Sei sulla pagina 1di 11

ADR-12644; No of Pages 11

Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Mechanisms of β-lactam antimicrobial resistance and epidemiology of


major community- and healthcare-associated
multidrug-resistant bacteria☆
Sarah S. Tang a,1, Anucha Apisarnthanarak b,1, Li Yang Hsu c,⁎
a
Singapore General Hospital, Outram Road, Singapore 169608, Singapore
b
Thammasat University Hospital, Pathumthani 12120, Thailand
c
National University Health System, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228, Singapore

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

Article history: Alexander Fleming's discovery of penicillin heralded an age of antibiotic development and healthcare advances
Accepted 11 August 2014 that are premised on the ability to prevent and treat bacterial infections both safely and effectively. The resultant
Available online xxxx evolution of antimicrobial resistant mechanisms and spread of bacteria bearing these genetic determinants of
resistance are acknowledged to be one of the major public health challenges globally, and threatens to unravel
Keywords:
the gains of the past decades. We describe the major mechanisms of resistance to β-lactam antibiotics – the
Antimicrobial resistance
Methicillin-resistant Staphylococcus aureus
most widely used and effective antibiotics currently – in both Gram-positive and Gram-negative bacteria, and
Vancomycin-resistant enterococci also briefly detail the existing and emergent pharmacological strategies to overcome such resistance. The global
Carbapenem resistance epidemiology of the four major types of bacteria that are responsible for the bulk of antimicrobial-resistant infec-
Enterobacteriaceae tions in the healthcare setting – methicillin-resistant Staphylococcus aureus, vancomycin-resistant enterococci,
β-Lactamases Enterobactericeae, and Acinetobacter baumannii – are also briefly described.
Acinetobacter baumannii © 2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. General mechanisms of antimicrobial resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Antimicrobial susceptibility testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. β-Lactam resistance in Gram-positive bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1. S. aureus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.2. Enterococci . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3. Drug development against Gram-positive bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5. β-lactam resistance in Gram-negative bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.1. Enterobacteriaceae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2. A. baumannii . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.3. Drug development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6. Drug delivery mechanisms to overcome antibiotic resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7. Global epidemiology of major antimicrobial-resistant bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7.1. MRSA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7.2. Vancomycin-resistant enterococci (VRE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7.3. ESBL-producing Enterobacteriaceae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7.4. Carbapenem-resistant Enterobacteriaceae (CRE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7.5. Carbapenem-resistant A. baumannii (CRA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

☆ 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: Department of Medicine, National University Health System, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228, Singapore. Tel.: +65 6772
3931; fax: +65 6779 5678.
E-mail addresses: sarah.tang.s.l@sgh.com.sg (S.S. Tang), anapisarn@yahoo.com (A. Apisarnthanarak), liyang_hsu@yahoo.com (L.Y. Hsu).
1
These authors contributed equally to the work.

http://dx.doi.org/10.1016/j.addr.2014.08.003
0169-409X/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
2 S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction and healthcare systems in lower income and less developed countries
more than those in the developed nations as a consequence of issues of
Antibiotics have been hailed as one of the greatest medical advances hygiene and healthcare access [4]. Nonetheless, a post-antibiotic era –
in the modern age [1]. Alexander Fleming's serendipitous observation of if this should ever arise – will profoundly affect all.
a zone of inhibition around Penicillium notatum on a staphylococcal cul- In this review, we focus on the mechanisms of resistance – in partic-
ture plate in 1928 is considered one of the most significant milestones in ular β-lactam resistance – as well as the existing and emergent pharma-
the timeline of antibiotic discovery, although it took more than a decade cological strategies to overcome such resistance. We also briefly
before penicillin was successfully isolated and purified, and underwent describe the global epidemiology of the four major types of bacteria
large-scale production and use [2]. However, Fleming's less celebrated that are responsible for the bulk of antimicrobial-resistant infections
observation was that non-lethal concentrations of penicillin could rap- in the healthcare setting — methicillin-resistant Staphylococcus aureus,
idly generate penicillin-resistant microbes [2], a process of evolution vancomycin-resistant enterococci, Enterobactericeae, and Acinetobacter
that has held true for all antimicrobial agents used in humans to date. baumannii.
As illustrated in Fig. 1, the accelerated growth, diversity and burden
of antimicrobial resistance became increasingly evident past the mid- 2. General mechanisms of antimicrobial resistance
1970s, although this was initially matched by the development of anti-
microbial compounds that could overcome or bypass these resistance Bacteria can acquire antibiotic resistance either through de novo gene
mechanisms. Since the 1990s, however, there has been a “discovery mutation(s) or by horizontal transfer of mobile genetic elements.
void” in the development of new classes of antimicrobial compounds, Mutation-based changes are usually spontaneous and random, involving
a problem compounded by the relentless increase in new and modified single nucleotide polymorphisms that occur at a rate of 10−9 to 10−10 per
mechanisms of antimicrobial resistance in disease-causing microbes gene. Consequently, drug targets may be altered, enzymes capable of
[3–5]. In the β-lactam class of antibiotics – the largest antibiotic inactivating the antibiotic may be expressed, efflux systems may be
class containing arguably the most important and effective antibiotics up-regulated and uptake mechanisms may be lost [7]. In some instances,
developed to date – there has been a ten-fold increase in the types of mutations occur in the regulatory genes or may not result in changes in
inactivating enzymes between 1990 and 2010 [6]. The reasons for the protein function at all. This route of resistance development is often
“discovery void” and current imbalance between antimicrobial resis- deemed to be of lesser importance as cumulative point mutations are usu-
tance and development have been described in detail in several other ally required for phenotypic resistance, although it is possible for the
excellent reviews and will not be further elaborated here [3–5]. strain harboring the mutated allele to become the predominant organism
Multiple attempts have been made to quantify the local and global under selective pressure such as antimicrobial drug use. In contrast, cen-
impact of antimicrobial resistance, and these were extensively covered tral to the rapid and broad propagation of drug resistance is the ability for
in recent reviews and reports [4,5]. To summarize, antimicrobial resis- bacteria to undergo horizontal gene transfer. Antibiotic resistance genes
tant infections result in greater mortality, morbidity and costs of treat- are carried on mobile genetic elements such as transposons, integrons
ment compared to infections caused by their antimicrobial susceptible and plasmids (extrachromosomal genetic material that may be large
counterparts, and the growing inability to prevent infections with anti- and can contain a variety of resistance genes), which serve as vectors
microbial prophylaxis will overwhelmingly and negatively affect the for transfer within the same species or between different species via
risk equation in performing more complex medical procedures such as processes of conjugation, transformation and transduction. Genetic
major surgery, cancer chemotherapy or organ/stem cell transplantation mechanisms of antibiotic resistance and their consequences have been
[4,5]. This burden is unevenly distributed at present, affecting patients previously reviewed [8–11].

Fig. 1. Timeline of antimicrobial drug discovery against the development of antimicrobial drug resistance.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 3

Basic mechanisms of antibiotic resistance can generally be summa- 4. β-Lactam resistance in Gram-positive bacteria
rized using the bullet–target concept [12]. Sites of drug activity (target)
may be altered by enzymatic modification (e.g. methylation of 16S rRNA β-Lactam antibiotics exert bactericidal activity by covalently binding
in aminoglycoside resistance), transformed by genomic mutations (e.g. to and inactivating enzymes known as penicillin-binding proteins
DNA gyrase in quinolone resistance), and/or bypassed metabolically (PBPs), resulting in interference with the synthesis and remodeling of
(e.g. sulfonamide resistance). On the other hand, the antibiotic (bullet) the bacterial peptidoglycan. In Gram-positive bacteria, β-lactam resis-
can undergo enzymatic inactivation (e.g. acetylation of aminoglyco- tance primarily occurs as a result of alteration of PBPs, with enzymatic
sides) and degradation (e.g. β-lactams by β-lactamases), reduced acces- degradation as a minor pathway (Fig. 2).
sibility in entering the bacteria cell (e.g. porin loss) and/or increased
extrusion from the cell (e.g. efflux pump) [13,14]. Fig. 2 depicts the com- 4.1. S. aureus
mon mechanisms of β-lactam antibiotic resistance by both Gram-positive
and Gram-negative bacteria. Penicillin was the primary antibiotic used for S. aureus infections
until the late 1950s when penicillinase-producing S. aureus became a
3. Antimicrobial susceptibility testing major problem, particularly during the Asian influenza pandemic
when such bacteria frequently caused post-influenza pneumonia [19].
Antimicrobial resistance detection in bacteria is generally performed The β-lactamase produced is encoded by the blaZ gene, which in turn
in clinical and research microbiology laboratories using phenotypic is regulated by two adjacent genes, the repressor blaI and anti-
susceptibility testing methods. These are variants of detection of micro- repressor blaR1 [20]. In the presence of β-lactam antibiotics, blaR1 pro-
bial growth from an initial specified amount and/or concentration of tein undergoes autolytic cleavage to become a protease for the cleavage
bacteria inoculated into culture media with stepwise dilution of target of the blaI protein, thereby unblocking the transcription of the blaZ gene
antimicrobial concentrations. The most commonly used methods – [21].
disk susceptibility testing, broth dilution, Etest strips, and automated sys- Methicillin, first-generation cephalosporins and subsequently the
tems – have been reviewed elsewhere [15]. Such methods produce ei- other semi-synthetic penicillinase-resistant penicillins (cloxacillin,
ther qualitative (disk susceptibility testing) or quantitative results, and flucloxacillin) were introduced in the late 1950s–early 1960s. Unfortu-
both the testing as well as the interpretation of test results – especially nately, this strategy was short-lived as methicillin-resistant S. aureus
in the clinical setting – are usually based on regularly updated standards (MRSA) appeared at the start of the 1960s [22]. Methicillin resistance
published by expert bodies such as the European Committee on Antimi- is not mediated by β-lactamase production but by the expression of a
crobial Susceptibility Testing (EUCAST) [16] or the Clinical Laboratory unique PBP (PBP 2a), which is in addition to the four PBPs (PBP1–4)
Standards Institute (CLSI) [17]. present in native S. aureus. PBP2a has low affinity for all β-lactam anti-
Phenotypic susceptibility testing methods are generally not able to biotics and acts as a substitute for the other PBPs, thus enabling the sur-
define the exact mechanism of resistance, although an accurate guess vival of the bacteria in the presence of high concentrations of β-lactam
can usually be made based on a combination of the distribution of test antibiotics [23]. The gene encoding PBP2a is known as mecA and is locat-
results as well as the species of microorganism tested [15]. The increas- ed in a specific mobile genetic element referred to as the staphylococcal
ing availability and reduced costs of next generation sequencing renders cassette chromosome mec (SCCmec) that integrates into the chromo-
whole genome sequencing an attractive possibility for bacterial resistance some using recombinases (ccrAB or ccrC) carried on the SCCmec
detection — and the results have been excellent in certain organisms such element itself [24]. Similar to the blaZ system, PBP2a synthesis is regu-
as S. aureus [18]. Nonetheless, for bacteria where novel resistance mecha- lated by mecR1 and the repressor gene mecI; in addition, due to high
nisms emerge frequently, whole genome sequencing and other molecular sequence homology with the proteins, expression of PBP2a can be in-
methods may not be able to detect significant antibiotic resistance as re- duced by the blaR1–blaI–blaZ system, if present. Constitutive expression
liably as phenotypic methods [15]. of methicillin resistance, on the other hand, requires activation of the

Fig. 2. Major mechanisms of β-lactam resistance in both Gram-positive and Gram-negative bacteria.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
4 S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

mecA gene by the mutation or deletion of the mecI gene or a mecA disruption of the cell wall (e.g. pore-forming drug, brilacidin), and (vii) in-
promotor/operator region [24]. hibition of fatty acid synthesis (e.g. FabI inhibitor, CG400549) [37,38].
Expression of methicillin resistance, however, also involves several In our opinion, β-lactams that can directly bind to altered PBPs (such
other factors. PBP2a, lacking a functional glycosyltransferase region, as ceftobiprole against PBP2a) are more likely to be effective in the clin-
requires the co-existence of PBP2 to effectively make peptidoglycan ical setting and have a higher likelihood of impacting clinical practice in
[25]; hence deletion of PBP2 will reduce the ability of PBP2a to confer the short and medium term. Other strategies as described above are
resistance. The fem (factor essential for methicillin resistance) or aux interesting but face significant hurdles in reaching clinical practice, in-
(auxiliary) factors, including llm, fmtA and sigB, have also been identified cluding tests of toxicity in animals and humans, pharmacokinetics,
to alter the expression of resistance. These genes are located on chromo- costs of clinical trials to demonstrate efficacy, etc.
somal DNA separate from the mec element and are also involved in
peptidoglycan synthesis [26,27].
5. β-lactam resistance in Gram-negative bacteria

4.2. Enterococci β-Lactam resistance in Gram-negative bacteria can arise through


three possible mechanisms: alteration of PBPs, production of β-
The intrinsic resistance of enterococci to β-lactam antibiotics stems lactamases, and limited access to target PBPs (Fig. 2). In contrast to
from the production of low-affinity PBP5 that is able to function in Gram-positive bacteria, PBPs are located in the periplasmic space of
place of other PBPs that are already saturated by β-lactam antibiotics. Gram-negative bacteria and β-lactam antibiotics must traverse across
Minimum inhibitory concentrations (MICs) of ampicillin, penicillin, the bacterial outer membrane in order to reach their target sites of ac-
piperacillin and imipenem have been reported to be approximately tion. Hence, any modulation that restricts entry (porin loss) or result
100-fold higher than that for streptococci [28], with MICs of penicillin in extrusion (efflux pumps) of β-lactam antibiotics will confer antibiotic
against Enterococcus faecalis and Enterococcus faecium typically in the resistance, with the potential for cross-resistance to multiple agents
ranges of 2–8 μg/ml and 8–16 μg/ml respectively [29]. Deletion of should these drugs share the same channel. This mechanism of resis-
PBP5 is associated with an 800-fold reduction in MIC of ampicillin tance is unique to Gram-negative bacteria. In addition, while changes
[30]. Accordingly, this low-level resistance renders penicillins bacterio- in PBPs are the foundational basis for antibiotic resistance in Gram-
static and cephalosporins practically ineffective. positive bacteria, they occur less frequently in Gram-negative bacteria,
Molecular mechanisms for intrinsic cephalosporin resistance have in which production of β-lactamases is the most prevalent resistance
yet to be clearly defined. Apart from inherent differences in binding af- mechanism [39]. Of note, the synergy achieved through the interplay
finity to PBP5, a two-compartment signal transduction system (croRS) of these diverse mechanisms plays an important role in determining
and a Ser/Thr kinase, known as ireK (as for intrinsic resistance of entero- the final phenotypic expression of resistance in Gram-negative bacteria.
cocci) and which is reciprocally regulated by the upstream protein
phosphatase ireP, have been reported to be important factors of cepha-
5.1. Enterobacteriaceae
losporin resistance [31,32]. IreB, which is found downstream of ireK, has
also recently been described to contribute to cephalosporin resistance
The profound impact of β-lactamases on antimicrobial resistance is
through ireK-dependent phosphorylation [33].
most evident in this group of bacteria. Interestingly, the first bacterial
High level resistance to β-lactams develops through acquisition of β-
enzyme reported to destroy penicillin was an AmpC cephalosporinase
lactamases or mutations in PBP5. The β-lactamases in enterococci are
identified from Escherichia coli in 1940, several years before the
encoded by plasmid-mediated bla genes, which are highly homologous
introduction of penicillin into clinical practice [40]. Several schemes of
to the blaZ genes of S. aureus, except for its constitutive expression
classification have been proposed to characterize the vast number of
presumably due to a lack of a repressor protein [28]. Resistance due to
β-lactamases identified. A commonly used system is the Ambler molec-
this mechanism is nonetheless rare and has been described predomi-
ular classification scheme whereby enzymes are grouped based on pro-
nantly in E. faecalis rather than E. faecium isolates [29]. Resistance in
tein homology — the need for serine in its active site (class A, C and D
E. faecium has been attributed to modulations in PBP5 — mutations in
serine-β-lactamases) and those that require zinc (class B metallo-β-
its penicillin binding site that causes further reduction in binding affin-
lactamases) [41]. Another commonly used scheme is the Bush–Jacoby–
ity to β-lactams and/or its overproduction. In particular, the former has
Medeiros functional classification system that categorizes the β-
been implicated in the development of highly resistant strains that have
lactamases into four main groups according to their substrate and inhib-
MICs of ampicillin up to 512 μg/ml [34].
itory properties. This system, which was originally proposed in 1995, was
updated and expanded by Bush and Jacoby in 2010 [42]. A list of β-
4.3. Drug development against Gram-positive bacteria lactamases in both classification systems is shown in Table 1.
Characteristics of the various β-lactamases have been extensively
Drug discovery and development against Gram-positive bacteria is reviewed in previous publications [42–47], and will only be briefly
primarily directed at the cell wall, in particular, the PBPs. As bacteria touched upon here. Extended-spectrum β-lactamases (ESBLs) are prod-
evolve in terms of their PBP expression, new compounds that exhibit ucts of point mutations in class A β-lactamases TEM-1, TEM-2 and SHV-
greater binding affinity to current PBPs and/or are capable of inhibiting 1 that lead to broadening of their substrate specificity. These enzymes
new PBPs have been designed. For instance, the newly approved ceph- confer resistance to penicillins, first-, second-, and third-generation
alosporins (ceftobiprole and ceftaroline) confer anti-MRSA activity by cephalosporins as well as aztreonam, but are inhibited by β-lactamase
their ability to bind to and inhibit PBP2a, whereas several noncovalent inhibitors and are not active against the cephamycins (e.g. cefoxitin)
compounds appear to be promising PBP2a inhibitors capable of restor- [48]. Cefepime resistance may be more frequent with the CTX-M-type
ing β-lactam antibiotic susceptibility to MRSA [35,36]. ESBL and its susceptibility appears to be dependent on the inoculum
Other strategies to modulate methicillin resistance have also been of the pathogen [46]. The CTX-M ESBLs, which are related to the chro-
explored and include (i) inhibition of mRNA expression of mecA (e.g. mosomal β-lactamases of the environmental Kluyvera species, emerged
polyoxometalates), (ii) use of DNAzymes to disrupt the bla and mec in the late 1980s and have since become very prevalent [49]. CTX-M en-
signal pathways (e.g. PS-DRZ1366 and PS-DRZ1694), (iii) inhibition of zymes typically hydrolyze cefotaxime more readily than ceftazidime,
Mur enzymes (e.g. fosfomycin), (iv) interference with cell wall precursor and unlike the TEM and SHV ESBLs, are more susceptible to inhibition
lipid I and lipid II (e.g. defensin mimetic BAS00127538), (v) blocking bio- by tazobactam than clavulanic acid [44,46,49]. OXA-type ESBLs are rare-
synthesis of wall teichoic acids (e.g. 1835F03 and targocil), (vi) direct ly found in Enterobacteriaceae.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 5

Table 1
Classification of bacterial β-lactamases (illustration of both the Ambler [33] and Bush–Jacoby classifications [34]).

Ambler Bush– Characteristics Examples of enzymes Substrates Specific laboratory confirmation


Jacoby method(s)

A 2a Penicillinases inhibited by CA1 PC1 Penicillins


2b Broad-spectrum enzymes inhibited TEM-1, TEM-2, TEM-13, SHV-1, Penicillins, cephalothin
by CA1 SHV-11
2be Extended broad-spectrum enzymes TEM-3, TEM-10, TEM-26, SHV-2, Penicillins, oxyimino-cephalosporins Cephalosporin/CA synergy double-
inhibited by CA1 SHV-3, Klebsiella oxytoca K1, CTX- (cefotaxime, ceftazidime, ceftriaxone, disk test, ESBL Etest
M, PER, VEB cefepime), monobactams
2br Broad-spectrum enzymes with TEM-30, TEM-31, SHV-10, SHV-72 Penicillins, resistant to CA, tazobactam and
reduced binding to CA1 (inhibitor- sulbactam
resistant TEMs)
2ber Extended-spectrum enzymes with TEM-50, TEM-158 Penicillins, oxyimino-cephalosporins,
relative resistance to CA1 monobactams, resistant to CA, tazobactam
and sulbactam
2c Carbenicillin-hydrolyzing enzymes PSE-1, CARB-3 Penicillins, carbenicillin
inhibited by CA1
2ce Extended-spectrum carbenicillinase RTG-4 (CARB-10) Carbenicillin, cefepime
2e Cephalosporinases inhibited by CA1 CepA Cephalosporins
2f Carbapenem-hydrolyzing KPC, SME, GES, IMI-1 All β-lactams, including carbapenems Modified Hodge test, Carba NP test
nonmetallo-β-lactamases
B 3 Metallo-β-lactamases IMP, VIM, IND All β-lactams, including carbapenems, with Modified Hodge test, Carba NP test
exception of monobactams
C 1 Cephalosporinases not inhibited by ACT-1, FOX-1, MIR-1, CMY Narrow and extended-spectrum cephalo- Cefoxitin/cloxacillin synergy
CA1 sporins, including cephamycins double-disk test, AmpC Etest,
modified Hodge test
D 2d Cloxacillin-hydrolyzing enzymes OXA-1, OXA-2, OXA-10 Cloxacillin, oxacillin
2de with variable inhibition by CA1 OXA-11, OXA-15 Cloxacillin, oxacillin, oxyimino-
cephalosporins, monobactams
2df OXA-23, OXA-51, OXA-58 Cloxacillin, oxacillin, carbapenems Modified Hodge test, Carba NP test
– 4 Penicillinases not inhibited by CA1 Penicillinases from Burkholderia Penicillins
cepacia
1
CA — clavulanic acid.

AmpC β-lactamases are characteristically resistant to inhibition by β- activity [53]. Nonetheless, high level resistance to carbapenems can
lactamase inhibitors and provide resistance to oxyimino-cephalosporins arise from synergy with loss of outer membrane proteins (OMPs). Carba-
along with the cephamycins; fourth-generation cephalosporins (e.g. penem resistance can also occur in porin-deficient AmpC β-lactamase or
cefepime) are considerably more stable [50]. Most Enterobacteriaceae, ESBL-producing isolates, although this mechanism is considered of lesser
apart from Klebsiella spp. and Proteus spp., have chromosomal AmpC clinical and public health importance because of the reduced risk of trans-
genes that are constitutively expressed at low levels. Expression of mission of resistance compared to plasmid-borne β-lactamase genes.
the blaAmpC gene, however, is inducible by derepression on exposure to
β-lactam antibiotics such as amoxicillin, imipenem and clavulanic acid. 5.2. A. baumannii
In E. coli, high levels of resistance to cephalosporins can also occur either
through up-regulation of the chromosomal AmpC gene or through the ac- A. baumannii has inherent chromosomally-encoded AmpC-type
quisition of a plasmid-borne AmpC gene that is unaccompanied by regu- cephalosporinases, now referred to as ADCs (Acinetobacter derived
latory genes [47]. cephalosporinases), that has not been shown to be inducible. Rather, it
Once thought to be rare, carbapenem-resistant Enterobacteriaceae is constitutively expressed but at low levels inadequate to affect its an-
(CRE) have been labeled an urgent (highest level) health threat as of tibiotic susceptibility profile. Nonetheless, resistance to penicillins and
2013 [51], with few effective antimicrobial agents against infections cephalosporins, with the exception of cefepime, can develop upon up-
caused by these organisms other than the polymyxins and tigecycline stream acquisition of insertion sequence ISAba1, and less frequently
(because such organisms typically are also resistant to the majority of ISAba125, which serve as strong promoters for the overproduction of
non-β-lactam antibiotics through other mechanisms of resistance). the enzyme [54,55]. Extended-spectrum AmpC-type cephalosporinases
Two groups of enzymes have been associated with carbapenem resis- (ESAC), such as ADC-33 and ADC-56, have been described as being ca-
tance — the metallo-β-lactamases and non-metallo-β-lactamases. The pable of hydrolyzing all cephalosporins including cefepime [56,57].
metallo-β-lactamases have a comparatively broader spectrum of activ- A. baumannii also produces an intrinsic chromosomally-encoded
ity as they exhibit strong hydrolytic activity against all β-lactam antibi- oxacillinase (OXA-51 and its variants). In like manner to the ADCs,
otics except for monobactams (i.e. aztreonam), and are not inhibited by genes encoding for these oxacillinases are normally expressed at low
β-lactamase inhibitors. The group of non-metallo-β-lactamases include levels but can be up-regulated and thereby confer carbapenem resis-
the class A carbapenemases and class D OXA-type carbapenemases. tance when ISAba1 or ISAba9 is inserted upstream [58].
Class A carbapenemases of the SME and IMI families can confer resistance Acquired β-lactamases include penicillinases (TEM-1, TEM-2 and
to penicillins and aztreonam but not the oxyimino-cephalosporins; the CARB-5), class D oxacillinases that lack carbapenemase activity but
Klebsiella pneumoniae carbapenemase (KPC) carbapenemases confer re- are clavulanic acid-resistant (OXA-20, OXA-21 and OXA-37), and the
sistance to all β-lactam antibiotics and are plasmid-encoded, and are extended-spectrum β-lactamases (ESBLs). Although the penicillinases
therefore potentially able to spread between Enterobacteriaceae with are generally regarded to be susceptible to inhibition by common
greater facility [50]. The OXA-type carbapenemases confer high level re- β-lactamase inhibitors, production of TEM-1 has been associated with
sistance to penicillins, have some hydrolytic activity against carbapenems sulbactam resistance [59]. Genes reported to encode for ESBL production
(imipenem more so than meropenem), but generally do not affect the include blaPER-1, blaPER-2, blaPER-7, blaVEB-1, blaCTX-M, blaSHV-5, blaSHV-12,
cephalosporins and monobactams [52]. In contrast to the metallo-β- blaTEM-92 and blaTEM-116 [60]. In addition, RTG-4 (CARB-10), which is a
lactams, the non-metallo-β-lactamases have fairly weak hydrolytic unique plasmid-encoded CARB-type β-lactamase that hydrolyzes

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
6 S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

cefepime more significantly than ceftazidime and cefotaxime, has been


identified [61].
The major problem with widespread development of carbapenem-
resistant A. baumannii and outbreaks arises from its ability to acquire
carbapenem-hydrolyzing β-lactamases, specifically the Ambler class B
metallo-β-lactamases and class D oxacillinases. Metallo-β-lactamases
are notably the most potent of the carbapenemases, although it is less
common than the OXA-type [62]. Four groups of metallo-β-lactamases
have been described thus far — imipenemase (IMP)-type, Verona
integrin-encoded metallo-β-lactamase (VIM)-type, SIM-1 and New
Delhi metallo-β-lactamase (NDM-1 and NDM-2). Apart from aztreo- Fig. 3. Mechanism of action of β-lactamase-inhibitor combinations, where the β-lactam
nam, these enzymes have the ability to hydrolyze all β-lactams. The antibiotic particles (green polygon) are able to bind with their target penicillin-binding
OXA carbapenemases have been classified into four clusters: (i) OXA- proteins (blue blocks) because the β-lactamase inhibitors (red polygons) bind to and in-
23-like (OXA-23, OXA-27 and OXA-49); (ii) OXA-24-like (OXA-24, hibit bacterial β-lactamases (purple blocks). (For interpretation of the references to
color in this figure legend, the reader is referred to the web version of this article.)
OXA-25, OXA-26, OXA-40 and OXA-72); (iii) OXA-58 and (iv) OXA-
143-like (OXA-143 and OXA-231) [63–65]. OXA-23, which can be either
intra-abdominal infections [73,74], and appears to have an important ad-
chromosome or plasmid mediated, was the first OXA-type β-lactamase
vantage over clavulanic acid — it does not induce AmpC β-lactamase pro-
to be identified from A. baumannii and remains the most prevalent
duction [75].
today. More recently, a novel subclass designated OXA-235-like (OXA-
Another class of agents that displays significant potential is boronic
235, OXA-236 and OXA-237) has been identified from isolates recov-
acid β-lactamase inhibitors (e.g. RPX7009), which exhibit very potent
ered from the U.S. states and Mexico [65]. Carbapenemases belonging
AmpC β-lactamase inhibitory activity when the carboxamide group of
to Ambler class A are also increasingly detected in A. baumannii and
glycylboronates are replaced with a sulfonamide. 4,7-Dichloro-1-
they include KPC-2, KPC-3, KPC-4, KPC-10, and GES-14 [60].
benzothien-2-yl sulfonylaminomethyl (DSABA) is a boronic acid-
A. baumannii is intrinsically resistant to several antimicrobial agents,
based compound that also exhibits activity against OXA β-lactamases
and this is believed to be contributed in part by the small number and
[76].
size of OMPs present, thereby making it less permeable, and/or the
Although the above compounds appear promising, they do not in-
low level constitutive expression of active efflux pumps [66]. To date,
hibit the emerging group of metallo-β-lactamases and there is an urgent
three OMPs have been associated with carbapenem resistance — a
need to develop effective inhibitors against these enzymes in view of
29 kDa protein (also known as CarO), a 33–36 kDa protein and
the changing epidemiology of bacterial resistance. Apart from the thiol
OmpW, which has yet to be fully characterized [60]. An additional
derivatives being the prime candidate, including use of the clinically
43 kDa protein, also referred to as OprD homologue, had been previous-
available anti-hypertensive agent captopril, work has also been per-
ly reported to play a role in carbapenem resistance. However, this claim
formed with classes such as pyridine dicarboxylates, maleic acid deriv-
has since been challenged by newer evidence, which revealed its lack of
atives, carbapenem analogs, and the tricyclic natural products, but none
involvement in carbapenem diffusion, and its closer similarity on genet-
are close to emerging from the developmental pipeline [70,77,78].
ic basis and function to the OprQ protein of Pseudomonas aeruginosa
Just as inhibitor-resistant strains have surfaced with current β-
[67]. The AdeABC efflux pump belongs to the resistance-nodulation-
lactamase inhibitors, it is highly probable that resistance will continue
division (RND) family of efflux systems and has been identified to be
to develop against these novel “second generation” β-lactamase inhibi-
the main efflux mechanism responsible for β-lactam resistance in
tors when they enter clinical use. It is thus necessary to consider addi-
A. baumannii. Upregulation of the gene encoding for this efflux pump
tional strategies to overcome the diverse mechanisms of bacterial
can confer resistance to a broad range of antimicrobial agents, namely
resistance that have been described above so that a multi-pronged ap-
aminoglycosides, β-lactams, fluoroquinolones, tetracyclines, tigecyc-
proach may be adopted. Efflux pump inhibitors are an attractive option
line, macrolides, chloramphenicol, trimethoprim and tigecycline [68].
as adjunctive therapy but their development process has been slow.
Among the class of β-lactam antibotics, cefepime, cefpirome and cefo-
Pagès and Bhardwarj provided a detailed analysis on efflux pump inhib-
taxime are most affected, while it has little impact on the remaining
itors, their ideal characteristics and the list of patented compounds thus
members, including carbapenems. Nonetheless, when present in syner-
far [79,80]. A schematic of the effect of efflux pumps as well as the var-
gy with production of oxacillinases, AdeABC can contribute to high level
ious types of inhibitors is shown in Fig. 4. It has been observed that
carbapenem resistance. AdeIJK is another RND efflux system present in
charge and molecular weight are important determinants of suscepti-
A. baumannii that is capable of extruding β-lactams; AdeFG is the third
bility to extrusion by efflux pump and may be useful parameters for
RND efflux system present, but it has no effect on β-lactam antibiotics
modifying existing compounds.
[69].

5.3. Drug development 6. Drug delivery mechanisms to overcome antibiotic resistance

With new bacterial β-lactamases against which currently available It is well recognized that the design of new compounds is an expen-
β-lactamase inhibitors (clavulanic acid, tazobactam, sulbactam) are no sive and time-consuming process [81]. There is thus value in exploring
longer active, recent efforts have been directed towards the develop- improvements in drug delivery mechanisms as an alternative strategy
ment of novel β-lactamase inhibitors and these have been reviewed in to cope with drug-resistant bacteria. The use of nanotechnology for
detail elsewhere [70–72]. Fig. 3 shows the effect of β-lactamase inhibi- drug formulation has been utilized largely in the field of oncology and
tors in binding β-lactamases while the active drug targets the PBPs. there are many theoretical benefits to also support its use in the treat-
Among the β-lactamase inhibitors in the pipeline, avibactam and MK- ment of infectious diseases [82].
7655, which belong to the class of non-β-lactam-β-lactamase inhibitors β-Lactam antibiotics are readily water-soluble, and thus they general-
(diazabicyclooctanes), are at the forefront of the developmental ly have poor oral bioavailability (most antibiotics have to be given paren-
process. They are active against serine class A, class C and some class terally) and depend on porins for entry into the bacteria. Nanoparticles
D β-lactamases including ESBLs, AmpC, KPCs and OXA-48. Avibactam are amphiphilic molecules that can be capitalized to increase the solubil-
has been successful in clinical trials when paired with ceftazidime ity of β-lactam antibiotics, which in turn enhances the ease of oral
and ceftaroline for the treatment of complicated urinary tract and drug absorption [82]. The most widely used nanocarrier system are the

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 7

Fig. 4. Schematic of bacterial efflux pump showing (i) extrusion of antibiotic in drug-resistant bacteria, (ii) efflux pump inhibitor plug blocking the outer membrane channel of the pump,
(iii) non-antibiotic molecule as either a competitive or non-competitive inhibitor of the efflux pump affinity binding site, and (iv) modification of the antibiotic molecule to avoid extrusion
by the efflux pump.

liposomes; its lipid bilayer rapidly fuses with the bacterial cell wall, there- use of antimicrobial agents in both humans and livestock, and better
by overcoming the problem of reduced drug uptake, and large quantities transportation links [3,5]. Although healthcare facilities have traditionally
of antibiotic are released directly into the periplasmic space [82,83]. This been associated with the most resistant bacterial pathogens, the rise of
high antibiotic concentration can potentially overwhelm β-lactamases community- and livestock-associated MRSA and ESBL-producing Entero-
and efflux pumps, rendering them “ineffective” (see Fig. 5). Liposomes bacteriaceae demonstrate that such bacteria are not limited to healthcare
can also be enhanced with “stealth” material (e.g. polyethylene glycol) settings, and pose a potentially escalating threat even in the community
that prolongs its circulatory lifetime and/or attached with cell-specific li- setting [5–7,10].
gands (e.g. antibodies, peptides) that allow target binding and release of
high antibiotic concentrations at its intended site [83]. As have already 7.1. MRSA
been seen with liposomal amphotericin B, such tissue specificity could
translate as a toxicity-sparing approach by minimizing unnecessary Since its emergence in UK in the 1960s [22], successive epidemic
drug exposure and injury to other organ tissues. strains of healthcare-associated MRSA (HA-MRSA) have spread world-
A slight variation to the liposomes is the polymeric nanoparticle, wide and become entrenched in the hospitals of most countries, with
which has been evaluated and used for controlled drug release [84]. few exceptions such as West Australia, Holland, and the Scandinavian
This feature is particularly valuable for β-lactam antibiotics, whereby countries [86]. The first cases of true community-associated MRSA [87,
the percentage time (of up to 70%) for which the antibiotic concentra- 88] passed virtually unnoticed by the majority of the medical communi-
tion is kept above the minimum inhibitory concentration is the key de- ty, until the deaths of four children from severe CA-MRSA infection were
terminant of bactericidal activity and optimizing on this parameter reported from North Dakota and Minnesota in 1997 [89].
could render the antibiotic effective against bacteria that would have Currently, a persistently high number of CA-MRSA infections are
otherwise been deemed resistant according to conventional antibiotic being observed, in particular in the US, but also in increasing frequency
dosing regimens. Nonetheless, there are far less data even in the labora- in other parts of the world, reaching pandemic proportions [90,91]. The
tory setting with regard to the efficacy of polymeric nanoparticles as an- CA-MRSA epidemic is particularly severe in the US possibly due to the
tibiotic carriers [85]. higher pathogenic potential of the US epidemic CA-MRSA strain
In summary, novel drug delivery methods represent an evolutionary USA300, which is now also spreading to other countries [90]. At present,
(but not quite revolutionary) advance on the treatment of infectious an increasing number of reports from the US and abroad indicates that
diseases, and may be used to overcome many of the current bacterial CA-MRSA strains may be gradually replacing HA-MRSA strains in hospi-
antimicrobial resistance mechanisms. Development in this field has tals. Several authors have suggested that this indicates yet another po-
remained slow, however, and there are as yet no clinical trials to deter- tential epidemiological shift in staphylococcal infections [92–94].
mine the efficacy of such methods in the treatment of human infections. Reports of livestock-associated MRSA (LA-MRSA) in European and
Asian animals have increased since 2000 [95,96]. Routine use of antimi-
7. Global epidemiology of major antimicrobial-resistant bacteria crobials as growth promoters in animal husbandry appears to be a
possible cause of emerging MRSA carriage [95]. Individuals including
In the past 3 decades, there has been an acceleration in the diversity veterinarians and farmers who have been exposed to LA-MRSA colo-
and number of antimicrobial resistant bacteria, coinciding with increased nized livestock are at higher risk for LA-MRSA carriage and infection

Fig. 5. Use of nanocarriers to overcome β-lactamase antibiotic resistance by enhancing drug transport across the cell membrane and saturating β-lactamases produced by the bacteria.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
8 S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

[95,97,98]. Additionally, LA-MRSA was found in 2.5–11.9% of livestock- the three countries with the highest reported incidences of CRE were
derived food products such as pork, turkey, lamb, beef and chicken in Greece (20–50%), India (4–24%), and the United States (US) (4–11%)
Europe and North America, although infections caused by LA-MRSA [125,127,129,130]. There is also a keen global interest in infections
from food products has not been reported to date [99–101]. due to KPC and NDM-1-producing Enterobacteriaceae. KPC-producing
Enterobacteriaceae were first reported in the US in 1996, with sub-
7.2. Vancomycin-resistant enterococci (VRE) sequent clonal dissemination through the US and to North Africa,
Europe, China, Canada, Brazil, Argentina, Columbia, and Venezuela
High-level resistance to the glycopeptide class of antibiotics, [124,131]. Nonetheless, underreporting of KPC-producing Entero-
vancomycin and teicoplanin, was first reported in Europe in 1986 bacteriaceae likely exists, given the lack of a structured global sur-
[102] and in the US in 1987 [103]. Since then, VRE have been isolated veillance program and difficulties in laboratory detection.
in multiple countries on several continents and its prevalence is on In contrast to the clonal spread of KPC-producing Enterobacteriace-
the rise [104]. Between 1989 and 1993, the rate of vancomycin resis- ae, cases with NDM-1 producing bacteria have been detected world-
tance reported in the USA by the National Nosocomial Infections Sur- wide since the first case was discovered in Sweden in 2008 [132]. The
veillance (NNIS) system increased from 0.3% to 7.9% [105]. The NNIS first recognized endemic region for NDM-1 producing bacteria was
system in 2004 showed that 28.5% of enterococcal isolates were resis- the subcontinent of India, inclusive of Pakistan [124]. Cases of coloniza-
tant to vancomycin — an increase of 12% compared with data averaged tion or infection with NDM-producing Enterobacteriaceae and non-
for the years between 1998 and 2002 [106]. In the USA, VRE is found Enterobacteriaceae have now been reported from the UK, the US,
mainly in patients exposed to healthcare settings. Studies conducted Canada, Kenya, China, Australia, Western Europe, and the Middle East
in the USA in the 1990s failed to detect VRE among farm animals [107, [124,125,133]. Clinical isolates with NDM-1 producing pathogens have
108]. The situation is different in Europe where a glycopeptide called been reported from the respiratory tract, urinary tract, skin and skin
avoparcin was used as a growth promoter in the animal industry until structures, surgical sites, and the bloodstream [124,125,134]. Risk
1997 [109]. That led to a high rate of VRE colonization in animals and, factors for the acquisition of NDM-1 producing pathogens include
subsequently, in healthy humans via the food chain, either by direct healthcare-associated risks, device-associated risks, severity of illness,
contact or by eating contaminated products [110]. Fortunately, the and use of several antibiotics, especially carbapenems, third- and
prevalence of VRE in farm animals decreased significantly after fourth-generation cephalosporins, and fluoroquinolones [123]. In addi-
avoparcin was withdrawn [111,112]. In South East Asia, the situation tion, the acquisition of NDM-1 producing Enterobacteriaceae can be
may be similar since avoparcin has been used in the animal industry food-borne, and repatriation, patient transfers from endemic areas,
in several Asian countries [113]. and medical tourism have been reported in case reports, case series,
and surveillance studies of these pathogens [123,135,136].
7.3. ESBL-producing Enterobacteriaceae
7.5. Carbapenem-resistant A. baumannii (CRA)
The first definitively characterized ESBL, TEM-3 (cefotaxime-
hydrolyzing enzyme type 1), was discovered in K. pneumoniae iso- Over the last 15 years, CRA has emerged globally as one of the most
lates recovered from intensive care unit patients in France [114]. challenging pathogens to treat when identified as a healthcare-
Since that initial report, TEM-type enzymes have become the most associated infection. Before the year 2005, regional variation in CRA
diverse class of ESBLs, with N 100 genetic variants now reported existed, with a range in incidence of 5.6% to 34.5% [137–142]. As report-
[115]. The prevalence of ESBL-producing Enterobacteriaceae can ed from reviews dating back to the 1970s [142], healthcare-associated
vary greatly from one location to another and even over time, pneumonia is still the most common infection caused by this organism.
given that ESBL-producing Enterobacteriaceae often arise in focal However, in more recent times, infections involving the central nervous
outbreaks [116]. As a result, regional and local estimates are proba- system, skin and soft tissue, and bone have emerged as highly problem-
bly more useful to clinical decision-making as compared to global atic for certain institutions. More recently, countries such as Australia,
assessments [116]. A further caveat when reviewing prevalence Brazil, Singapore, Canada, South Korea, Taiwan, and Thailand have re-
data is that different criteria are used to determine whether an En- ported CRA incidence rates of 47–80%, with consequent therapeutic
terobacteriaceae produces ESBL. Moreover, Enterobacteriaceae that and infection control challenges [123,137,142–144].
test positive for ESBL at screening might possess a resistance mech- Patients with CRA present with a panoply of healthcare-associated
anism other than ESBL production (e.g., AmpC-type β-lactamase or infections such as pneumonia, urinary tract infection, skin and skin
permeability effects that result from changes in OMPs) [116]. structure infections, and bloodstream infection [123,145]. Patients
The SENTRY Antimicrobial Surveillance Program estimated that the with prolonged hospital stay, severe illness, and catheter- or intravascu-
prevalence of ESBL-producing strains of E. coli and K. pneumoniae in lar device placement have increased risk for CRA acquisition [123,146,
the United States is relatively low (prevalence of 6–7%) [117–120]. In 147]. Additionally, colonization with CRA, exposure to other patients
contrast, the prevalence of ESBL-producing K. pneumoniae is high in in the same geographic unit with CRA, and exposure to group 2
the Asia-Pacific region (prevalence of 10–25%) and in Latin America. carbapenems (imipenem-cilastatin, meropenem, and doripenem), and
Data from Latin America suggest that the presence of ESBL-producing aminopenicillins are identified risks for CRA infection [145,148,149]. In-
strains of K. pneumoniae has increased dramatically in recent years terestingly, the emergence and international spread of CRA is limited to
(prevalence of 5–46%) [117–120]. In recent years, the spread of a small number of epidemic clones, particularly global clones I and II
extended-spectrum β-lactamases (ESBLs), particularly CTX-M enzymes, [150,151].
in Enterobacteriaceae has become a serious public health problem
worldwide [46,121,122]. In fact, ESBL-producing Escherichia coli (ESBL- 8. Conclusion
EC) are now a frequent cause of infection in the community and in
healthcare centers [122]. Bacterial antimicrobial resistance has emerged as one of the leading
public health problems globally, with all facts currently suggesting that
7.4. Carbapenem-resistant Enterobacteriaceae (CRE) the problem will become worse in the future due to the slow develop-
ment of new antimicrobial compounds vis-à-vis the escalating develop-
Isolated case reports and case series of CRE have increased world- ment and diversity of antimicrobial resistance mechanisms [3–5]. In
wide over the last decade [123]. The reported incidence of CRE varies particular, the emergence and spread of β-lactam-resistant bacteria
based on region, study methodology, and setting [124–128]. Notably, with an astonishing depth and heterogeneity of resistance mechanisms

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 9

within the healthcare and community settings signal the gravity of the [21] H.Z. Zhang, C.J. Hackbarth, K.M. Chansky, H.F. Chambers, A proteolytic transmem-
brane signaling pathway and resistance to beta-lactams in staphylococci, Science
situation, as the β-lactam antibiotics are by far the most widely pre- 291 (2001) 1962–1965.
scribed and effective of all antimicrobial compounds. There is an urgent [22] M.P. Jevons, A. Coe, M.T. Parker, Methicillin resistance in staphylococci, Lancet 1
need for a global concerted response towards both development of (1963) 904–907.
[23] A. Hartman, B. Tomasz, Altered penicillin-binding proteins in methicillin-resistant
novel antimicrobial compounds and strategies, as well as retarding the strains of Staphylococcus aureus, Antimicrob. Agents Chemother. 19 (1991)
spread and further development of antimicrobial-resistant bacteria. 726–735.
Relevant measures include facilitating as well as speeding up the devel- [24] L. Wang, G.L. Archer, Roles of ccrA and ccrB in excision and integration of staphy-
lococcal cassette chromosome mec, a Staphylococcus aureus genomic island, J.
opment, testing and approval of new antibiotics and novel drug devel- Bacteriol. 192 (2010) 3204–3212.
opment systems, and segregating different classes of antimicrobial [25] M.G. Pinho, H. de Lencastre, A. Tomasz, An acquired and a native penicillin-binding
compounds for human and animal use as far as is possible. Naturally, protein cooperate in building the cell wall of drug-resistant staphylococci, Proc.
Natl. Acad. Sci. U. S. A. 98 (2001) 10886–10891.
better efforts at infection control, surveillance and diagnostics are also
[26] H.F. Chambers, Methicillin resistance in staphylococci: molecular and biochemical
important. basis and clinical implications, Clin. Microbiol. Rev. 10 (1997) 781–791.
[27] H. Komatsuzawa, K. Ohta, H. Labischinski, M. Sugai, H. Suginaka, Characterization of
fmtA, a gene that modulates the expression of methicillin resistance in Staphylococcus
Acknowledgments aureus, Antimicrob. Agents Chemother. 43 (1999) 2121–2125.
[28] R. Leclercq, Enterococci acquire new kinds of resistance, Clin. Infect. Dis. 24 (Suppl.
1) (1997) S80–S84.
We would like to thank A/Prof. Tse Hsien Koh for vetting the [29] B.L. Hollenbeck, L.B. Rice, Intrinsic and acquired resistance mechanisms in
manuscript. Enterococcus, Virulence 35 (2012) 421–433.
[30] F. Sifaoui, M. Arthur, L. Rice, L. Gutmann, Role of penicillin-binding protein 5 in ex-
No funding was required for this work. pression of ampicillin resistance and peptidoglycan structure in Enterococcus
faecium, Antimicrob. Agents Chemother. 45 (2001) 2594–2597.
[31] Y. Comenge, R. Quintiliani Jr., L. Li, L. Dubost, J.P. Brouard, J.E. Hugonnet, M. Arthur,
References The CroRS two-component regulatory system is required for intrinsic beta-lactam
resistance in Enterococcus faecalis, J. Bacteriol. 185 (2003) 7184–7192.
[1] A. Ferriman, BMJ readers choose the “sanitary revolution” as greatest medical ad- [32] C.J. Kristich, J.L. Little, C.L. Hall, J.S. Hoff, Reciprocal regulation of cephalosporin re-
vance since 1840, BMJ 334 (2007) 111.2. sistance in Enterococcus faecalis, MBio 2 (2011) e00199-11.
[2] A. Fleming, Nobel Lecture: Penicillin, Available at: http://www.nobelprize.org/ [33] C.L. Hall, M. Tschannen, E.A. Worthey, C.J. Kristich, IreB, a Ser/Thr kinase substrate,
nobel_prizes/medicine/laureates/1945/fleming-lecture.pdf (Last accessed: 6 May influences antimicrobial resistance in Enterococcus faecalis, Antimicrob. Agents
2014). Chemother. 57 (2013) 6179–6186.
[3] G.H. Talbot, J. Bradley, J.E. Edwards Jr., D. Gilbert, M. Scheld, J.G. Bartlett, Antimicro- [34] R. Fontana, M. Aldegheri, M. Ligozzi, H. Lopez, A. Sucari, G. Satta, Overproduction of
bial Availability Task Force of the Infectious Diseases Society of America. Bad bugs a low-affinity penicillin-binding protein and high-level ampicillin resistance in
need drugs: an update on the development pipeline from the Antimicrobial Avail- Enterococcus faecium, Antimicrob. Agents Chemother. 38 (1994) 1980–1983.
ability Task Force of the Infectious Diseases Society of America, Clin. Infect. Dis. 42 [35] S. Turk, O. Verlaine, T. Gerards, M. Zivec, J. Humljan, I. Sosič, A. Amoroso, A.
(2006) 657–668. Zervosen, A. Luxen, B. Joris, S. Gobec, New noncovalent inhibitors of penicillin-
[4] World Economic Forum, Insight Report: Global Risks 2013 Eighth Edition, Avail- binding proteins from penicillin-resistant bacteria, PLoS ONE 6 (2011) e19418.
able at: http://www3.weforum.org/docs/WEF_GlobalRisks_Report_2013.pdf [36] G. Dive, C. Bouillon, A. Sliwa, B. Valet, O. Verlaine, E. Sauvage, J. Marchand-Brynaert,
(Last accessed: 6 May 2014). Macrocycle-embedded β-lactams as novel inhibitors of the penicillin binding pro-
[5] R. Laxminarayan, A. Duse, C. Wattal, A.K. Zaidi, H.F. Wertheim, N. Sumpradit, E. tein PBP2a from MRSA, Eur. J. Med. Chem. 64 (2013) 365–376.
Vlieghe, G.L. Hara, I.M. Gould, H. Goossens, C. Greko, A.D. So, M. Bigdeli, G. Tomson, [37] H. Hao, G. Cheng, M. Dai, Q. Wu, Z. Yuan, Inhibitors targeting on cell wall biosyn-
W. Woodhouse, E. Ombaka, A.Q. Peralta, F.N. Qamar, F. Mir, S. Kariuki, Z.A. Bhutta, A. thesis pathway of MRSA, Mol. BioSyst. 8 (2012) 2828–2838.
Coates, R. Bergstrom, G.D. Wright, E.D. Brown, O. Cars, Antibiotic resistance — the [38] K. Kumar, S. Chopra, New drugs for methicillin-resistant Staphylococcus aureus: an
need for global solutions, Lancet Infect. Dis. 13 (2013) 1057–1098. update, J. Antimicrob. Chemother. 68 (2013) 1465–1470.
[6] J. Davies, D. Davies, Origins and evolution of antibiotic resistance, Microbiol. Mol. [39] J.D.D. Pitout, C.C. Sanders, E. Sanders, Antimicrobial resistance with focus on
Biol. Rev. 74 (2010) 417–433. β-lactam resistance in gram-negative bacilli, Am. J. Med. 103 (1997) 51–59.
[7] D.M. Livermore, Bacterial resistance: origins, epidemiology, and impact, Clin. Infect. [40] E.P. Abraham, E. Chain, An enzyme from bacteria able to destroy penicillin, Nature
Dis. 36 (Suppl. 1) (2003) S11–S23. 146 (1940) 837.
[8] M.C. Maiden, Horizontal genetic exchange, evolution, and spread of antibiotic re- [41] B.G. Hall, M. Barlow, Revised Ambler classification of β-lactamases, J. Antimicrob.
sistance in bacteria, Clin. Infect. Dis. 27 (Suppl. 1) (1998) S12–S20. Chemother. 55 (2005) 1050–1051.
[9] H. Ochman, J.G. Lawrence, E.A. Groisman, Lateral gene transfer and the nature of [42] K. Bush, G.A. Jacoby, Updated functional classification of β-lactamases, Antimicrob.
bacterial innovation, Nature 405 (2000) 299–304. Agents Chemother. 54 (2010) 969–976.
[10] S.B. Levy, B. Marshall, Antibacterial resistance worldwide: causes, challenges and [43] P. Nordmann, Trends in β-lactam resistance among Enterobacteriaceae, Clin. Infect.
responses, Nat. Med. 10 (12 Suppl.) (2004) S122–S129. Dis. 27 (Suppl. 1) (1998) S100–S106.
[11] A. Beceiro, M. Tomás, G. Bou, Antimicrobial resistance and virulence: a successful [44] G.A. Jacoby, L.S. Munoz-Price, The new β-lactamases, N. Engl. J. Med. 352 (2005)
or deleterious association in the bacterial world? Clin. Microbiol. Rev. 26 (2013) 380–391.
185–230. [45] G.A. Jacoby, AmpC β-lactamases, Clin. Microbiol. Rev. 22 (2009) 161–182.
[12] R.I. Aminov, A brief history of the antibiotic era: lessons learned and challenges for [46] D.L. Paterson, R.A. Bonomo, Extended-spectrum β-lactamases: a clinical update,
the future, Front. Microbiol. 1 (2010) 134. Clin. Microbiol. Rev. 18 (2005) 657–686.
[13] M.J. Cloutier, Antibiotics: mechanisms of action and the acquisition of resistance — [47] Y. Pfeifer, A. Cullik, W. Witte, Resistance to cephalosporins and carbapenems in
when magic bullets lose their magic, Am. J. Pharm. Educ. 59 (1995) 167–172. Gram-negative bacterial pathogens, Int. J. Med. Microbiol. 300 (2010) 371–379.
[14] A.Y. Peleg, D.C. Hooper, Hospital-acquired infections due to gram-negative bacte- [48] D.L. Paterson, Resistance in gram-negative bacteria: Enterobacteriaceae, Am. J.
ria, N. Engl. J. Med. 362 (2010) 1804–1813. Infect. Control 34 (5 Suppl. 1) (2006) S20–S28.
[15] J.H. Jorgensen, M.J. Ferraro, Antimicrobial susceptibility testing: a review of general [49] W.H. Zhao, Z.Q. Hu, Epidemiology and genetics of CTX-M extended-spectrum
principles and contemporary practices, Clin. Infect. Dis. 49 (2009) 1749–1755. β-lactamases in Gram-negative bacteria, Crit. Rev. Microbiol. 39 (2013) 79–101.
[16] European Committee on Antimicrobial Susceptibility Testing, Clinical Breakpoints, [50] D.M. Livermore, N. Woodford, The β-lactamase threat in Enterobacteriaceae.
Available at: http://www.eucast.org/clinical_breakpoints/ (Last accessed: 6 May Pseudomonas and Acinetobacter, Trends Microbiol. 14 (2006) 413–420.
2014). [51] CDC, Antibiotic Resistance Threats in the United States, 2013, http://www.cdc.gov/
[17] Clinical and Laboratory Standards Institute, Microbiology Standards, Available at: drugresistance/threat-report-2013/pdf/ar-threats-2013-508.pdf 2013 (Accessed
http://clsi.org/standards/micro/ (Last accessed: 6 May 2014). 6th December 2013).
[18] M.T. Holden, L.Y. Hsu, K. Kurt, L.A. Weinert, A.E. Mather, S.R. Harris, B. Strommenger, F. [52] J. Walter-Rasmussen, N. Hoiby, OXA-type carbapenemases, J. Antimicrob.
Layer, W. Witte, H. de Lencastre, R. Skov, H. Westh, H. Zemlicková, G. Coombs, A.M. Chemother. 57 (2006) 373–383.
Kearns, R.L. Hill, J. Edgeworth, I. Gould, V. Gant, J. Cooke, G.F. Edwards, P.R. McAdam, [53] T.R. Walsh, Clinically significant carbapenemases: an update, Curr. Opin. Infect. Dis.
K.E. Templeton, A. McCann, Z. Zhou, S. Castillo-Ramírez, E.J. Feil, L.O. Hudson, M.C. 21 (2008) 367–371.
Enright, F. Balloux, D.M. Aanensen, B.G. Spratt, J.R. Fitzgerald, J. Parkhill, M. Achtman, [54] S. Corvec, N. Caroff, E. Espaze, C. Giraudeau, H. Drugeon, A. Reynaud, AmpC
S.D. Bentley, U. Nübel, A genomic portrait of the emergence, evolution, and global cephalosporinase hyperproduction in Acinetobacter baumanii clinical strains, J.
spread of a methicillin-resistant Staphylococcus aureus pandemic, Genome Res. 23 Antimicrob. Chemother. 52 (2003) 629–635.
(2013) 653–664. [55] M. Hamidian, D.P. Hancock, R.M. Hall, Horizontal transfer of an ISAba125-activated
[19] C.M. Martin, C.M. Kunin, L.S. Gottlieb, M. Finland, Asian influenza A in Boston, AmpC gene between Acinetobacter baumannii strains leading to cephalosporin
1957–1958. II. Severe staphylococcal pneumonia complicating influenza, AMA resistance, J. Antimicrob. Chemother. 68 (2013) 244–245.
Arch. Intern. Med. 103 (1959) 532–542. [56] J.M. Rodríguez-Martínez, P. Nordmann, E. Ronco, L. Poirel, Extended-spectrum
[20] F.D. Lowy, Antimicrobial resistance: the example of Staphylococcus aureus, J. Clin. cephalosporinase in Acinetobacter baumannii, Antimicrob. Agents Chemother. 54
Invest. 111 (2003) 1265–1273. (2010) 3484–3488.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
10 S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

[57] G.B. Tian, J.M. Adams-Haduch, M. Taracila, R.A. Bonomo, H.N. Wang, Y. Doi, [87] L.D. Saravolatz, N. Markowitz, L. Arking, D. Pohlod, E. Fisher, Methicillin-resistant
Extended-spectrum AmpC cephalosporinase in Acinetobacter baumannii: ADC-56 Staphylococcus aureus. Epidemiologic observations during a community-acquired
confers resistance to cefepime, Antimicrob. Agents Chemother. 55 (2011) outbreak, Ann. Intern. Med. 96 (1982) 11–16.
4922–4925. [88] E.E. Udo, J.W. Pearman, W.B. Grubb, Genetic analysis of community isolates of
[58] J.F. Turton, M.E. Ward, N. Woodford, M.E. Kaufmann, R. Pike, D.M. Livermore, T.L. methicillin-resistant Staphylococcus aureus in Western Australia, J. Hosp. Infect.
Pitt, The role of ISAba1 in expression of OXA carbapenemase genes in Acinetobacter 25 (1993) 97–108.
baumannii, FEMS Microbiol. Lett. 258 (2006) 72–77. [89] CDC, Four pediatric deaths from community-acquired methicillin-resistant
[59] L. Krizova, L. Poirel, P. Nordmann, A. Nemec, TEM-1 β-lactamase as a source of Staphylococcus aureus — Minnesota and North Dakota, 1997–1999, JAMA 282
resistance to sulbactam in clinical strains of Acinetobacter baumannii, J. Antimicrob. (1999) 1123–1125.
Chemother. 68 (2013) 2786–2791. [90] F.R. DeLeo, M. Otto, B.N. Kreiswirth, H.F. Chambers, Community-associated
[60] L. Poirel, R.A. Bonnin, P. Nordmann, Genetic basis of antibiotic resistance in pathogenic meticillin-resistant Staphylococcus aureus, Lancet 375 (2010) 1557–1558.
Acinetobacter species, IUBMB Life 63 (2011) 1061–1067. [91] M. Otto, MRSA virulence and spread, Cell. Microbiol. 14 (2012) 1513–1521.
[61] A. Potron, L. Poirel, J. Croizé, V. Chanteperdrix, P. Nordmann, Genetic and biochem- [92] J.N. Jimenez, A.M. Ocampo, J.M. Vanegas, E.A. Rodriguez, J.R. Mediavilla, L. Chen, C.E.
ical characterization of the first extended-spectrum CARB-type beta-lactamase, Muskus, L.A. Vélez, C. Rojas, A.V. Restrepo, S. Ospina, C. Garcés, L. Franco, P. Bifani,
RTG-4, from Acinetobacter baumannii, Antimicrob. Agents Chemother. 53 (2009) B.N. Kreiswirth, M.M. Correa, CC8 MRSA strains harboring SCCmec type IVc are
3010–3016. predominant in Colombian hospitals, PLoS ONE 7 (2012) e38576.
[62] R. Zarrilli, S. Pournaras, M. Giannouli, A. Tsakris, Global evolution of multidrug- [93] P. Tattevin, B.A. Diep, M. Jula, F. Perdreau-Remington, Long-term follow-up of
resistant Acinetobacter baumannii clonal lineages, Int. J. Antimicrob. Agents 41 methicillin-resistant Staphylococcus aureus molecular epidemiology after emer-
(2013) 11–19. gence of clone USA300 in San Francisco jail populations, J. Clin. Microbiol. 46
[63] S. Brown, S. Amyes, OXA β-lactamases in Acinetobacter: the story so far, J. (2008) 4056–4057.
Antimicrob. Chemother. 57 (2006) 1–3. [94] P. Tattevin, B.A. Diep, M. Jula, F. Perdreau-Remington, Methicillin-resistant
[64] B. Gionco, J.S. Pelayo, E.J. Venancio, R. Cayô, A.C. Gales, F.E. Carrara-Marroni, Detection Staphylococcus aureus USA300 clone in long-term care facility, Emerg. Infect. Dis.
of OXA-231, a new variant of blaOXA-143, in Acinetobacter baumannii from Brazil: a case 15 (2009) 953–955.
report, J. Antimicrob. Chemother. 67 (2012) 2531–2532. [95] H. Graveland, B. Duim, E. van Duijkeren, D. Heederik, J.A. Wagenaar, Livestock-
[65] P.G. Higgins, L. Poirel, M. Lehmann, P. Nordmann, H. Seifert, OXA-143, a novel car- associated methicillin-resistant Staphylococcus aureus in animals and humans,
bapenem hydrolyzing class D β-lactamase in Acinetobacter baumannii, Antimicrob. Int. J. Med. Microbiol. 301 (2011) 630–634.
Agents Chemother. 53 (2009) 5035–5038. [96] L. Guardabassi, M. O'Donoghue, A. Moodley, J. Ho, M. Boost, Novel lineage
[66] J. Villa, S. Marti, J. Sanchez-Cespedes, Porins, efflux pumps and multidrug resistance of methicillin-resistant Staphylococcus aureus, Hong Kong, Emerg. Infect. Dis. 15
in Acinetobacter baumannii, J. Antimicrob. Chemother. 59 (2007) 1210–1215. (12) (2009) 1998–2000.
[67] M. Catel-Ferreira, R. Nehme, V. Molle, J. Aranda, E. Bouffartigues, S. Chevalier, G. [97] M.N. Mulders, A.P.J. Haenen, P.L. Geenen, P.C. Vesseur, E.S. Poldervaart, T. Bosch, X.W.
Bou, T. Jouenne, E. De, Deciphering the function of the outer membrane protein Huijsdens, P.D. Hengeveld, W.D. Dam-Deisz, E.A. Graat, D. Mevius, A. Voss, A.W. Van
OprD homologue of Acinetobacter baumannii, Antimicrob. Agents Chemother. 56 De Giessen, Prevalence of livestock-associated MRSA in broiler flocks and risk factors
(2012) 3826–3832. for slaughterhouse personnel in The Netherlands, Epidemiol. Infect. 138 (2010)
[68] P. Wieczorek, P. Sacha, T. Hauschild, M. Zorawski, M. Krawczyk, E. TTryniszewska, 743–755.
Multidrug resistant Acinetobacter baumannii — the role of AdeABC (RND family) [98] C. Garcia-Graells, J. Antoine, J. Larsen, B. Catry, R. Sko, O. Denis, Livestock veterinarians
efflux pump in resistance to antibiotics, Folia Histochem. Cytobiol. 46 (2008) at high risk of acquiring methicillin-resistant Staphylococcus aureus ST398, Epidemiol.
257–267. Infect. 140 (2012) 383–389.
[69] S. Coyne, P. Courvalin, B. Perichon, Efflux-mediated antibiotic resistance in [99] J.A.J.W. Kluytmans, Methicillin-resistant Staphylococcus aureus in food products:
Acinetobacter spp. Antimicrob. Agents Chemother. 55 (2011) 947–953. cause for concern or case for complacency? Clin. Microbiol. Infect. 16 (2010)
[70] S.M. Drawz, R.A. Bonomo, Three decades of β-lactamase inhibitors, Clin. Microbiol. 11–15.
Rev. 23 (2010) 160–201. [100] E. de Boer, J.T.M. Zwartkruis-Nahuis, B. Wit, X.W. Huijsdens, A.J. de Neeling, T.
[71] R.R. Watkins, K.M. Papp-Wallace, S.M. Drawz, R.A. Bonomo, Novel β-lactamase Bosch, R.A. van Oosterom, A. Vila, A.E. Heuvelink, Prevalence of methicillin-resistant
inhibitors: a therapeutic hope against the scourge of multidrug resistance, Front. Staphylococcus aureus in meat, Int. J. Food Microbiol. 134 (2009) 52–56.
Microbiol. 4 (2013) 392. [101] I.H.M. van Loo, B.M.W. Diederen, P.H.M. Savelkoul, J.H. Woudenberg, R. Roosendaal, A.
[72] S.M. Drawz, K.M. Papp-Wallace, R.A. Bonomo, New β-lactamase inhibitors: van Belkum, N. Lemmens-den Toom, C. Verhulst, P.H. van Keulen, J.A. Kluytmans,
a therapeutic renaissance in an MDR world, Antimicrob. Agents Chemother. 58 Methicillin-resistant Staphylococcus aureus in meat products, the Netherlands,
(2014) 1835–1846. Emerg. Infect. Dis. 13 (11) (2007) 1753–1755.
[73] C. Lucasti, I. Popescu, M.K. Ramesh, J. Lipka, C. Sable, Comparative study of the [102] R. Leclercq, E. Derlot, J. Duval, P. Courvalin, Plasmid-mediated resistance to
efficacy and safety of ceftazidime/avibactam plus metronidazole versus meropenem vancomycin and teicoplanin in Enterococcus faecium, N. Engl. J. Med. 319
in the treatment of complicated intra-abdominal infections in hospitalized adults: (1988) 157–161.
results of a randomized, double-blind, Phase II trial, J. Antimicrob. Chemother. 68 [103] A.H. Uttley, C.H. Collins, J. Naidoo, R.C. George, Vancomycin-resistant enterococci,
(2013) 1183–1192. Lancet 1 (1988) 57–58.
[74] J.A. Vazquez, L.D. González Patzán, D. Stricklin, et al., Efficacy and safety of [104] D.E. Low, N. Keller, A. Barth, R.N. Jones, Clinical prevalence, antimicrobial susceptibility,
ceftazidime-avibactam versus imipenem-cilastatin in the treatment of complicat- and geographic resistance patterns of enterococci: results from the SENTRY Antimi-
ed urinary tract infections, including acute pyelonephritis, in hospitalized adults: crobial Surveillance Program, 1997–1999, Clin. Infect. Dis. 32 (Suppl. 2) (2001)
results of a prospective, investigator-blinded, randomized study, Curr. Med. Res. S133–S145.
Opin. 28 (2012) 1921–1931. [105] Nosocomial enterococci resistant to vancomycin — United States, 1989–1993,
[75] K. Coleman, Diazabicyclooctanes (DBOs): a potent new class of non-β-lactam MMWR Morb. Mortal. Wkly Rep. 42 (1993) 597–599.
β-lactamase inhibitors, Curr. Opin. Microbiol. 14 (2011) 550–555. [106] National Nosocomial Infections Surveillance (NNIS) System Report, data summary
[76] Q. Tan, A.M. Ogawa, R.E. Painter, Y.W. Park, K. Young, F.P. DiNinno, 4,7-Dichloro from January 1992 through June 2004, issued October 2004, Am. J. Infect. Control
benzothien-2-yl sulfonylaminomethyl boronic acid: first boronic acid-derived 32 (2004) 470–485.
beta-lactamase inhibitor with class A, C, and D activity, Bioorg. Med. Chem. Lett. [107] T.M. Coque, J.F. Tomayko, S.C. Ricke, P.C. Okhyusen, B.E. Murray, Vancomycin-
20 (2010) 2622–2624. resistant enterococci from nosocomial, community, and animal sources in the
[77] J.D. Buynak, β-Lactamase inhibitors: a review of the patent literature (2010–2013), United States, Antimicrob. Agents Chemother. 40 (1996) 2605–2609.
Expert Opin. Ther. Pat. 23 (2013) 1469–1481. [108] L.A. Thal, J.W. Chow, R. Mahayni, et al., Characterization of antimicrobial resis-
[78] Faridoon, N. Ul Islam, An update on the status of potent inhibitors of metallo-β- tance in enterococci of animal origin, Antimicrob. Agents Chemother. 39
lactamases, Sci. Pharm. 81 (2013) 309–327. (1995) 2112–2115.
[79] Ashima K. Bhardwaj, Priyabrata Mohanty, Bacterial efflux pumps involved in [109] M.J. Bonten, R. Willems, R.A. Weinstein, Vancomycin-resistant enterococci: why
multidrug resistance and their inhibitors: rejuvenating the antimicrobial chemo- are they here, and where do they come from? Lancet Infect. Dis. 1 (2001) 314–325.
therapy, Recent Pat. Antiinfect Drug Discov. 7 (2012) 73–89. [110] T.L. Sorensen, M. Blom, D.L. Monnet, N. Frimodt-Moller, R.L. Poulsen, F. Espersen,
[80] J.M. Pagès, L. Amaral, Mechanisms of drug efflux and strategies to combat them: Transient intestinal carriage after ingestion of antibioticresistant Enterococcus
challenging the efflux pump of Gram-negative bacteria, Biochim. Biophys. Acta faecium from chicken and pork, N. Engl. J. Med. 345 (2001) 1161–1166.
1794 (2009) 826–833. [111] A.E. van den Bogaard, N. Bruinsma, E.E. Stobberingh, The effect of banning
[81] K.I. Kaitin, Deconstructing the drug development process: the new face of innovation, avoparcin on VRE carriage in The Netherlands, J. Antimicrob. Chemother. 46
Clin. Pharmacol. Ther. 87 (2010) 356–361. (2000) 146–148.
[82] A.J. Huh, Y.J. Kwon, “Nanoantibiotics”: a new paradigm for treating infectious [112] F.M. Aarestrup, A.M. Seyfarth, H.D. Emborg, K. Pedersen, R.S. Hendriksen, F. Bager,
diseases using nanomaterials in the antibiotics resistant era, J. Control. Release Effect of abolishment of the use of antimicrobial agents for growth promotion on
156 (2011) 128–145. occurrence of antimicrobial resistance in fecal enterococci from food animals in
[83] T.M. Allen, P.R. Cullis, Liposomal drug delivery systems: from concept to clinical Denmark, Antimicrob. Agents Chemother. 45 (2001) 2054–2059.
applications, Adv. Drug Deliv. Rev. 65 (2013) 36–48. [113] S.K. Lim, T.S. Kim, H.S. Lee, H.M. Nam, Y.S. Joo, H.B. Koh, Persistence of vanA-type
[84] M.H. Xiong, Y. Bao, X.Z. Yang, Y.H. Zhu, J. Wang, Delivery of antibiotics with Enterococcus faecium in Korean livestock after ban on avoparcin, Microb. Drug
polymeric particles, Adv. Drug Deliv. Rev. (2014) (Epub ahead of print). Resist. 12 (2006) 136–139.
[85] K. Hadinoto, S. Chewo, Nano-antibiotics in chronic lung infection therapy against [114] D. Sirot, J. Sirot, R. Labia, A. Morand, P. Courvalin, A. Darfeuille-Michaud, R. Perroux, R.
Pseudomonas aeruginosa, Colloids Surf. B: Biointerfaces 116 (2014) 772–785. Cluzel, Transferable resistance to third-generation cephalosporins in clinical isolates of
[86] L.Y. Hsu, L. Wijaya, B.H. Tan, Management of healthcare-associated methicillin- Klebsiella pneumoniae: identification of CTX-1, a novel β-lactamase, J. Antimicrob.
resistant Staphylococcus aureus, Expert Rev. Anti-Infect. Ther. 3 (2005) 893–905. Chemother. 20 (1987) 323–334.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003
S.S. Tang et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 11

[115] P.A. Bradford, Extended-spectrum b-lactamases in the 21st century: characterization, pneumoniae sequence type 14 from India, Antimicrob. Agents Chemother. 53
epidemiology, and detection of this important resistance threat, Clin. Microbiol. Rev. (2009) 5046–5054.
14 (2001) 933–951. [133] M.J. Struelens, D.L. Monnet, A.P. Magiorakos, F. Santos O'Connor, J. Giesecke, New
[116] M.A. Pfaller, J. Segreti, Overview of the epidemiological profile and laboratory Delhi metallo-beta-lactamase 1-producing Enterobacteriaceae: emergence and
detection of extended-spectrum beta-lactamases, Clin. Infect. Dis. 42 (Suppl. 4) response in Europe, Euro Surveill. 15 (2010) (pii:19716).
(2006) S153–S163. [134] H.C. Maltezou, P. Giakkoupi, A. Maragos, M. Bolikas, V. Raftopoulos, H. Papahatzaki, G.
[117] R.N. Jones, D.J. Biedenbach, A.C. Gales, Sustained activity and spectrum of selected Vrouhos, V. Liakou, A.C. Vatopoulos, Outbreak of infections due to KPC-2-producing
extended-spectrum b-lactams (carbapenems and cefepime) against Enterobacter Klebsiella pneumoniae in a hospital in Crete (Greece), J. Infect. 58 (2009) 213–219.
spp. and ESBL-producing Klebsiella spp.: report from the SENTRY antimicrobial [135] P. Savard, T.M. Perl, A call for action: managing the emergence of multidrug-
surveillance program (USA, 1997–2000), Int. J. Antimicrob. Agents 21 (2003) 1–7. resistant Enterobacteriaceae in the acute care settings, Curr. Opin. Infect. Dis. 25
[118] A.C. Gales, H.S. Sader, R.N. Jones, Urinary tract infection trends in Latin American (2012) 371–377.
hospitals: report from the SENTRY antimicrobial surveillance program (1997–2000), [136] D.A. Williamson, H.E. Sidjabat, J.T. Freeman, S.A. Roberts, A. Silvey, R. Woodhouse,
Diagn. Microbiol. Infect. Dis. 44 (2002) 289–299. E. Mowat, K. Dyet, D.L. Paterson, T. Blackmore, A. Burns, H. Heffernan, Identification
[119] J.M. Bell, J.D. Turnidge, A.C. Gales, M.A. Pfaller, R.N. Jones, Prevalence of extended and molecular characterisation of New Delhi metallo-β-lactamase-1 (NDM-1)-
spectrum β-lactamase (ESBL)-producing clinical isolates in the Asia-Pacific region and NDM-6-producing Enterobacteriaceae from New Zealand hospitals, Int. J.
and South Africa: regional results from SENTRY Antimicrobial Surveillance Pro- Antimicrob. Agents 39 (2012) 529–533.
gram (1998–99), Diagn. Microbiol. Infect. Dis. 42 (2002) 193–198. [137] C.M. Ying, T.K. Ling, C.C. Lee, J.M. Ling, Characterization of carbapenem-resistant
[120] P.L. Winokur, R. Canton, J.M. Casellas, N. Legakis, Variations in the prevalence of Acinetobacter baumannii in Shanghai and Hong Kong, J. Med. Microbiol. 55
strains expressing an extended-spectrum β-lactamase phenotype and characteri- (2006) 799–802.
zation of isolates from Europe, the Americas, and the Western Pacific region, [138] T.H. Koh, T.T. Tan, C.T. Khoo, S.Y. Ng, T.Y. Tan, L.Y. Hsu, E.E. Ooi, T.J. Van Der Reijden,
Clin. Infect. Dis. 32 (Suppl. 2) (2001) S94–S103. L. Dijkshoorn, Acinetobacter calcoaceticus–Acinetobacter baumannii complex species
[121] J.D.D. Pitout, K.B. Laupland, Extended-spectrum β-lactamase-producing in clinical specimens in Singapore, Epidemiol. Infect. 140 (2012) 535–538.
Enterobacteriaceae: an emerging public-health concern, Lancet Infect. Dis. 8 [139] C. Routsi, M. Pratikaki, E. Platsouka, C. Sotiropoulou, S. Nanas, V. Markaki, C.
(2008) 159–166. Vrettou, O. Paniara, H. Giamarellou, C. Roussos, Carbapenem-resistant versus
[122] A. Apisarnthanarak, P. Kiratisin, P. Saifon, R. Kitphati, S. Dejsirilert, L.M. Mundy, carbapenem-susceptible Acinetobacter baumannii bacteremia in a Greek intensive
Clinical and molecular epidemiology of community-onset, extended-spectrum care unit: risk factors, clinical features and outcomes, Infection 38 (2010) 173–180.
beta-lactamase-producing Escherichia coli infections in Thailand: a case–case–control [140] S. Le Hello, V. Falcot, F. Lacassin, M. Mikulski, F. Baumann, Risk factors for
study, Am. J. Infect. Control 35 (2007) 606–612. carbapenem-resistant Acinetobacter baumannii infections at a tertiary care hospital
[123] A. Apisarnthanarak, L.Y. Hsu, T. Khawcharoenporn, L.M. Mundy, Carbapenem- in New Caledonia, South Pacific, Scand. J. Infect. Dis. 42 (2010) 821–826.
resistant Gram-negative bacteria: how to prioritize infection prevention and [141] A. Asensio, R. Cantón, J. Vaqué, F. Calbo-Torrecillas, R. Herruzo, J.L. Arribas, M.C.
control interventions in resource-limited settings? Expert Rev. Anti-Infect. Ther. Sáenz, Grupo de Trabajo EPINE, Prevalence of infection by carbapenem-resistant
11 (2013) 147–157. Acinetobacter baumannii in Spain (1999–2005), Enferm. Infecc. Microbiol. Clin. 21
[124] N. Gupta, B.M. Limbago, J.B. Patel, A.J. Kallen, Carbapenem-resistant Entero- (2008) 199–204.
bacteriaceae: epidemiology and prevention, Clin. Infect. Dis. 53 (2011) 60–67. [142] A.Y. Peleg, C. Franklin, J.M. Bell, D.W. Spelman, Emergence of carbapenem resis-
[125] K.K. Kumarasamy, M.A. Toleman, T.R. Walsh, J. Bagaria, F. Butt, R. Balakrishnan, U. tance in Acinetobacter baumannii recovered from blood cultures in Australia, Infect.
Chaudhary, M. Doumith, C.G. Giske, S. Irfan, P. Krishnan, A.V. Kumar, S. Maharjan, S. Control Hosp. Epidemiol. 27 (2006) 759–761.
Mushtaq, T. Noorie, D.L. Paterson, A. Pearson, C. Perry, R. Pike, B. Rao, U. Ray, J.B. [143] Y.J. Kim, S.I. Kim, Y.R. Kim, K.W. Hong, S.H. Wie, Y.J. Park, H. Jeong, M.W. Kang,
Sarma, M. Sharma, E. Sheridan, M.A. Thirunarayan, J. Turton, S. Upadhyay, M. Carbapenem-resistant Acinetobacter baumannii: diversity of resistant mechanisms
Warner, W. Welfare, D.M. Livermore, N. Woodford, Emergence of a new antibiotic and risk factors for infection, Epidemiol. Infect. 140 (2012) 137–145.
resistance mechanism in India, Pakistan, and the UK: a molecular, biological, and [144] S. Dejsirilert, S. Tiengrim, P. Sawanpanyalert, N. Aswapokee, K. Malathum, Antimi-
epidemiological study, Lancet Infect. Dis. 10 (2010) 597–602. crobial resistance of Acinetobacter baumannii: six years of National Antimicrobial
[126] L.F. Mataseje, E. Bryce, D. Roscoe, D.A. Boyd, J. Embree, D. Gravel, K. Katz, P. Kibsey, Resistance Surveillance Thailand (NARST) surveillance, J. Med. Assoc. Thail. 92
M. Kuhn, A. Mounchili, A. Simor, G. Taylor, E. Thomas, N. Turgeon, M.R. Mulvey, (2009) S34–S45.
Canadian Nosocomial Infection Surveillance Program. Carbapenem-resistant [145] C.H. Su, J.T. Wang, C.A. Hsiung, L.J. Chien, C.L. Chi, H.T. Yu, F.Y. Chang, S.C. Chang,
Gram-negative bacilli in Canada 2009–10: results from the Canadian Nosocomial Increase of carbapenem-resistant Acinetobacter baumannii infection in acute care
Infection Surveillance Program (CNISP), J. Antimicrob. Chemother. 67 (2012) hospitals in Taiwan: association with hospital antimicrobial usage, PLoS ONE 7
1359–1367. (2012) e37788.
[127] A. Vatopoulos, High rates of metallo-beta-lactamase-producing Klebsiella [146] E. Akinci, A. Colpan, H. Bodur, N. Balaban, A. Erbay, Risk factors for ICU-acquired
pneumoniae in Greece—a review of the current evidence, Euro Surveill. 13 imipenem-resistant Gram-negative bacterial infections, J. Hosp. Infect. 59 (2005)
(2008) (pii:8023). 317–323.
[128] A. Leavitt, S. Navon-Venezia, I. Chmelnitsky, M.J. Schwaber, Y. Carmeli, Emergence [147] J.M. Cisneros, J. Rodríguez-Baño, F. Fernández-Cuenca, A. Ribera, J. Vila, A. Pascual,
of KPC-2 and KPC-3 in carbapenem-resistant Klebsiella pneumoniae strains in an L. Martínez-Martínez, G. Bou, J. Pachón, Spanish Group for Nosocomial Infection
Israeli hospital, Antimicrob. Agents Chemother. 51 (2007) 3026–3029. (GEIH) for the Spanish Society of Infectious Diseases and Clinical Microbiology
[129] A.I. Hidron, J.R. Edwards, J. Patel, T.C. Horan, D.M. Sievert, D.A. Pollock, S.K. Fridkin, (SEIMC), Risk-factors for the acquisition of imipenem-resistant Acinetobacter
National Healthcare Safety Network Team, Participating National Healthcare Safety baumannii in Spain: a nationwide study, Clin. Microbiol. Infect. 11 (2005) 874–879.
Network Facilities. NHSN annual update: antimicrobial-resistant pathogens associ- [148] E.G. Playford, J.C. Craig, J.R. Iredell, Carbapenem-resistant Acinetobacter baumannii
ated with healthcare-associated infections: annual summary of data reported to the in intensive care unit patients: risk factors for acquisition, infection and their
National Healthcare Safety Network at the Centers for Disease Control and Prevention, consequences, J. Hosp. Infect. 65 (2007) 204–211.
2006–2007, Infect. Control Hosp. Epidemiol. 29 (2008) 996–1011. [149] H. Aydemir, G. Celebi, N. Piskin, N. Oztoprak, A.S. Keskin, E. Aktas, V. Sumbuloglu, D.
[130] P.R. Rhomberg, R.N. Jones, Summary trends for the Meropenem Yearly Susceptibil- Akduman, Mortality attributable to carbapenem-resistant nosocomial Acinetobacter
ity Test Information Collection program: a 10-year experience in the United States baumannii infections in a Turkish university hospital, Jpn. J. Infect. Dis. 65 (2012)
(1999–2008), Diagn. Microbiol. Infect. Dis. 65 (2009) 414–426. 66–71.
[131] H. Yigit, A.M. Queenan, G.J. Anderson, A. Domenech-Sanchez, J.M. Biddle, C.D. Steward, [150] P.G. Higgins, C. Dammhayn, M. Hackel, H. Seifert, Global spread of carbapenem-
S. Alberti, K. Bush, F.C. Tenover, Novel carbapenem hydrolyzing β-lactamase, KPC-1, resistant Acinetobacter baumannii, J. Antimicrob. Chemother. 65 (2010) 233–238.
from a carbapenem-resistant strain of Klebsiella pneumoniae, Antimicrob. Agents [151] D.H. Kim, J.Y. Choi, H.W. Kim, S.H. Kim, D.R. Chung, K.R. Peck, V. Thamlikitkul, T.M.
Chemother. 45 (2011) 1151–1161. So, R.M. Yasin, P.R. Hsueh, C.C. Carlos, L.Y. Hsu, L. Buntaran, M.K. Lalitha, J.H. Song,
[132] D. Yong, M.A. Toleman, C.G. Giske, H.S. Cho, K. Sundman, K. Lee, T.R. Walsh, K.S. Ko, Spread of carbapenem-resistant Acinetobacter baumannii global clone 2 in
Characterization of a new metallo-beta-lactamase gene, bla (NDM-1), and a Asia and AbaR-type resistance islands, Antimicrob. Agents Chemother. 57 (2013)
novel erythromycin esterase gene carried on a unique genetic structure in Klebsiella 5239–5246.

Please cite this article as: S.S. Tang, et al., Mechanisms of β-lactam antimicrobial resistance and epidemiology of major community- and
healthcare-associated multidrug-resistant..., Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.003

Potrebbero piacerti anche