiratta, Bacteriophage therapy: an alternative to crobial efflux systems and inhibitors: approaches to drug discovery and the chal-
conventional antibiotics, J. Assoc. Physicians India 51 (2003) 593–596. lenge of clinical implementation, Open Microbiol. J. 7 (2013) 34–52, http://dx.
[74] B.R. Levin, J.J. Bull, Population and evolutionary dynamics of phage therapy, Nat. doi.org/10.2174/1874285801307010034.
Rev. Microbiol. 2 (2004) 166–173, http://dx.doi.org/10.1038/nrmicro822. [102] K. Markoishvili, G. Tsitlanadze, R. Katsarava, J. Glenn, A. Sulakvelidze, A novel
[75] H. Brüssow, Phage therapy: the Escherichia coli experience, Microbiol. Read. Eng. sustained-release matrix based on biodegradable poly(ester amide)s and impreg-
151 (2005) 2133–2140, http://dx.doi.org/10.1099/mic. 0.27849-0. nated with bacteriophages and an antibiotic shows promise in management of
[76] R.D. Joerger, Alternatives to antibiotics: bacteriocins, antimicrobial peptides and infected venous stasis ulcers and other poorly healing wounds, Int. J. Dermatol.
bacteriophages, Poult. Sci. 82 (2003) 640–647. 41 (2002) 453–458, http://dx.doi.org/10.1046/j.1365-4362.2002.01451.x.
[77] A.M. Comeau, F. Tétart, S.N. Trojet, M.-F. Prère, H.M. Krisch, Phage-Antibiotic [103] N.K. Petty, T.J. Evans, P.C. Fineran, G.P.C. Salmond, Biotechnological exploitation of
Synergy (PAS): beta-lactam and quinolone antibiotics stimulate virulent bacteriophage research, Trends Biotechnol. 25 (2007) 7–15, http://dx.doi.org/10.
phage growth, PLoS One 2 (2007) e799, http://dx.doi.org/10.1371/journal. 1016/j.tibtech.2006.11.003.
pone.0000799. [104] K. Joshi-Navare, A. Prabhune, A biosurfactant-sophorolipid acts in synergy with an-
[78] E. Pajtasz-Piasecka, J. Rossowska, D. Duś, B. Weber-Dąbrowska, A. Zabłocka, A. tibiotics to enhance their efficiency, Biomed. Res. Int. 2013 (2013) 512495, http://
Górski, Bacteriophages support anti-tumor response initiated by DC-based vaccine dx.doi.org/10.1155/2013/512495.
against murine transplantable colon carcinoma, Immunol. Lett. 116 (2008) 24–32, [105] N.A. Barekzi, A.G. Felts, K.A. Poelstra, J.B. Slunt, D.W. Grainger, Locally delivered
http://dx.doi.org/10.1016/j.imlet.2007.11.004. polyclonal antibodies potentiate intravenous antibiotic efficacy against gram-
[79] A.E. Willis, R.N. Perham, D. Wraith, Immunological properties of foreign peptides in negative infections, Pharm. Res. 19 (2002) 1801–1807.
multiple display on a filamentous bacteriophage, Gene 128 (1993) 79–83. [106] D.J. Payne, M.N. Gwynn, D.J. Holmes, D.L. Pompliano, Drugs for bad bugs:
[80] Y.-H. Chen, C.-F. Peng, P.-L. Lu, J.-J. Tsai, T.-P. Chen, In vitro activities of antibiotic confronting the challenges of antibacterial discovery, Nat. Rev. Drug Discov. 6
combinations against clincal isolates of Pseudomonas aeruginosa, Kaohsiung J. (2007) 29–40, http://dx.doi.org/10.1038/nrd2201.
Med. Sci. 20 (2004) 261–267. [107] A.F. Read, S. Huijben, Evolutionary biology and the avoidance of antimicrobial re-
[81] R.J. Worthington, C. Melander, Combination approaches to combat multidrug- sistance, Evol. Appl. 2 (2009) 40–51, http://dx.doi.org/10.1111/j.1752-4571.2008.
resistant bacteria, Trends Biotechnol. 31 (2013) 177–184, http://dx.doi.org/10. 00066.x.
1016/j.tibtech.2012.12.006. [108] Z. Ahmed, S. Saeed Khan, M. Khan, In vitro trials of some antimicrobial combina-
[82] P.D. Tamma, S.E. Cosgrove, L.L. Maragakis, Combination therapy for treatment of tions against Staphylococcus aureus and Pseudomonas aeruginosa, Saudi J. Biol. Sci.
infections with gram-negative bacteria, Clin. Microbiol. Rev. 25 (2012) 450–470, 20 (2013) 79–83, http://dx.doi.org/10.1016/j.sjbs.2012.10.005.
http://dx.doi.org/10.1128/CMR. 05041-11. [109] E. Moran, S. Masters, A.R. Berendt, P. McLardy-Smith, I. Byren, B.L. Atkins, Guiding
[83] P. Wu, D.W. Grainger, Drug/device combinations for local drug therapies and infec- empirical antibiotic therapy in orthopaedics: the microbiology of prosthetic joint
tion prophylaxis, Biomaterials 27 (2006) 2450–2467, http://dx.doi.org/10.1016/j. infection managed by debridement, irrigation and prosthesis retention, J. Infect.
biomaterials.2005.11.031. 55 (2007) 1–7, http://dx.doi.org/10.1016/j.jinf.2007.01.007.
26 B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27

[110] C.E. Marculescu, J.R. Cantey, Polymicrobial prosthetic joint infections: risk factors [140] M. Stigter, J. Bezemer, K. de Groot, P. Layrolle, Incorporation of different antibiotics
and outcome, Clin. Orthop. 466 (2008) 1397–1404, http://dx.doi.org/10.1007/ into carbonated hydroxyapatite coatings on titanium implants, release and antibi-
s11999-008-0230-7. otic efficacy, J. Control. Release 99 (2004) 127–137, http://dx.doi.org/10.1016/j.
[111] V.K. Aggarwal, C. Higuera, G. Deirmengian, J. Parvizi, M.S. Austin, Swab cultures are jconrel.2004.06.011.
not as effective as tissue cultures for diagnosis of periprosthetic joint infection, Clin. [141] S. Ozaki, A. Saito, H. Nakaminami, M. Ono, N. Noguchi, N. Motomura, Comprehen-
Orthop. 471 (2013) 3196–3203, http://dx.doi.org/10.1007/s11999-013-2974-y. sive evaluation of fibrin glue as a local drug-delivery system-efficacy and safety of
[112] K. Wood, S. Nishida, E.D. Sontag, P. Cluzel, Mechanism-independent method for sustained release of vancomycin by fibrin glue against local methicillin-resistant
predicting response to multidrug combinations in bacteria, Proc. Natl. Acad. Sci. Staphylococcus aureus infection, J. Artif. Organs (2013), http://dx.doi.org/10.1007/
U. S. A. 109 (2012) 12254–12259, http://dx.doi.org/10.1073/pnas.1201281109. s10047-013-0746-9.
[113] P.J. Yeh, M.J. Hegreness, A.P. Aiden, R. Kishony, Drug interactions and the evolution [142] K. Tofuku, H. Koga, M. Yanase, S. Komiya, The use of antibiotic-impregnated fibrin
of antibiotic resistance, Nat. Rev. Microbiol. 7 (2009) 460–466, http://dx.doi.org/ sealant for the prevention of surgical site infection associated with spinal instru-
10.1038/nrmicro2133. mentation, Eur. Spine J. 21 (2012) 2027–2033, http://dx.doi.org/10.1007/s00586-
[114] M. Hegreness, N. Shoresh, D. Damian, D. Hartl, R. Kishony, Accelerated evolution of 012-2435-4.
resistance in multidrug environments, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) [143] I.A. Rojas, J.B. Slunt, D.W. Grainger, Polyurethane coatings release bioactive anti-
13977–13981, http://dx.doi.org/10.1073/pnas.0805965105. bodies to reduce bacterial adhesion, J. Control. Release 63 (2000) 175–189.
[115] R. Pena-Miller, D. Lahnemann, H. Schulenburg, M. Ackermann, R. Beardmore, The [144] A.E. Brooks, B.D. Brooks, S.N. Davidoff, P.C. Hogrebe, M.A. Fisher, D.W. Grainger,
optimal deployment of synergistic antibiotics: a control-theoretic approach, J. R. Polymer-controlled release of tobramycin from bone graft void filler, Drug Deliv.
Soc. Interface 9 (2012) 2488–2502, http://dx.doi.org/10.1098/rsif.2012.0279. Transl. Res. 3 (2013) 518–530, http://dx.doi.org/10.1007/s13346-013-0155-x.
[116] V. Lázár, G. Pal Singh, R. Spohn, I. Nagy, B. Horváth, M. Hrtyan, et al., Bacterial evo- [145] B.D. Brooks, K.D. Sinclair, S.N. Davidoff, S. Lawson, A.G. Williams, B. Coats, et al.,
lution of antibiotic hypersensitivity, Mol. Syst. Biol. 9 (2013) 700, http://dx.doi.org/ Molded polymer-coated composite bone void filler improves tobramycin con-
10.1038/msb.2013.57. trolled release kinetics, J. Biomed. Mater. Res. B Appl. Biomater. (2013), http://dx.
[117] L. Kalan, G.D. Wright, Antibiotic adjuvants: multicomponent anti-infective strategies, doi.org/10.1002/jbm.b.33089.
Expert Rev. Mol. Med. 13 (2011), http://dx.doi.org/10.1017/S1462399410001766 [146] N. Ahola, N. Mannisto, M. Veiranto, M. Karp, J. Rich, A. Efimov, et al., An in vitro
(null–null). study of composites of poly(L-lactide-co-?-caprolactone),5?-tricalcium phosphate
[118] D.E. Goldberg, R.F. Siliciano, W.R. Jacobs Jr., Outwitting evolution: fighting drug- and ciprofloxacin intended for local treatment of osteomyelitis, Biomaterials 3
resistant TB, malaria, and HIV, Cell 148 (2012) 1271–1283, http://dx.doi.org/10. (2013), http://dx.doi.org/10.4161/biom.23162.
1016/j.cell.2012.02.021. [147] A. Cavallaro, S. Taheri, K. Vasilev, Responsive and “smart” antibacterial surfaces:
[119] H.E. Hasper, N.E. Kramer, J.L. Smith, J.D. Hillman, C. Zachariah, O.P. Kuipers, et al., An common approaches and new developments, Biointerphases 9 (2014) 029005,
alternative bactericidal mechanism of action for lantibiotic peptides that target lipid http://dx.doi.org/10.1116/1.4866697.
II, Science 313 (2006) 1636–1637, http://dx.doi.org/10.1126/science.1129818. [148] K. Vasilev, J. Cook, H.J. Griesser, Antibacterial surfaces for biomedical devices,
[120] T. Bollenbach, S. Quan, R. Chait, R. Kishony, Non-optimal microbial response to an- Expert Rev. Med. Devices 6 (2009) 553–567.
tibiotics underlies suppressive drug interactions, Cell 139 (2009) 707–718, http:// [149] K. Vasilev, V. Sah, K. Anselme, C. Ndi, M. Mateescu, B. Dollmann, et al., Tunable an-
dx.doi.org/10.1016/j.cell.2009.10.025. tibacterial coatings that support mammalian cell growth, Nano Lett. 10 (2010)
[121] B. Brooks, A. Brooks, D.W. Grainger, Antimicrobial technologies in preclinical and 202–207, http://dx.doi.org/10.1021/nl903274q.
clinical medical devices, Biomater. Assoc. Infect. Immunol. Asp. Antimicrob. Strateg. [150] F. Siedenbiedel, J.C. Tiller, Antimicrobial polymers in solution and on surfaces:
N. Y. Springer (2012) 307–354. overview and functional principles, Polymers 4 (2012) 46–71, http://dx.doi.org/
[122] D. Campoccia, L. Montanaro, P. Speziale, C.R. Arciola, Antibiotic-loaded biomaterials 10.3390/polym4010046.
and the risks for the spread of antibiotic resistance following their prophylactic and [151] L. Timofeeva, N. Kleshcheva, Antimicrobial polymers: mechanism of action, factors
therapeutic clinical use, Biomaterials 31 (2010) 6363–6377, http://dx.doi.org/10. of activity, and applications, Appl. Microbiol. Biotechnol. 89 (2011) 475–492,
1016/j.biomaterials.2010.05.005. http://dx.doi.org/10.1007/s00253-010-2920-9.
[123] S.P. Noel, H.S. Courtney, J.D. Bumgardner, W.O. Haggard, Chitosan sponges to locally [152] K. Kuroda, G.A. Caputo, Antimicrobial polymers as synthetic mimics of host-
deliver amikacin and vancomycin: a pilot in vitro evaluation, Clin. Orthop. 468 defense peptides, Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 5 (2013)
(2010) 2074–2080, http://dx.doi.org/10.1007/s11999-010-1324-6. 49–66, http://dx.doi.org/10.1002/wnan.1199.
[124] D.J. Stinner, S.P. Noel, W.O. Haggard, J.T. Watson, J.C. Wenke, Local antibiotic deliv- [153] S. Ng, B. Goodson, A. Ehrhardt, W.H. Moos, M. Siani, J. Winter, Combinatorial dis-
ery using tailorable chitosan sponges: the future of infection control? J. Orthop. covery process yields antimicrobial peptoids, Bioorg. Med. Chem. 7 (1999)
Trauma 24 (2010) 592–597, http://dx.doi.org/10.1097/BOT.0b013e3181ed296c. 1781–1785, http://dx.doi.org/10.1016/S0968-0896(99)00132-7.
[125] J.S. Price, A.F. Tencer, D.M. Arm, G.A. Bohach, Controlled release of antibiotics from [154] S. Fernandez-Lopez, H.-S. Kim, E.C. Choi, M. Delgado, J.R. Granja, A. Khasanov, et al.,
coated orthopedic implants, J. Biomed. Mater. Res. 30 (1996) 281–286, http://dx. Antibacterial agents based on the cyclic d, l-α-peptide architecture, Nature 412
doi.org/10.1002/(SICI)1097-4636(199603)30:3b281::AID-JBM2N3.0.CO;2-M. (2001) 452–455, http://dx.doi.org/10.1038/35086601.
[126] M. Zilberman, A. Kraitzer, O. Grinberg, J.J. Elsner, Drug-eluting medical implants, [155] X. Ren, C. Zhu, L. Kou, S.D. Worley, H.B. Kocer, R.M. Broughton, et al., Acyclic N-
Handb. Exp. Pharmacol. 299–341 (2010), http://dx.doi.org/10.1007/978-3-642- halamine polymeric biocidal films, J. Bioact. Compat. Polym. 25 (2010) 392–405,
00477-3_11. http://dx.doi.org/10.1177/0883911510370387.
[127] M. Zilberman, J.J. Elsner, Antibiotic-eluting medical devices for various applica- [156] G.N. Tew, D. Clements, H. Tang, L. Arnt, R.W. Scott, Antimicrobial activity of an abi-
tions, J. Control. Release 130 (2008) 202–215, http://dx.doi.org/10.1016/j.jconrel. otic host defense peptide mimic, Biochim. Biophys. Acta 1758 (2006) 1387–1392,
2008.05.020. http://dx.doi.org/10.1016/j.bbamem.2006.03.001.
[128] M.B. Colton, E. Ehrlich, Bactericidal effect obtained by addition of antibiotics to den- [157] M. Gorbunova, Novel guanidinium and phosphonium polysulfones: synthesis and
tal cements and direct filling resins, J. Am. Dent. Assoc. 1939 (47) (1953) 524–531. antimicrobial activity, J. Chem. Pharm. Res. 5 (2013) 185–192.
[129] F. Munson, D. Heron, Facial resconstruction with acrylic resin, Am. J. Surg. 3 (1941) [158] A.M. Carmona-Ribeiro, L.D. de Melo Carrasco, Cationic antimicrobial polymers and
18–21. their assemblies, Int. J. Mol. Sci. 14 (2013) 9906–9946, http://dx.doi.org/10.3390/
[130] R.P. Howlin, M.J. Brayford, J.S. Webb, J.J. Cooper, S.S. Aiken, P. Stoodley, Antibiotic- ijms14059906.
loaded synthetic calcium sulfate beads for the prevention of bacterial colonisation [159] R.J. Cornell, L.G. Donaruma, 2-Methacryloxytropones. intermediates for the synthe-
and biofilm formation in periprosthetic infections, Antimicrob. Agents Chemother. sis of biologically active polymers, J. Med. Chem. 8 (1965) 388–390.
(2014), http://dx.doi.org/10.1128/AAC.03676-14. [160] B. Dizman, M.O. Elasri, L.J. Mathias, Synthesis, characterization, and antibacterial ac-
[131] H.L. Tan, W.T. Lin, T.T. Tang, The use of antimicrobial-impregnated PMMA to man- tivities of novel methacrylate polymers containing norfloxacin, Biomacromolecules
age periprosthetic infections: controversial issues and the latest developments, Int. 6 (2005) 514–520, http://dx.doi.org/10.1021/bm049383.
J. Artif. Organs 35 (2012) 832–839, http://dx.doi.org/10.5301/ijao.5000163. [161] M.C. Lawson, R. Shoemaker, K.B. Hoth, C.N. Bowman, K.S. Anseth, Polymerizable
[132] J. Kopecek, Soluble biomedical polymers, Polim. Med. 7 (1977) 191–221. vancomycin derivatives for bactericidal biomaterial surface modification: struc-
[133] A. Gristina, Biomaterial-centered infection: microbial adhesion versus tissue inte- ture–function evaluation, Biomacromolecules 10 (2009) 2221–2234, http://dx.
gration1987 Clin. Orthop. (2004) 4–12. doi.org/10.1021/bm900410a.
[134] C.G. Zalavras, M.J. Patzakis, P. Holtom, Local antibiotic therapy in the treatment of [162] V.E. Wagner, J.T. Koberstein, J.D. Bryers, Protein and bacterial fouling characteristics
open fractures and osteomyelitis, Clin. Orthop. (2004) 86–93. of peptide and antibody decorated surfaces of PEG-poly(acrylic acid) co-polymers,
[135] Z. Ruszczak, W. Friess, Collagen as a carrier for on-site delivery of antibacterial Biomaterials 25 (2004) 2247–2263.
drugs, Adv. Drug Deliv. Rev. 55 (2003) 1679–1698. [163] B. Gottenbos, H.C. van der Mei, F. Klatter, P. Nieuwenhuis, H.J. Busscher, In vitro and
[136] M.N. Mavros, P.K. Mitsikostas, V.G. Alexiou, G. Peppas, M.E. Falagas, Gentamicin in vivo antimicrobial activity of covalently coupled quaternary ammonium silane
collagen sponges for the prevention of sternal wound infection: a meta-analysis of coatings on silicone rubber, Biomaterials 23 (2002) 1417–1423, http://dx.doi.org/
randomized controlled trials, J. Thorac. Cardiovasc. Surg. 144 (2012) 1235–1240, 10.1016/S0142-9612(01)00263-0.
http://dx.doi.org/10.1016/j.jtcvs.2012.06.040. [164] A.E. Madkour, J.M. Dabkowski, K. Nusslein, G.N. Tew, Fast disinfecting antimicrobial
[137] J. Cai, Y. Zhang, W. Du, K. Nan, Preparation and in vitro release of spray-dried chi- surfaces, Langmuir 25 (2009) 1060–1067, http://dx.doi.org/10.1021/la802953v.
tosan microspheres for levofloxacin delivery, J. Control. Release 152 (2011) [165] L.G. Harris, S. Tosatti, M. Wieland, M. Textor, R.G. Richards, Staphylococcus au-
e70–e71, http://dx.doi.org/10.1016/j.jconrel.2011.08.129. reus adhesion to titanium oxide surfaces coated with non-functionalized and
[138] W. Lv, J. Luo, Y. Deng, Y. Sun, Biomaterials immobilized with chitosan for recharge- peptide-functionalized poly(L-lysine)-grafted-poly(ethylene glycol) copoly-
able antimicrobial drug delivery, J. Biomed. Mater. Res. A 101 (2013) 447–455, mers, Biomaterials 25 (2004) 4135–4148, http://dx.doi.org/10.1016/j.bioma-
http://dx.doi.org/10.1002/jbm.a.34350. terials.2003.11.033.
[139] E.M. Pritchard, T. Valentin, B. Panilaitis, F. Omenetto, D.L. Kaplan, Antibiotic- [166] D. Neut, H. van de Belt, I. Stokroos, J.R. van Horn, H.C. van der Mei, H.J. Busscher,
releasing silk biomaterials for infection prevention and treatment, Adv. Funct. Biomaterial-associated infection of gentamicin-loaded PMMA beads in orthopae-
Mater. 23 (2013) 854–861, http://dx.doi.org/10.1002/adfm.201201636. dic revision surgery, J. Antimicrob. Chemother. 47 (2001) 885–891.
B.D. Brooks, A.E. Brooks / Advanced Drug Delivery Reviews 78 (2014) 14–27 27

[167] A.L.Z. Lee, V.W.L. Ng, W. Wang, J.L. Hedrick, Y.Y. Yang, Block copolymer mixtures as an- [182] C. Jones, D.W. Grainger, In vitro assessments of nanomaterial toxicity, Adv. Drug
timicrobial hydrogels for biofilm eradication, Biomaterials 34 (2013) 10278–10286, Deliv. Rev. 61 (2009) 438–456, http://dx.doi.org/10.1016/j.addr.2009.03.005.
http://dx.doi.org/10.1016/j.biomaterials.2013.09.029. [183] K.K. Assil, J. Frucht-Perry, E. Ziegler, D.J. Schanzlin, T. Schneiderman, R.N. Weinreb,
[168] A.M. Fayaz, K. Balaji, M. Girilal, R. Yadav, P.T. Kalaichelvan, R. Venketesan, Biogenic Tobramycin liposomes. Single subconjunctival therapy of pseudomonal keratitis,
synthesis of silver nanoparticles and their synergistic effect with antibiotics: a Invest. Ophthalmol. Vis. Sci. 32 (1991) 3216–3220.
study against gram-positive and gram-negative bacteria, Nanomedicine 6 (2010) [184] M. Alhariri, A. Azghani, A. Omri, Liposomal antibiotics for the treatment of infec-
103–109, http://dx.doi.org/10.1016/j.nano.2009.04.006. tious diseases, Expert Opin. Drug Deliv. 10 (2013) 1515–1532, http://dx.doi.org/
[169] J.T. Seil, T.J. Webster, Reduced Staphylococcus aureus proliferation and biofilm for- 10.1517/17425247.2013.822860.
mation on zinc oxide nanoparticle PVC composite surfaces, Acta Biomater. 7 [185] Z. Drulis-Kawa, A. Dorotkiewicz-Jach, Liposomes as delivery systems for antibiotics,
(2011) 2579–2584, http://dx.doi.org/10.1016/j.actbio.2011.03.018. Int. J. Pharm. 387 (2010) 187–198, http://dx.doi.org/10.1016/j.ijpharm.2009.11.033.
[170] A. Azam, A.S. Ahmed, M. Oves, M.S. Khan, S.S. Habib, A. Memic, Antimicrobial activ- [186] A. Sharma, Liposomes in drug delivery: progress and limitations, Int. J. Pharm. 154
ity of metal oxide nanoparticles against Gram-positive and Gram-negative bacte- (1997) 123–140, http://dx.doi.org/10.1016/S0378-5173(97)00135-X.
ria: a comparative study, Int. J. Nanomedicine 7 (2012) 6003–6009, http://dx.doi. [187] O.K. Nag, V. Awasthi, Surface engineering of liposomes for stealth behavior,
org/10.2147/IJN.S35347. Pharmaceutics 5 (2013) 542–569, http://dx.doi.org/10.3390/pharmaceutics5040542.
[171] R.Y. Pelgrift, A.J. Friedman, Nanotechnology as a therapeutic tool to combat micro- [188] P.K. Tosh, L.C. McDonald, Infection control in the multidrug-resistant era: tending
bial resistance, Adv. Drug Deliv. Rev. 65 (2013) 1803–1815, http://dx.doi.org/10. the human microbiome, Clin. Infect. Dis. 54 (2012) 707–713, http://dx.doi.org/
1016/j.addr.2013.07.011. 10.1093/cid/cir899.
[172] F. Andrade, D. Rafael, M. Videira, D. Ferreira, A. Sosnik, B. Sarmento, Nanotechnolo- [189] T. Weber, In silico tools for the analysis of antibiotic biosynthetic pathways, Int. J.
gy and pulmonary delivery to overcome resistance in infectious diseases, Adv. Drug Med. Microbiol. (2014), http://dx.doi.org/10.1016/j.ijmm.2014.02.001.
Deliv. Rev. 65 (2013) 1816–1827, http://dx.doi.org/10.1016/j.addr.2013.07.020. [190] R. Laxminarayan, A. Duse, C. Wattal, A.K.M. Zaidi, H.F.L. Wertheim, N. Sumpradit,
[173] L. Zhang, D. Pornpattananangkul, C.-M. Hu, C.-M. Huang, Development of nanopar- et al., Antibiotic resistance—the need for global solutions, Lancet Infect. Dis. 13
ticles for antimicrobial drug delivery, Curr. Med. Chem. 17 (2010) 585–594, http:// (2013) 1057–1098, http://dx.doi.org/10.1016/S1473-3099(13)70318-9.
dx.doi.org/10.2174/092986710790416290. [191] D. Pornpattananangkul, L. Zhang, S. Olson, S. Aryal, M. Obonyo, K. Vecchio, et al.,
[174] F. Andrade, M. Videira, D. Ferreira, B. Sarmento, Micelle-based systems for pulmo- Bacterial toxin-triggered drug release from gold nanoparticle-stabilized liposomes
nary drug delivery and targeting, Drug Deliv. Lett. 1 (2011) 171–185, http://dx.doi. for the treatment of bacterial infection, J. Am. Chem. Soc. 133 (2011) 4132–4139,
org/10.2174/2210304x11101020171. http://dx.doi.org/10.1021/ja111110e.
[175] H. Pinto-Alphandary, A. Andremont, P. Couvreur, Targeted delivery of antibiotics [192] N.J. Irwin, C.P. McCoy, D.S. Jones, S.P. Gorman, Infection-responsive drug delivery from
using liposomes and nanoparticles: research and applications, Int. J. Antimicrob. urinary biomaterials controlled by a novel kinetic and thermodynamic approach,
Agents 13 (2000) 155–168. Pharm. Res. 30 (2013) 857–865, http://dx.doi.org/10.1007/s11095-012-0927-x.
[176] K.I. Wolska, K. Grześ, A. Kurek, Synergy between novel antimicrobials and conven- [193] M.T. Madigan, Brock Biology of Microorganisms, 11th ed. Pearson Prentice Hall,
tional antibiotics or bacteriocins, Pol. J. Microbiol. 61 (2012) 95–104. Upper Saddle River, NJ, 2006.
[177] E. Taylor, T.J. Webster, Reducing infections through nanotechnology and nanoparti- [194] P.M. Shah, The need for new therapeutic agents: what is in the pipeline? Clin.
cles, Int. J. Nanomedicine 6 (2011) 1463–1473, http://dx.doi.org/10.2147/IJN.S22021. Microbiol. Infect. 11 (2005) 36–42, http://dx.doi.org/10.1111/j.1469-0691.2005.
[178] R.T. Tom, V. Suryanarayanan, P.G. Reddy, S. Baskaran, T. Pradeep, Ciprofloxacin- 01141.x.
protected gold nanoparticles, Langmuir 20 (2004) 1909–1914. [195] M.S. Butler, M.A. Blaskovich, M.A. Cooper, Antibiotics in the clinical pipeline in 2013,
[179] H. Gu, P.L. Ho, E. Tong, L. Wang, B. Xu, Presenting vancomycin on nanoparticles to J. Antibiot. (Tokyo) 66 (2013) 571–591, http://dx.doi.org/10.1038/ja.2013.86.
enhance antimicrobial activities, Nano Lett. 3 (2003) 1261–1263, http://dx.doi.org/ [196] L. Freire-Moran, B. Aronsson, C. Manz, I.C. Gyssens, A.D. So, D.L. Monnet, et al., Critical
10.1021/nl034396z. shortage of new antibiotics in development against multidrug-resistant bacteria—
[180] G. Burygin, B. Khlebtsov, A. Shantrokha, L. Dykman, V. Bogatyrev, N. Khlebtsov, On the time to react is now, Drug Resist. Updat. 14 (2011) 118–124, http://dx.doi.org/10.
enhanced antibacterial activity of antibiotics mixed with gold nanoparticles, Nano- 1016/j.drup.2011.02.003.
scale Res. Lett. 4 (2009) 794–801, http://dx.doi.org/10.1007/s11671-009-9316-8. [197] P.M. Shah, The need for new therapeutic agents: what is in the pipeline? Clin.
[181] R. Duncan, R. Gaspar, Nanomedicine(s) under the microscope, Mol. Pharm. 8 Microbiol. Infect. 11 (2005) 36–42, http://dx.doi.org/10.1111/j.1469-0691.2005.
(2011) 2101–2141, http://dx.doi.org/10.1021/mp200394t. 01141.x.

Potrebbero piacerti anche