Antimicrobial Resistance in ESKAPE Pathogens
Antimicrobial Resistance in ESKAPE Pathogens
Antimicrobial Resistance in ESKAPE Pathogens
crossm
a School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
b Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
c
UQ Centre for Clinical Research, The University of Queensland, QLD, Australia
SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
VANCOMYCIN-RESISTANT ENTEROCOCCI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
METHICILLIN-RESISTANT STAPHYLOCOCCUS AUREUS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
KLEBSIELLA PNEUMONIAE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
ACINETOBACTER BAUMANNII . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
PSEUDOMONAS AERUGINOSA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
ENTEROBACTER SPECIES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
ESCHERICHIA COLI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
ESKAPE PATHOGEN MECHANISMS OF ANTIBIOTIC RESISTANCE . . . . . . . . . . . . . . . . . . . . . . . . . 8
Antibiotic Inactivation/Alteration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
-Lactamases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Aminoglycoside-modifying enzymes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Target Site Modifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Target enzyme modifications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Ribosomal target site alterations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Cell wall precursor alterations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Reduced Antibiotic Penetration and Accumulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Porins. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Efflux pumps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Other Mechanisms and Survival Strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Biofilms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Antibiotic tolerance and persistence. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Intracellular survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
MOBILE GENETIC ELEMENTS CONFERRING ANTIMICROBIAL RESISTANCE . . . . . . . . . . . . . 16
Insertion Sequences and Transposons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Plasmids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Genomic Islands and Integrative Conjugative Elements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Contribution of Horizontal Gene Transfer to the Spread of Mobile Genetic
Elements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Coselection of Antimicrobial Resistance with Detergents and Biocides . . . . . . . . . . . . . . . . . 20
THERAPEUTIC ADVANCES AGAINST ESKAPE PATHOGENS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Recently Approved Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
New Drug Classes in Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
New Drugs in Clinical Trials—Overcoming Antibiotic Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Alternative Drug Trial Approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Combinational Drug Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Blocking resistance mechanisms against existing antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
(i) Class I adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
(ii) Class II adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Alternative Nondrug Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Citation De Oliveira DMP, Forde BM, Kidd TJ,
Bacteriophage therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Harris PNA, Schembri MA, Beatson SA, Paterson
Repurposing existing drugs used for noninfectious disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
DL, Walker MJ. 2020. Antimicrobial resistance in
Monoclonal antibody therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
ESKAPE pathogens. Clin Microbiol Rev
Vaccine development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
33:e00181-19. https://doi.org/10.1128/CMR
FMT strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
.00181-19.
OUTLOOK . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 Copyright © 2020 American Society for
AUTHOR BIOS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49 Microbiology. All Rights Reserved.
Address correspondence to Mark J. Walker,
[email protected].
Published 13 May 2020
INTRODUCTION
was recommended in December 2019, and approval was provided in February 2020)
(22–25). Apart from these antimicrobials, during this time frame, the E.U. EMA addi-
tionally approved ceftobiprole (Zeftera; also approved by the Australian Therapeutic
Goods Agency in 2016 and by Health Canada in 2015), whereas the Japanese Pharma-
ceutical and Medical Devices Agency (PMDA) approved lascufloxacin (Lasvic) (26–29).
Global initiatives to deliver new stand-alone antibacterial therapies or complementing
alternative therapies are urgently needed. In this review, we assess the current state of
AMR in ESKAPE pathogens, with a focus on current and emerging drug development
avenues in the response against AMR.
VANCOMYCIN-RESISTANT ENTEROCOCCI
Enterococcus faecium is a prominent cause of health care-associated infections, and
hospital-adapted lineages are increasingly resistant to vancomycin (30) (Table 1). The
dissemination of Enterococcus in the United States occurred in two separate waves. The
first wave began in the 1980s and was associated with the introduction of third-
generation cephalosporins driving the emergence of vancomycin- and ampicillin-
resistant Enterococcus faecalis (31). The second wave, dominated by vancomycin-
resistant E. faecium (VREfm), was hypothesized to have spread from the United States
to other parts of the world. Several European countries have now reported increases in
VREfm prevalence in hospitalized patients (32, 33). In Australia, 47% of E. faecium blood
culture isolates are VREfm, contributing to an incidence rate of vancomycin-resistant
enterococci (VRE) which surpasses that of many other high-income nations (34, 35).
VREfm multilocus sequence types (ST) pertaining to clonal complex 17 (CC17) are
currently responsible for a significant burden of hospital-acquired infection (36). Highly
prevalent in the gut microbiome of wild and domesticated animals (37, 38), CC17
strains have been associated with outbreaks in Europe, Asia, South America, and
Australia (34, 39–42). Although the zoonotic transfer of CC17 strains from animals to
humans is largely attributed to the spread of this complex, fresh food has also been
found to be a significant reservoir (36). Despite spread in the community appearing
high, community-associated infections caused by CC17 strains are uncommon.
Compared to the durations of outbreaks caused by the other ESKAPE pathogens,
VREfm outbreaks have a long duration, approximating 11 months, on average (43, 44).
The entry of VREfm into the bloodstream of hospitalized patients is typically preceded
by antibiotic exposure, enabling VREfm to become the predominant species in the
gastrointestinal tract (45, 46). The duration of prior antibiotic exposure is strongly
associated with a subsequent risk of VRE infection (47). In a 2016 national survey of
1,058 bloodstream infections caused by Enterococcus in Australia, almost 50% of E.
faecium isolates were vancomycin resistant (48). In the United States, the incidence of
hospital- and community-acquired VRE infection between 2012 and 2017 significantly
decreased (4). The management of patients infected with VRE is complicated by the
excess cost and disruption resulting from the need for isolation rooms, contact pre-
cautions, and dedicated room cleaning. The treatment of significant infection relies
upon second-line antibiotic therapies (e.g., tigecycline and daptomycin), which are
often associated with increased cost, diminished efficacy, and a greater risk of toxicity
compared with the cost, efficacy, and risk of toxicity of first-line antibiotic therapies
(Table 1) (49, 50). Defining the additional risk of a poor outcome attributable to
vancomycin resistance in enterococci has been challenging, largely because of the
confounding effects of comorbidity (51). Most studies have demonstrated an associa-
tion of VRE infection with excess mortality, the duration of hospital admission, and
treatment costs (52, 53), especially when VRE cause a bloodstream infection (54).
(474), cephalosporins (64), and pelvic infection, E. faecalis CC2 (477), CC9 (477), compared to that from chloramphenicol (483), demonstrates significantly higher levels
multidrug resistant (184) endocarditis (51) ST6 (478), and ST16 (479) bacteremia caused by doxycycline (483), high-dose of resistance than E. faecalis (35);
vancomycin-sensitive bacteria ampicillin and sulbactam commonly encountered as
(473) (483), omadacycline (396) asymptomatic colonization (487)
Methicillin-resistant Aminoglycosides (488), -lactams Acute bacterial skin and skin ST5 (65), ST8 (493), ST22 (35), Greater than 20% for bloodstream Vancomycin (488), clindamycin In Asia, 50% of all S. aureus bloodstream
S. aureus (489), chloramphenicol (488), structure infection (490), ST30 (494), ST59 (495), ST72 infection (76, 77); overall (499), daptomycin (500), infections are caused by MRSA (507); in
trimethoprim (313), bacteremia (488), pneumonia (CC8) (496), ST80 (70), ST398 mortality ranges from 15–50% linezolid (501), tedizolid (502), USA, HA-MRSA infections have
macrolides (313), tetracycline (491), osteoarticular infection (livestock associated) (71, 497) (498) dalbavancin (503), tigecycline decreased by 54% (508); in Europe, the
(313), fluoroquinolones (64), (492), endocarditis (488) (504), trimethoprim and total proportion of reported MRSA
multidrug resistant (488) sulfamethoxazole (505), infections among S. aureus infections
pristinamycin (506), decreased from 19.6% in 2014 to 16.4%
cmr.asm.org 4
Clinical Microbiology Reviews
Antimicrobial Resistance Profiles of ESKAPE Pathogens Clinical Microbiology Reviews
incidence of hospital-acquired MRSA (HA-MRSA) is declining (4, 56) (Table 1). Opposite
this finding, the incidence of community-acquired MRSA (CA-MRSA) infections in the
same region has significantly increased (56). CA-MRSA infections emerged among the
indigenous population of Australia in the 1980s (57) and in otherwise healthy commu-
nities of the United States and Canada in the 1990s (58). In North America, where
CA-MRSA is prevalent, MRSA epidemics are largely attributed to the emergence of
either of two unrelated MRSA clones (59, 60). The MRSA clone USA400, isolated from
the pediatric population, initiated the first epidemic wave and remains a common
cause of community-onset disease among indigenous populations in Alaska and the
Pacific Northwest (61). Since 2001, USA400 has been superseded by an epidemic
caused by MRSA USA300 (61, 62) and closely related variants, which are now the most
prevalent CA-MRSA isolates in North America and northern parts of South America.
Today, the burden of MRSA across the world varies substantially (4, 63, 64). In China,
the prevalence of HA- and CA-MRSA infections wavered remarkably between 2007 and
2018. The prevalence of HA-MRSA clones ST239-t030 and ST239-t037 was significantly
reduced (from 20.3% to 1% and 18.4% to 0.5%, respectively), and these have now been
replaced by the ST5-t2460 clone (from 0% to 17.3%), which has seen a rapid emer-
gence. Furthermore, the incidence of CA-MRSA clones ST59 and ST398 also increased
over the same period (from 1.0% to 5.8% and 1.8% to 10.5%, respectively) (65). In
Northern Europe (i.e., the United Kingdom and France), a steady decrease in the
prevalence of HA-MRSA was observed between 2015 and 2018 and was largely
attributed to improved national infection control programs (64, 66, 67). In comparison,
the rates of HA-MRSA in Southern Europe (i.e., Portugal, Spain, Italy, and Greece) remain
high (5, 64).
CA-MRSA strains have typically been associated with skin and soft tissue infections,
whereas HA-MRSA strains are associated with severe pneumonia and bloodstream
infections (68). The division between CA- and HA-MRSA strains is becoming indistinct,
with CA-MRSA strains now identified to be a causative agent of bloodstream infections
in nosocomial settings. MRSA ST80 is a well-defined agent of CA-MRSA in Europe.
Although it is now becoming less prevalent in select European countries (69), CA-MRSA
ST80 is now a major contributor of infection in defined health care settings (70).
Furthermore, examples of CA-MRSA (e.g., ST398) have been shown to be associated
with exposure to livestock (particularly pigs) in Europe (71) (Table 1). Although indi-
viduals with direct exposure to livestock are the most at risk from livestock-associated
MRSA (LA-MRSA), it has now been reported that LA-MRSA substantially contributes to
the burden of nosocomial infection in Europe (72). One of the less-defined and
neglected subgroups of S. aureus is borderline oxacillin-resistant S. aureus (BORSA).
Found both in community settings and in hospital settings, BORSA is characterized by
intermediate resistance to penicillinase-resistant penicillins, with oxacillin MICs being
between 1 and 8 g/ml (73). Lacking the mecA gene, BORSA is not truly either
methicillin resistant or methicillin sensitive, and frequent misidentification poses a
significant threat to patient treatment and outcome, as severe BORSA infections may be
nonresponsive to high doses of oxacillin (74). Overall, MRSA infections carry additional
health care burdens in terms of morbidity, length of hospital stay, health care costs, and
quality of life (75). The rate of mortality following S. aureus bloodstream infection
exceeds 20%, and the presence of methicillin resistance is independently associated
with increased mortality (76, 77).
KLEBSIELLA PNEUMONIAE
Cephalosporin- and carbapenem-class antibiotics have been a mainstay of treat-
ment for serious infections caused by Enterobacterales, such as K. pneumoniae, but
efficacy has been compromised by the widespread acquisition of genes encoding
enzymes, such as extended-spectrum -lactamases (ESBLs) and carbapenemases,
which mediate the respective resistance to these critical drugs (19). High rates of
mortality, often exceeding 40%, have been associated with severe infections caused by
carbapenem-resistant Enterobacterales (CRE) (78). Effective antimicrobial options are
often lacking, and treatment typically requires reliance on drugs with a risk of toxicity
(e.g., aminoglycosides, polymyxins) or other safety concerns (e.g., tigecycline) (79)
(Table 1). Carbapenem-resistant K. pneumoniae (CRKP) strains are the most clinically
prominent CRE (64, 80). In the United States, carbapenemases carried by K. pneumoniae
were originally reported in 2001 (81). Since then, the genes encoding these
-lactamases have spread among several Gram-negative bacterial species. Between
2005 and 2010, an increase in CRKP isolates causing invasive infections was reported
across Europe (64). The spread of CRKP in Europe has been driven by direct and indirect
patient-to-patient transmission in nosocomial settings, largely attributed to ST11, ST15,
ST101, and ST258 strains, along with the ST258 derivative ST512 (82) (Table 1). The
global burden of CRKP has now been further exacerbated by successive waves of CRKP
emerging from several locations across the Indian Ocean rim, the United States, and
China (83–87). The global dissemination of CRKP is exemplified by the CRKP clone
ST307. The ST307 clone has successfully disseminated across every major continent
(88), demonstrating extremely high transmission rates in health care settings (89).
Recent reports suggest that AMR hypervirulent K. pneumoniae (hvKP) strains are also
emerging. In Taiwan, hvKP causes as many cases of necrotizing fasciitis as Streptococcus
pyogenes and is associated with a higher mortality rate (47% versus 19%) (90). The
detection of hvKP is now being reported around the world in both high- and low-
income settings (87, 91, 92). An important laboratory feature frequently seen in hvKP
strains is the presence of a hypermucoviscous phenotype (in association with the K1
and K2 capsular serotypes) (93).
ACINETOBACTER BAUMANNII
A. baumannii infections typically occur in hospitalized patients or patients with
significant contact with the health care system (94). Historically, A. baumannii has been
associated with hot and/or humid geographic climates (95, 96). Between 1987 and
1996, the frequency of both community- and hospital-acquired infections across the
United States was observed to rise by 50% between the months of July and October
(97). Since the 1970s, A. baumannii has become increasingly common in temperate
climates, a shift largely attributed to improved environmental persistence mechanisms
and MDR development (98). Community-acquired pneumonia due to A. baumannii has
been described in tropical regions of Asia and Australia among individuals with a
history of alcohol abuse (99). Although A. baumannii infection rates are comparatively
low compared to those of other ESKAPE pathogens (100, 101), approximately 45% of all
global A. baumannii isolates are considered MDR, with rates exceeding 60% in the
United States (4, 101), Latin America, and the Middle East (102). Turkey and Greece have
reported MDR rates exceeding 90% (103). These levels of MDR for A. baumannii are over
four times higher than those observed in K. pneumoniae and P. aeruginosa (3). A key
aspect of A. baumannii physiology is the propensity to develop rapid resistance. From
2011 to 2016, the rate of identification of A. baumannii isolates resistant to
carbapenem- and -lactam-class antibiotics has increased by over 30% globally (103).
The spread of MDR and carbapenem-resistant A. baumannii (CRAB) isolates is largely
associated with three international clonal lineages: CC1, CC2, and CC3 (104, 105). CC1
is prevalent worldwide, while CC2 and CC3 are highly prevalent in Europe and North
America. CC15 and CC79 are also predominant in Central and South America (106, 107).
With the emergence of pandrug-resistant isolates, last-resort carbapenem- and
polymyxin-class antibiotics are no longer effective (103, 108) (Table 1). Without ade-
quate action via improved epidemiological surveillance and therapeutic development,
A. baumannii has the capacity to potentiate a global epidemic.
PSEUDOMONAS AERUGINOSA
Widely present in aquatic environments, P. aeruginosa is a Gram-negative opportu-
nistic human pathogen commonly associated with severe respiratory infections in
patients with impaired immunity. While P. aeruginosa is responsible for 10% of all
ENTEROBACTER SPECIES
Over the last 35 years, Enterobacter aerogenes (now renamed Klebsiella aerogenes)
and Enterobacter cloacae species have presented as significant threats to neonatal
wards and patients in intensive care units, particularly those dependent on mechanical
ventilation (119). The emergence of these two Enterobacter species as clinically signif-
icant MDR pathogens has occurred in concurrent epidemic waves. From the early 1990s
to 2003, E. aerogenes was the most clinically prevalent cause of Enterobacter nosocomial
infection (119). During this period, the hospital-acquired E. aerogenes infection inci-
dence was high in Western Europe (120, 121), largely attributed to the dispersion of a
single epidemic clone (122, 123). In about 2010, E. aerogenes was superseded by E.
cloacae as the most common clinically isolated species of the genus (124). It is worth
noting that other members of the E. cloacae complex, especially Enterococcus hormae-
chei, are clinically relevant and are often difficult to discriminate at the species level
based on standard phenotypic assays (125, 126).
MDR Enterobacter species are an increasing cause of hospital-acquired infection. In
the United States, E. aerogenes ST4 and ST93 currently represent prevalent lineages
associated with nosocomial infection (127). For the E. cloacae complex, recent data
suggest that carbapenem resistance has directionally spread across the United States
due to the dissemination of hospital-associated carbapenem-resistant E. cloacae ST178
and ST78 isolates (128). Prior to 2005, an estimated 99.9% of Enterobacter strains were
sensitive to carbapenems (129). Carbapenem resistance is now reported in all WHO
health regions (3). Moreover, pandrug-resistant E. aerogenes has also emerged, display-
ing resistance to the last-resort antibiotic colistin (130) (Table 1). To complicate the
treatment of bacterial infections further, E. aerogenes is capable of harboring subpopu-
lations of colistin-resistant bacteria which are undetectable using current diagnostic
testing strategies (131).
ESCHERICHIA COLI
Although not formally recognized as part of the ESKAPE group of pathogens, AMR
Escherichia coli is identified as a major cause of bloodstream and urinary tract infection
(UTI) in both community and health care settings globally (5, 35, 64). Sepsis is one of
the most common manifestations of E. coli UTI. In Australian inpatient and emergency
department settings, E. coli is the most prevalent Gram-negative bacterial species
isolated from both blood and urine cultures (35). Over the past decade, several
pandemic clones of MDR uropathogenic E. coli (e.g., ST131 and ST95) have dissemi-
nated worldwide (132, 133). Through horizontal gene transfer, E. coli typically acquires
resistance genes from other members of the Enterobacterales. High rates of resistance
to aminopenicillins, fluoroquinolones, aminoglycosides, and third-generation cephalo-
sporins are noted across Europe (64). Although carbapenem resistance is rare in
invasive E. coli strains, the general situation in Europe for CRE, including E. coli, was
shown to worsen between 2010 and 2018 (134). Furthermore, in 2016, resistance to the
last-resort polymyxin, colistin, was identified in E. coli strains isolated from pig farms in
China (135). Although not discussed further in this review, AMR E. coli is currently one
of the largest clinical burdens facing both human and animal health. In order not to
exacerbate these challenges further, organizations involved in AMR policy, research and
development (R&D), and surveillance need to consider this pathogen as a critical public
health concern.
Antibiotic Inactivation/Alteration
One of the most common AMR mechanisms employed by ESKAPE pathogens
involves the production of enzymes that irreversibly destroy or neutralize antibiotics.
Such enzymes are particularly prevalent among the Gram-negative pathogens and
comprise those (i) that destroy the active antibiotic site (e.g., hydrolytic cleavage of the
-lactam ring by -lactamases) or (ii) that covalently modify key structural elements of
the drug to hinder bacterial target site interaction (e.g., aminoglycoside-modifying
enzymes [AMEs] that catalyze hydroxyl/amino group modifications).
-Lactamases. -Lactamase enzymes were first identified soon after the initial
discovery and purification of penicillin (136). Since then, ⬎2,600 unique -lactamases
enabling resistance to one or more -lactams (i.e., penicillins, cephalosporins, mono-
bactams, and carbapenems) have been described (137). -Lactamases remain the most
important resistance mechanism among Gram-negative ESKAPE pathogens, where they
are concentrated within the periplasm, thus hydrolyzing the -lactam agents prior to
reaching the penicillin-binding protein (PBP) target in the cell wall.
-Lactamase enzymes are typically classified according to their primary molecular
structure (i.e., the Ambler scheme [138]) or combined hydrolytic and inhibition func-
tional properties (i.e., the Bush-Jacoby system [139]). Ambler class A enzymes contain
serine in their active site and comprise penicillinases, cephalosporinases, narrow- and
broad-spectrum -lactamases, extended-spectrum -lactamases (ESBLs), and carbap-
enemases. Overall, they represent the largest cluster of -lactamase enzymes and
collectively are capable of inactivating most -lactam classes, including the penicillins,
early cephalosporins, third-generation oxyimino-cephalosporins, monobactams, cepha-
mycins, and carbapenems. Their susceptibility to inhibition by clavulanic acid and
tazobactam is variable, though all are inhibited by novel -lactamase inhibitor agents,
including avibactam, relebactam, and vaborbactam (139, 140).
Ambler class A enzymes comprise various important -lactamases that are fre-
quently observed in Gram-negative (e.g., TEM, SHV, CTX-M, and KPC) and Gram-positive
FIG 1 Mediators of ESKAPE pathogen antimicrobial resistance. Mechanisms facilitating antimicrobial resistance in ESKAPE pathogens can be broadly categorized
into four groups: (i) enzyme-mediated antimicrobial inactivation, which either irreversibly destroys the active antibiotic site (e.g., hydrolytic cleavage of the
-lactam ring by -lactamases) or covalently modifies key structural elements of the drug to hinder the bacterial target site interaction (e.g., aminoglycoside-
modifying enzymes that catalyze hydroxyl/amino group modifications); (ii) bacterial target site modification, which prevents the binding or which reduces the
affinity of the antibiotic molecule at the cell surface (e.g., LPS modification, PBP2a expression with reduced -lactam affinity, and van gene cluster-mediated
peptidoglycan modification) or intracellularly (e.g., 16S RNA methylation); (iii) reduced antibiotic accumulation through the mutation or loss of outer membrane
channels (e.g., OprD in P. aeruginosa, CarO in A. baumannii, and OmpK36 in K. pneumoniae) and expression of efflux systems to actively extrude drugs out of
the cell (e.g., RND, MFS, MATE, SMR, ABC, and PACE); and (iv) persistence through biofilm-embedded cells which demonstrate a markedly higher tolerance to
antimicrobial agents than planktonic bacteria. AMEs, aminoglycoside-modifying enzymes; AACs, aminoglycoside acetyltransferases; ANTs, aminoglycoside
nucleotidyltransferases; APHs, aminoglycoside phosphotransferases; LPS, lipopolysaccharide; PBP, penicillin-binding protein; RND, resistance-nodulation-
division; MFS, major facilitator superfamily; MATE, multidrug and toxic compound extrusion; SMR, small multidrug resistance; ABC, ATP-binding cassette; PACE,
proteobacterial antimicrobial compound efflux; EPS, extracellular polymeric substance.
enzymes remain susceptible to clavulanic acid, though Bush-Jacoby subgroup 2br and
2ber ESBLs (e.g., TEM-30, SHV-10, and TEM-50) show reduced susceptibility to various
-lactamase inhibitors (146). Concerningly, inhibitor-resistant -lactamases have also
been reported in K. pneumoniae strains harboring KPC serine carbapenemase enzymes
(147). Plasmid-encoded KPCs have been associated with major outbreaks worldwide (e.g.,
the outbreak caused by K. pneumoniae ST258) and hydrolyze virtually all -lactams,
including carbapenems (148). Despite this, there is emerging evidence that infections with
KPC-producing organisms can be successfully targeted with various new -lactamase–-
lactamase inhibitor combinations, including imipenem-cilastatin-relebactam, meropenem-
vaborbactam, and ceftazidime-avibactam (149). Unfortunately, the rapid evolution of
ceftazidime-avibactam resistance has already been reported in K. pneumoniae ST258
blaKPC-3-harboring isolates and in non-ST258 clonal backgrounds and additional blaKPC
variants (17, 150, 151).
Ambler class B metallo--lactamases (MBLs) represent another clinically important
group of enzymes capable of hydrolyzing most -lactams, including carbapenems.
However, in contrast to other -lactamases, they require Zn2⫹ at their active site,
display a low affinity for aztreonam, and are inhibited by EDTA (139). The most
prominent MBLs encountered in the Gram-negative ESKAPE pathogens (e.g., MBLs of
the IMP, VIM, and NDM families) are encoded on conjugative plasmids. IMP- and
VIM-type MBLs were first detected in clinical P. aeruginosa isolates (152, 153) but have
since been identified in K. pneumoniae, E. cloacae complex isolates, and Acinetobacter
spp. (154–157). NDM-type enzymes have also been detected across all Gram-negative
ESKAPE bacteria and are of particular concern due to the fact that they are incorporated
into transferable genetic elements that also encode determinants for resistance to
other antibiotic classes (157, 158).
Group C -lactamases comprise chromosomally encoded cephalosporinases, such
as AmpC, that are found in many Enterobacterales (including Enterobacter spp.), P.
aeruginosa, and Acinetobacter spp. (159). They are most active on narrow- to
intermediate-spectrum cephalosporins plus aztreonam and are usually resistant to
clavulanic acid. The rate of constitutive expression of AmpC is usually low, but clinically
relevant resistance is inducible during therapy (139). Plasmid-mediated resistance
involving group C enzymes has also been reported widely, including reports of plas-
mids in organisms, such as K. pneumoniae, that do not normally contain genes
encoding these enzymes on their chromosome (159).
-Lactamases belonging to Ambler class D primarily consist of oxacillin-hydrolyzing
enzymes (OXA), which are able to hydrolyze oxacillin and its derivatives, which display
ESBL-like substrate properties, and which show variable resistance to -lactam inhibi-
tors (139). Importantly, some OXA-type -lactamases, such as OXA-48 and its deriva-
tives, also confer carbapenem resistance. OXA-type enzymes are most frequently found
in Acinetobacter spp., where they are often located on the chromosome. However,
plasmid-borne OXA-48-like enzymes are now widely distributed in many Enterobacte-
rales species, including K. pneumoniae and Enterobacter spp. (160), many of which
express other ESBLs, such as CTX-M-15, and thus provide resistance to most -lactam
agents (161).
Aminoglycoside-modifying enzymes. The most common aminoglycoside resis-
tance mechanism encountered among ESKAPE pathogens occurs through the produc-
tion of AMEs. During transportation of the drug across the cytoplasmic membrane,
these enzymes covalently catalyze specific hydroxyl or amino group modifications of
the aminoglycoside molecule, thus reducing antibacterial activity through diminished
bacterial ribosomal subunit binding. Based on their biochemical activity, there are three
classes of AMEs (i.e., aminoglycoside acetyltransferases [AACs], aminoglycoside phos-
photransferases [APHs], and aminoglycoside nucleotidyltransferases [ANTs]). Enzymes
within each class are then further subdivided according to the position of the modifi-
cation site, resistance profile, and specific protein designation (162). Earlier work has
shown that the global distribution of AMEs varies with respect to geography, antibiotic
selection pressure, and bacterial species (163, 164). Depending on the specific enzyme
and the host organism, genes coding for AMEs are located on plasmids, on transposons,
or in the chromosome (162), though the high frequency of these resistance determi-
nants among ESKAPE pathogens is largely attributable to acquisition via horizontal
gene transfer (165).
AACs encompass the largest AME class and in an acetyl coenzyme A-dependent
manner catalyze the acetylation of specific amino groups present on the antibiotic
acceptor molecule. Of the four AAC subclasses, the AAC(1) and AAC(3) enzymes target
amino group positions 1 and 3 of the central 2-deoxystreptamine ring, respectively,
whereas the AAC(2=) and AAC(6=) subclasses modify the respective 2= and 6= amino
group positions of the 2,6-dideoxy-2,6-diamino-glucose ring (166). While comprehen-
sive analyses of global AAC epidemiology remain relatively scarce, recent investigations
conducted in the United States, Europe, and Asia indicate that Gram-negative ESKAPE
pathogens most frequently encode AAC(3) and AAC(6=) enzymes, which collectively
confer resistance to gentamicin, tobramycin, and amikacin (165, 167, 168).
APHs comprise the second most abundant class of AMEs, which decrease aminogly-
coside binding affinity by catalyzing ATP-dependent phosphorylation of —OH groups
on the antibiotic molecule. Of the seven different APH subclasses [i.e., APH(4), APH(6),
APH(9), APH(3=), APH(2⬙), APH(3⬙), and APH(7⬙)], APH(3=) is the most widely distributed
among clinical isolates, with the aph(3=)-IIIa gene being recognized as a key determi-
nant of plasmid-mediated amikacin resistance in both S. aureus and Enterococcus spp.
(165).
The final class of AMEs encompasses the ANTs, which reduce aminoglycoside
toxicity via the magnesium-dependent transfer of a nucleoside monophosphate
to —OH groups on the antibiotic molecule. Overall, there are five subclasses of ANTs
[i.e., ANT(6), ANT(9), ANT(4=), ANT(2⬙), and ANT(3⬙)], of which ANT(4=) and ANT(2⬙) are
the most clinically relevant. ANT(4=) enzymes conferring resistance to amikacin and
tobramycin have been detected in S. aureus, Enterococcus spp., K. pneumoniae, and P.
aeruginosa. ANT(2⬙), encoded by the ant(2⬙)-Ia (or aadB) gene, is frequently associated
with gentamicin and tobramycin resistance across all the Gram-negative ESKAPE
organisms (165).
Most importantly, broad-spectrum aminoglycoside resistance in the ESKAPE patho-
gens is often conferred through the presence of multiple or bifunctional AMEs. This
frequently occurs among Gram-negative organisms, where multiple AMEs result in
significantly increased aminoglycoside resistance (169–171). Likewise, expression of the
bifunctional AAC(6=)-APH(2⬙) enzyme, which resides on the common Tn4001 trans-
poson, accounts for high-level gentamicin resistance in both S. aureus and Enterococcus
spp. (including MRSA and VRE strains) worldwide (152). More recently, a variant enzyme
termed AAC(6=)-Ib-cr, which confers low-level plasmid-mediated aminoglycoside and
ciprofloxacin resistance, has been described in K. pneumoniae, Enterobacter spp., A.
baumannii, and P. aeruginosa (172–175).
mission, the vast majority of hVISA and VISA infections arise via in vivo evolution within
individual patients and typically involve pandemic HA-MRSA lineages (e.g., ST239 and
ST5). However, it should be noted that CA-MRSA clones, including the USA300 clone,
can also exhibit this resistance phenotype (146, 226).
Resistance to daptomycin, an agent that has activity against Gram-positive bacteria
and that is related to host cationic antimicrobial peptides (AMPs), has also been
observed in both S. aureus and enterococci in recent years. The precise mechanisms of
resistance are yet to be fully elucidated, but it has been postulated that alterations in
cell surface charge, phospholipid composition/metabolism, and membrane stress re-
sponses are involved (227). Recent studies also highlight the emergence of acquired
polymyxin (another cationic AMP) resistance in K. pneumoniae, A. baumannii, and P.
aeruginosa arising from remodeling of outer membrane (OM) lipopolysaccharide (LPS)
lipid A structures. These modifications contribute to reduce the net negative charge of
the LPS, thus reducing its polymyxin binding efficiency. In K. pneumoniae, loss-of-
function mutations of the mgrB gene (a negative feedback regulator of the PhoPQ
TCRS), mutations driving the expression of the PhoPQ, PmrAB, and CrrAB TCRS, as well
as the acquisition of the plasmid-mediated mcr gene all give rise to resistance-
associated lipid A modifications (e.g., addition of 4-amino-4-deoxy-L-arabinose [Ara4N],
phosphoethanolamine [PEtN], and 2-hydroxymyristate through increased expression of
the pmrHFIJKLM operon, pmrC, and lpxO, respectively) (135, 228–230). Of these, mgrB
inactivation has been reported the most frequently and, interestingly, also gives rise to
other modifications that collectively promote virulence and that attenuate early host
defense responses (228). The primary mechanisms of polymyxin resistance in A. bau-
mannii comprise mutations in the PmrAB TCRS leading to PEtN synthesis and the loss
of LPS through inactivation of the lpxA, lpxC, and lpxD lipid A biosynthesis genes (229).
Polymyxin resistance in P. aeruginosa is conveyed by five TCRS, including PmrAB,
PhoPQ, ParRS, ColRS, and CpsRS, most frequently resulting in the constitutive expres-
sion of pmrHFIJKLM and the addition of Ara4N (229).
terial antimicrobial compound efflux (PACE) families (234, 237). All six families are
represented within the ESKAPE group, with individual exporters varying in terms of
their substrate specificity. Of note, RND-type efflux pump-mediated resistance is of
particular concern with respect to AMR among Gram-negative bacteria. For example,
the chromosomally encoded MexAB-OprM efflux system in P. aeruginosa exhibits broad
substrate specificity and when overexpressed confers fluoroquinolone, aminoglycoside,
and -lactam resistance. Likewise, the overproduction of AcrAB-TolC is characteristic of
multidrug-resistant K. pneumoniae and Enterobacter strains. The A. baumannii AdeABC,
AdeFGH, and AdeIJK RND-type efflux pumps are also associated with broad-range AMR
(234, 238–240). More recently, the chromosomally encoded OqxAB efflux pump, which
contributes to reduced quinolone and chloramphenicol susceptibility, has been iden-
tified in K. pneumoniae (195, 241). OqxAB homologues have also been observed in
some Enterobacter spp., though, aside from tigecycline (242), these elements are not
thought to contribute to clinically relevant drug resistance under in vitro conditions
(241).
Methicillin-resistant S. aureus -Lactams Tn Tn552-like elements Tn552 is a complex unit transposon responsible for mobilization and 272
dissemination of -lactam resistance in staphylococci; associated
with a pSK1-like and pSK41-like plasmid
Aminoglycosides, -lactams, vancomycin Plasmid pSK41-like plasmids (e.g., pSK41, Conjugative; associated with carriage of the Tn552-like -lactam 318–321
pGO1, and pLW103) resistance transposons; associated with carriage of vanA on the
Tn1546 glycopeptide resistance transposon
Methicillin, penicillin, -lactams GI SCCmec element SCCmec has a limited distribution and is restricted to 11 major 336–338
clonal lineages of S. aureus from 5 clonal complexes
Methicillin, glycopeptides IS IS1182, IS256 Insertional deactivation, resulting in increased resistance 566, 567
Enterobacterales (K. pneumoniae Multidrug: -lactams, carbapenems, IS ISEcp1, ISCR Typically encodes a promiscuous transposase which can mobilize 273–276, 280,
and Enterobacter spp.) trimethoprim, chloramphenicol, adjacent genes when they fail to identify terminal repeat 568, 569
fluoroquinolones sequences; ISEcp1 elements are commonly associated with
mobilization of -lactamase genes; ISCR elements are commonly
associated with complex class 1 integrons
Carbapenems, colistin IS Various IS, including ISEcp1 Insertional deactivation resulting in increased resistance 276, 281, 282,
570
Multidrug: -lactams, aminoglycosides, Plasmid IncF-type, IncI, IncH (HiI1 and HI2), Typically, low-copy-number plasmids; broad and narrow host 293, 294, 571
trimethoprim, antiseptics, carbapenems, IncL, IncC, IncN, IncH, IncX3 ranges; can act as vehicles for carriage of other mobile resistance
colistin, cephalosporins elements, e.g., transposons and integrons
A. baumannii Multidrug: -lactams, aminoglycosides Tn AbaR, AbGR11 Tn7-like unit transposons 270, 271
chloramphenicol, tetracycline, sulfonamide
Carbapenems Tn Tn2006 The Tn2006 composite transposon (ISAb1) is responsible for
mobilization of the blaOXA-23 carbapenemase
Carbapenems IS ISAb825, ISAb125 Insertional deactivation of the outer membrane protein carO results 223
in elevated carbapenem MICs
Carbapenems IS ISAba1 IS insertion upstream of blaOXA-53 drives expression of the gene; 283
IS-mediated constitutive expression of blaOXA-53 confers high-
level carbapenem resistance
Gentamicin, tobramycin Plasmid pRAY-like plasmids Small (6- to 10-kb), widely distributed plasmids 303, 304
Multidrug: carbapenems and kanamycin Plasmid RepAci6-like plasmids (e.g., pAB-G7-2 Kanamycin resistance is associated with carriage of the TnaphA6 302, 305, 306
and pACICU2), pNDM-BJ01-like transposon; carbapenem resistance is associated with carriage of
plasmids the Tn2006 transposon; pNDM-BJ01-like plasmids from A. lwoffii
carry the blaNDM-1 carbapenemase genes
P. aeruginosa Carbapenems, -lactams IS IS21, ISPA26 Insertional deactivation, resulting in increased resistance 572, 573
Carbapenems Plasmid IncP-2 plasmids Carbapenemases associated with class 1 integrons carried on 309–311
IncP-2-type plasmids
Carbapenems, aminoglycosides ICE P. aeruginosa pathogenicity island Multidrug resistance is associated with carriage of antimicrobial resistance 333
(e.g., PAPI-1, PAGI-2/PAGI-3-like) genes on Tn6162 and Tn6163 elements in a genomic island
aAbbreviations:MGEs, mobile genetic elements; ICE, integrative and conjugative elements; Tn, transposon; IS, insertion sequence; GI, genomic island; MLS, macrolide lincosamide sulfonamides; SCCmec, staphylococcal
cassette chromosome mec.
cmr.asm.org 17
Clinical Microbiology Reviews
De Oliveira et al. Clinical Microbiology Reviews
emase gene (273–276). ISEcp1 elements encode promiscuous transposases that can
recognize a variety of different sequences as right IRs (IRr), thereby allowing them to
capture adjacent genes. Related IS (IS1247, ISKpn23, and ISEnca1) have also been
associated with the mobilization of adjacent resistance genes in a manner similar to
that for ISEcp1 (277, 278). Recently, it has been demonstrated that IS26 can produce a
circular intermediate consisting of a single copy of IS26 and a DNA segment immedi-
ately adjacent to it. This structure, termed a translocatable unit (TU), can then move by
a replicative mechanism (279). ISCR elements also form circular intermediates. They
move and can capture adjacent genes by rolling-circle replication (280).
IS elements can also impact the evolution of AMR in the host by the transpositional
deactivation of genes and by modulating the expression of adjacent genes through the
delivery of promoter or terminator sequences (reviewed in reference 264). The inser-
tional deactivation of uptake systems is a common mechanism by which IS elements
can affect antibiotic susceptibilities. For example, IS-mediated deactivation of the
ompK36 porin in K. pneumoniae results in elevated carbapenem MICs (281). Similarly,
insertional inactivation of the mgrB regulatory gene in K. pneumoniae drives the
overexpression of the pmrHFIJKLM operon, conferring colistin resistance (276, 282).
Many IS carry strong promoter sequences, and their insertion upstream of chromo-
somal genes can drive the expression of that gene and influence AMR. This mechanism
is clearly evident in A. baumannii, where insertion of an ISAba1 element upstream of the
blaOXA-51 gene confers carbapenem resistance (283). Similar mechanisms of IS-
mediated constitutive expression of resistance genes have been observed in K. pneu-
moniae and P. aeruginosa (284, 285). Alternatively, an IS may provide only the ⫺35
region promoter component, which, together with a ⫺10 region donated by an
adjacent gene, forms a hybrid promoter to drive the expression of neighboring genes.
Hybrid IS promoters have been identified in at least 17 different bacterial species (286),
including A. baumannii, K. pneumoniae, and P. aeruginosa (283, 287, 288).
Plasmids
Plasmids are an important vehicle for gene transfer in both Gram-negative and
Gram-positive bacteria (289). Typically, plasmids are circular, double-stranded, and
self-replicating DNA molecules that are readily vertically inherited in a growing popu-
lation (290). While it is clear that the MGEs described thus far (IS elements, transposons,
etc.) are primarily responsible for mobilizing resistance genes, the dissemination of
these genes is mainly attributed to conjugative plasmids. Plasmids are rich in IS and
other MGEs carrying AMR genes and facilitate the intra- and interspecies horizontal
transfer of these elements (135, 291–293).
MDR Enterobacterales (K. pneumoniae and Enterobacter species) carry plasmids from
a wide variety of different incompatibility (Inc) groups (294) (Table 2), but those from
Inc group types F (multiple F-type replicons can be found together in multireplicon
plasmids), I, H (HI1 and HI2), L, C, and N are most frequently associated with multidrug
resistance (293, 294). Of particular concern among the Enterobacterales is the role that
these plasmids continue to play in the emergence and dissemination of ESBLs, partic-
ularly those of the blaCTX-M type (19, 295, 296); AmpC-type cephalosporinases (blaCMY-2
and blaDHA-1) (297–299); carbapenemase-encoding genes (blaVIM, blaKPC, blaNDM, and
blaOXA-48) (161, 300, 301); and plasmid-mediated colistin resistance (mcr) (229). Com-
paratively little is known about plasmids from A. baumannii compared to what is known
about plasmids from the Enterobacterales. However, in 2011, a Europe-wide study of
clinical A. baumannii isolates found that resistance plasmids were mainly associated
with carriage of the blaOXA (blaOXA-23, blaOXA-58-like, and blaOXA-40) carbapenemase
genes (302), kanamycin and amikacin resistance, and gentamicin and tobramycin
resistance (303–306). Additionally, plasmids related to pNDM-BJ01 from Acinetobacter
lwoffii carry the globally distributed blaNDM-1 carbapenemase gene (307).
In P. aeruginosa, resistance genes are typically found on chromosomal resistance
islands rather than plasmids. However, P. aeruginosa may carry large (⬃300- to 500-kb),
transferable IncP-2 plasmids (308) associated with the carriage of carbapenemase
genes, specifically, blaIMP (blaIMP-9 and blaIMP-45) (309, 310) and blaVIM (blaVIM-2) (311)
carbapenemases, on class 1 integrons.
AMR plasmids are frequently found in clinical staphylococcal isolates (312, 313).
These include small (1- to 10-kb) multicopy plasmids typically encoding a single
resistance gene (314–316) and larger (⬎15-kb) multiresistance plasmids, such as the
prototype pSK1 family of multiresistance plasmids (317) (Table 2). A single family of
larger (⬎30-kb) conjugative multiresistance plasmids, including pSK41, pGO1, and
pLW1043 (318, 319), can also be found in clinical strains of staphylococci and are
credited with the emergence of aminoglycoside, -lactam, and vancomycin resistance
in S. aureus populations (267, 320–326).
In the enterococci, AMR is largely encoded on Inc18 and RepA_N plasmids (262).
Both Inc18 and RepA_N plasmids have a broad host range, allowing for their transfer
into a variety of bacterial species, and are responsible for introducing vancomycin
resistance into MRSA (218).
bacterial species (352). Consequently, the potential role that transformation plays in the
spread of AMR genes cannot be overlooked.
been estimated to be €31.5 million, which in turn considerably weakens the economic
incentive (375). Today, the majority of pharmaceutical companies which do undertake
R&D have few antimicrobial products on the market aimed at the WHO’s priority
pathogen list (376). Due to the absence of revenue from sales, these companies are
largely dependent on external partnering funding sources, such as the U.S. Biomedical
Advanced Research and Development Authority (BARDA), Combating Antibiotic Resis-
tant Bacteria Biopharmaceutical Accelerator (CARB-X), the Wellcome Trust, the National
Institute of Allergy and Infectious Diseases (NIAID), and the Innovative Medicines
Initiative (IMI; a joint undertaking between the European Union and the European
Pharmaceutical Industry), initiatives funded solely and/or in partnership (376–379).
Identifying antibiotic-related health priorities, defining appropriate stewardship prac-
tices, and developing new sustainable economic models to stimulate antibiotic inno-
vation are key intersecting themes currently requiring a cooperative restructure.
Funded by IMI, Driving Reinvestment in Research and Development and Responsible
Antibiotic Use (DRIVE-AB) is a pioneering project composed of 15 public and 7 private
partners from 12 countries aiming to tackle this issue (380). Through the use of
surveillance systems data, antibiotic prescription databases, and published literature,
DRIVE-AB aims to develop models which transform the way in which policy makers
stimulate and financially incentivize antibiotic innovation (380, 381). Despite collabor-
ative projects like these, the majority of new R&D programs aimed at tackling AMR are
currently funded by public and nonprofit partnerships (374). This approach often
neglects obstacles, such as licensing, affordability, and stewardship, which affect access
to new antimicrobials in low- and middle-income countries. Even though antibiotic
drug discovery has reduced and investment from the pharmaceutical industry has
receded, promising antimicrobial strategies are still being developed (Table 3) (371,
382–389).
(cephalosporin) 2019
Imipenem ⫹ cilastatin ⫹ -Lactam (carbapenem) ⫹ PBP, -lactamase U.S. FDA approved on 17 July 2019; Merck & Co., Inc. CRE and CRPA cUTI including
relebactam (Recarbrio) -lactamase inhibitor E.U. EMA approved on 13 pyelonephritis,
(diazabicyclooctane) February 2020 cIAI, and HA and
VA bacterial
pneumonia
Meropenem- vaborbactam -Lactam (carbapenem) ⫹ PBP, -lactamase U.S. FDA approved on 30 August Rempex Pharmaceuticals E. coli, K. pneumoniae, E. cloacae cUTI including Anaphylactic and central
(Vabomere/Vaborem) -lactamase inhibitor 2017; E.U. EMA approved on 20 Inc. (wholly owned species complex, and CRE pyelonephritis nervous system
(cyclic boronate) November 2018 subsidiary of The reactions have been
Medicines Co.) reported
Cefepime ⫹ zidebactam -Lactam (cephalosporin) ⫹ PBP, -lactamase Phase I Wockhardt Ltd. ESKAPE pathogens, pneumococci, cUTI and HA and VA Highly broad-spectrum
cmr.asm.org 22
Clinical Microbiology Reviews
TABLE 3 (Continued)
Drug name Drug class Drug target Development stage Company Pathogen target Applicationb Information of noteb
Cefilavancin (TD-1792) Glycopeptide–-lactam PG chain elongation, Phase III R-Pharm/Theravance ABSSSI
(cephalosporin) hybrid PBP Biopharma Inc.
Delafloxacin Fluoroquinolone Bacterial type II U.S. FDA approved on 19 June Melinta Therapeutics Inc. ESKAPE pathogens except A. ABSSSI Serious adverse
(Baxdela/Quofenix) topoisomerase 2017; E.U. EMA approved on 16 baumannii reactions, including
December 2019 tendinitis, tendon
rupture, and
neuropathy
Lascufloxacin (Lasvic) Fluoroquinolone Bacterial type II Japanese PMDA approved on 20 Kyorin Pharmaceutical Klebsiella spp, Enterobacter spp., CA bacterial Activity against MRSA
topoisomerase September 2019 Co. Ltd. MRSA, and S. pneumoniae pneumonia comparable to that
of linezolid and
vancomycin
Alalevonadifloxacin ⫹ Fluoroquinolone Bacterial type II Phase III Wockhardt Ltd. MRSA, VRSA, and MDR ABSSSI and HA Active against MSRA
levonadifloxacin (WCK topoisomerase pneumococci, E. coli, and K. bacterial biofilm
771/WCK 2349) pneumoniae pneumonia
cmr.asm.org 23
Clinical Microbiology Reviews
TABLE 3 (Continued)
Drug name Drug class Drug target Development stage Company Pathogen target Applicationb Information of noteb
TP-271 Tetracycline 30S subunit of bacterial Phase I Tetraphase Respiratory pathogens CA bacterial Designated a QIDP by
ribosome Pharmaceuticals Inc. pneumonia U.S. FDA and
awarded fast-track
De Oliveira et al.
review
TP-6076 Tetracycline 30S subunit of bacterial Phase I Tetraphase CRE and CRAB Bacterial infections CARB-X funded ($4
ribosome Pharmaceuticals Inc. million)
Lefamulin (Xelenta) Pleuromutilin 50S subunit of bacterial U.S. FDA approved on 19 August Nabriva Therapeutics AG MRSA ABSSSI and CA and Low toxicity profile;
ribosome 2019; E.U. EMA marketing HA bacterial available as both
authorization application pneumonia intravenous and oral
submitted in May 2019 (now treatments
accepted); Chinese NMPA
granted clinical trial application
approval on 13 June 2019
Delpazolid (LCB01-0371) Oxazolidinone 50S subunit of bacterial Phase II LegoChem Biosciences Bacterial infections Granted orphan drug
cmr.asm.org 24
Clinical Microbiology Reviews
TABLE 3 (Continued)
Drug name Drug class Drug target Development stage Company Pathogen target Applicationb Information of noteb
Murepavadin (POL7080) Antimicrobial peptide LptD Phase III Polyphor Ltd. CRPA HA and VA bacterial Designated a QIDP by
mimetic pneumonia U.S. FDA and
cmr.asm.org 25
Clinical Microbiology Reviews
De Oliveira et al. Clinical Microbiology Reviews
Carbapenems are still considered the most effective therapy for select ESKAPE
pathogen infections, particularly those caused by ESBL-producing Enterobacterales.
Unfortunately, the increased use of carbapenems in clinical settings has created a
selection pressure for the emergence of carbapenem resistance. In an effort to define
carbapenem-sparing alternatives, a recent drug trial (MERINO trial) examined whether
piperacillin-tazobactam combinations could demonstrate efficacy comparable to that
of carbapenems (419). Among patients with E. coli and K. pneumoniae bloodstream
infections, 30-day survival did not improve upon piperacillin-tazobactam treatment
compared to that achieved with meropenem therapy (420). Although results from the
MERINO trial did not support the use of piperacillin-tazobactam as a carbapenem-
sparing treatment, the trial highlighted the need for carbapenem-sparing strategies
and for further study into -lactam–-lactamase inhibitor drug combinations.
Reducing the length of hospital stay following the initial stage of infection is
associated with a decreased risk of further complications and better outcomes of the
patient disease state (421, 422). In 2018, with the objective of reducing patient hospital
admission times, the Partial Oral Treatment of Endocarditis (POET) trial evaluated the
efficacy of intravenous antibiotics compared with that of oral antibiotics among stable
patients with infective endocarditis (423). The premise of this study was to allow patient
treatment to take place outside of hospitals, without the requirement for intravenous
catheters. The primary outcome from the POET study showed that oral antibiotics were
noninferior to intravenous antibiotics at preventing adverse events (i.e., all-cause death,
unplanned cardiac surgery, embolic events, or relapse of bacteremia) (423). The impact
of this landmark study may significantly minimize the challenges associated with
parenteral treatment, including logistics, monitoring, and risks of complications asso-
ciated with intravenous catheters (i.e., secondary local and systemic infections).
OUTLOOK
AMR represents one of the few challenges that unites global interests and concerns
for human and animal health and the food and agricultural sectors. Exacerbated by the
acquisition of AMR genes, ESKAPE pathogens represent the paradigm for resistance,
pathogenesis, and disease transmission in both the community and clinical settings.
Although heterogeneous at the genetic level, the general mechanisms surrounding
ESKAPE pathogen emergence and persistence are broadly shared. Mediated in part
through HGT, resistance strategies encompassing drug inactivation, modification of the
antibiotic target site, and a reduction of antibiotic accumulation in the bacterial cell are
common strategies shared by all ESKAPE pathogens. Combined with the collective
ability to form biofilm on innate and biological surfaces, ESKAPE pathogens remain
highly prevalent in clinical settings.
To constrain the spread of ESKAPE pathogens, it is now well recognized that
collaborative global and regional efforts are required by policy makers, funders, and
those responsible for the treatment and management of ESKAPE pathogens (12, 35, 64).
Aside from novel drug development, these collaborative endeavors will require sus-
tainable stewardship practices to reduce the inappropriate use of antibiotics in both the
human health and agricultural sectors. Despite efforts to coordinate international and
national AMR surveillance, it would be well-advised for AMR policy, drug development,
and surveillance efforts to include both ESKAPE pathogens and other serious health
threats, such as AMR E. coli, Neisseria gonorrhoeae, and Campylobacter spp. These will
require equal attention to avoid selecting for a new group of AMR pathogen threats
(12). Improvements in factors encompassing AMR surveillance, diagnostics, patient
education, and patient treatment options will together help facilitate the control
of AMR.
REFERENCES
1. Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, A review on antibiotic resistance: alarm bells are ringing. Cureus
Harbarth S, Hindler JF, Kahlmeter G, Olsson-Liljequist B, Paterson DL, 9:e1403. https://doi.org/10.7759/cureus.1403.
Rice LB, Stelling J, Struelens MJ, Vatopoulos A, Weber JT, Monnet DL. 15. Naylor NR, Atun R, Zhu N, Kulasabanathan K, Silva S, Chatterjee A,
2012. Multidrug-resistant, extensively drug-resistant and pandrug- Knight GM, Robotham JV. 2018. Estimating the burden of antimicrobial
resistant bacteria: an international expert proposal for interim standard resistance: a systematic literature review. Antimicrob Resist Infect Con-
definitions for acquired resistance. Clin Microbiol Infect 18:268 –281. trol 7:58. https://doi.org/10.1186/s13756-018-0336-y.
https://doi.org/10.1111/j.1469-0691.2011.03570.x. 16. Iguchi S, Mizutani T, Hiramatsu K, Kikuchi K. 2016. Rapid acquisition of
2. Sommer MOA, Munck C, Toft-Kehler RV, Andersson DI. 2017. Prediction linezolid resistance in methicillin-resistant Staphylococcus aureus: role
of antibiotic resistance: time for a new preclinical paradigm? Nat Rev of hypermutation and homologous recombination. PLoS One 11:
Microbiol 15:689 – 696. https://doi.org/10.1038/nrmicro.2017.75. e0155512. https://doi.org/10.1371/journal.pone.0155512.
3. World Health Organization. 2014. Antimicrobial resistance: global report 17. Giddins MJ, Macesic N, Annavajhala MK, Stump S, Khan S, McConville
on surveillance 2014. https://www.who.int/antimicrobial-resistance/ TH, Mehta M, Gomez-Simmonds A, Uhlemann AC. 2017. Successive
publications/surveillancereport/en/. Accessed 26 June 2019. emergence of ceftazidime-avibactam resistance through distinct genomic
4. Centers for Disease Control and Prevention. 2019. Antibiotic resistant adaptations in bla(KPC-2)-harboring Klebsiella pneumoniae sequence type
threats in the United States 2019. https://www.cdc.gov/drugresistance/ 307 isolates. Antimicrob Agents Chemother 62:e02101-17. https://doi.org/
pdf/threats-report/2019-ar-threats-report-508.pdf. Accessed 15 January 10.1128/AAC.02101-17.
2020. 18. Herc ES, Kauffman CA, Marini BL, Perissinotti AJ, Miceli MH. 2017.
5. Cassini A, Högberg LD, Plachouras D, Quattrocchi A, Hoxha A, Simonsen Daptomycin nonsusceptible vancomycin resistant Enterococcus blood-
GS, Colomb-Cotinat M, Kretzschmar ME, Devleesschauwer B, Cecchini stream infections in patients with hematological malignancies: risk
M, Ouakrim DA, Oliveira TC, Struelens MJ, Suetens C, Monnet DL, factors and outcomes. Leuk Lymphoma 58:2852–2858. https://doi.org/
Strauss R, Mertens K, Struyf T, Catry B, Latour K, Ivanov IN, Dobreva EG, 10.1080/10428194.2017.1312665.
Tambic Andraševic A, Soprek S, Budimir A, Paphitou N, Žemlicková H, 19. Paterson DL, Bonomo RA. 2005. Extended-spectrum beta-lactamases: a
Schytte Olsen S, Wolff Sönksen U, Märtin P, Ivanova M, Lyytikäinen O, clinical update. Clin Microbiol Rev 18:657– 686. https://doi.org/10.1128/
Jalava J, Coignard B, Eckmanns T, Abu Sin M, Haller S, Daikos GL, Gikas CMR.18.4.657-686.2005.
A, Tsiodras S, Kontopidou F, Tóth Á, Hajdu Á, Guólaugsson Ó, Kristin- 20. Kussmann M, Karer M, Obermueller M, Schmidt K, Barousch W,
Moser D, Nehr M, Ramharter M, Poeppl W, Makristathis A, Winkler S,
sson KG, Murchan S, Burns K, Pezzotti P, Gagliotti C, Dumpis U, et al.
Thalhammer F, Burgmann H, Lagler H. 2018. Emergence of a dalba-
2019. Attributable deaths and disability-adjusted life-years caused by
vancin induced glycopeptide/lipoglycopeptide non-susceptible
infections with antibiotic-resistant bacteria in the EU and the European
Staphylococcus aureus during treatment of a cardiac device-related
Economic Area in 2015: a population-level modelling analysis. Lancet
endocarditis. Emerg Microbes Infect 7:202. https://doi.org/10.1038/
Infect Dis 19:56 – 66. https://doi.org/10.1016/S1473-099(18)30605-4.
s41426-018-0205-z.
6. Weist K, Hogberg LD. 2016. ECDC publishes 2015 surveillance data on
21. CenterWatch. 2020. FDA approved drugs. https://www.centerwatch.com/
antimicrobial resistance and antimicrobial consumption in Europe.
directories/1067-fda-approved-drugs/topic/546-bacterial-infections. Ac-
Euro Surveill 21(46):pii⫽30401. https://doi.org/10.2807/1560-7917.ES
cessed 10 January 2020.
.2016.21.46.30399.
22. European Medicines Agency. 2020. Xerava. https://www.ema.europa
7. Center for Disease Dynamics Economics & Policy. 2015. The state of the
.eu/en/medicines/human/EPAR/xerava. Accessed 13 January 2020.
world’s antibiotics, 2015. https://cddep.org/publications/state_worlds 23. European Medicines Agency. 2020. Quofenix. https://www.ema.europa
_antibiotics_2015/. Accessed 1 November 2019. .eu/en/medicines/human/EPAR/quofenix. Accessed 13 January 2020.
8. Kayange N, Kamugisha E, Mwizamholya DL, Jeremiah S, Mshana SE. 24. European Medicines Agency. 2020. Recarbrio. https://www.ema.europa
2010. Predictors of positive blood culture and deaths among neonates .eu/en/medicines/human/EPAR/recarbrio. Accessed 13 January 2020.
with suspected neonatal sepsis in a tertiary hospital, Mwanza-Tanzania. 25. European Medicines Agency. 2020. Vaborem. https://www.ema.europa
BMC Pediatr 10:39. https://doi.org/10.1186/1471-2431-10-39. .eu/en/medicines/human/EPAR/vaborem. Accessed 11 March 2020.
9. Roca A, Quinto L, Abacassamo F, Morais L, Valles X, Espasa M, Sigauque 26. Giacobbe DR, De Rosa FG, Del Bono V, Grossi PA, Pea F, Petrosillo N,
B, Sacarlal J, Macete E, Nhacolo A, Mandomando I, Levine MM, Alonso Rossolini GM, Tascini C, Tumbarello M, Viale P, Bassetti M. 2019.
PL. 2008. Invasive Haemophilus influenzae disease in children less than Ceftobiprole: drug evaluation and place in therapy. Expert Rev Anti Infect
5 years of age in Manhica, a rural area of southern Mozambique. Trop Ther 17:689–698. https://doi.org/10.1080/14787210.2019.1667229.
Med Int Health 13:818 – 826. https://doi.org/10.1111/j.1365-3156.2008 27. PMDA. 2019. Pharmaceuticals and medical devices agency: list of
.02061.x. approved products. https://www.pmda.go.jp/english/review-services/
10. Akova M. 2016. Epidemiology of antimicrobial resistance in bloodstream reviews/approved-information/drugs/0002.html. Accessed 10 January
infections. Virulence 7:252–266. https://doi.org/10.1080/21505594.2016 2020.
.1159366. 28. Australian Government Therapeutic Goods Administration. 2016. Aus-
11. Rice LB. 2008. Federal funding for the study of antimicrobial resistance tralian public assessment report for ceftobiprole medocaril sodium.
in nosocomial pathogens: no ESKAPE. J Infect Dis 197:1079 –1081. https://www.tga.gov.au/sites/default/files/auspar-ceftobiprole
https://doi.org/10.1086/533452. -medocaril-sodium-160208.pdf. Accessed 11 March 2020.
12. World Health Organization. 2017. Global priority list of antibiotic- 29. Government of Canada. 2020. Drug product database. https://www
resistant bacteria to guide research, discovery, and development of .canada.ca/en/health-canada/services/drugs-health-products/drug
new antibiotics. http://www.who.int/medicines/publications/WHO-PPL -products/drug-product-database.html. Accessed 10 January 2020.
-Short_Summary_25Feb-ET_NM_WHO.pdf?ua⫽1. Accessed 2 Novem- 30. Lebreton F, van Schaik W, McGuire AM, Godfrey P, Griggs A, Mazumdar
ber 2019. V, Corander J, Cheng L, Saif S, Young S, Zeng Q, Wortman J, Birren B,
13. Beatson SA, Walker MJ. 2014. Microbiology. Tracking antibiotic resistance. Willems RJ, Earl AM, Gilmore MS. 2013. Emergence of epidemic
Science 345:1454–1455. https://doi.org/10.1126/science.1260471. multidrug-resistant Enterococcus faecium from animal and commensal
14. Bin Zaman S, Hussain MA, Nye R, Mehta V, Mamun KT, Hossain N. 2017. strains. mBio 4:e00534-13. https://doi.org/10.1128/mBio.00534-13.
31. Murray BE. 1990. The life and times of the Enterococcus. Clin Microbiol Robson J, Peachey G, Papanaoum K, Wells N, Warner M, Smith K, Cooley
Rev 3:46 – 65. https://doi.org/10.1128/cmr.3.1.46. L, Jones D, Kalukottege P, Wilcox K, Spelman D, Bernhard R, Johnson P,
32. Pinholt M, Larner-Svensson H, Littauer P, Moser CE, Pedersen M, Lem- Hurren F, Korman T, Kotsanas D, Daley A, Gonis G. 2018. Australian
ming LE, Ejlertsen T, Sondergaard TS, Holzknecht BJ, Justesen US, Dzajic Group on Antimicrobial Resistance (AGAR) Australian Enterococcal Sep-
E, Olsen SS, Nielsen JB, Worning P, Hammerum AM, Westh H, Jakobsen sis Outcome Programme (AESOP) annual report 2016. Commun Dis
L. 2015. Multiple hospital outbreaks of vanA Enterococcus faecium in Intell 42:PII⫽S2209-6051(18)00020-9.
Denmark, 2012–13, investigated by WGS, MLST and PFGE. J Antimicrob 49. Prasad P, Sun JF, Danner RL, Natanson C. 2012. Excess deaths associ-
Chemother 70:2474 –2482. https://doi.org/10.1093/jac/dkv142. ated with tigecycline after approval based on noninferiority trials. Clin
33. Gastmeier P, Schroder C, Behnke M, Meyer E, Geffers C. 2014. Dramatic Infect Dis 54:1699 –1709. https://doi.org/10.1093/cid/cis270.
increase in vancomycin-resistant enterococci in Germany. J Antimicrob 50. Silverman JA, Mortin LI, VanPraagh ADG, Li TC, Alder J. 2005. Inhibition
Chemother 69:1660 –1664. https://doi.org/10.1093/jac/dku035. of daptomycin by pulmonary surfactant: in vitro modeling and clinical
34. Coombs GW, Pearson JC, Daley DA, Le T, Robinson OJ, Gottlieb T, impact. J Infect Dis 191:2149 –2152. https://doi.org/10.1086/430352.
Howden BP, Johnson PD, Bennett CM, Stinear TP, Turnidge JD, Austra- 51. Cetinkaya Y, Falk P, Mayhall CG. 2000. Vancomycin-resistant entero-
lian Group on Antimicrobial Resistance. 2014. Molecular epidemiology cocci. Clin Microbiol Rev 13:686 –707. https://doi.org/10.1128/cmr.13.4
of enterococcal bacteremia in Australia. J Clin Microbiol 52:897–905. .686-707.2000.
https://doi.org/10.1128/JCM.03286-13. 52. Prematunge C, MacDougall C, Johnstone J, Adomako K, Lam F, Robertson
35. Australian Commission on Safety and Quality in Health Care. 2019. J, Garber G. 2016. VRE and VSE bacteremia outcomes in the era of effective
AURA 2019. Third Australian report on antimicrobial use and resistance VRE therapy: a systematic review and meta-analysis. Infect Control Hosp
in human health. https://www.safetyandquality.gov.au/sites/default/ Epidemiol 37:26–35. https://doi.org/10.1017/ice.2015.228.
files/2019-06/AURA-2019-Report.pdf. Accessed 10 November 2019. 53. Chiang HY, Perencevich EN, Nair R, Nelson RE, Samore M, Khader K,
36. Lee T, Pang S, Abraham S, Coombs GW. 2019. Antimicrobial-resistant Chorazy ML, Herwaldt LA, Blevins A, Ward MA, Schweizer ML. 2017.
CC17 Enterococcus faecium: the past, the present and the future. J Glob Incidence and outcomes associated with infections caused by
Antimicrob Resist 16:36 – 47. https://doi.org/10.1016/j.jgar.2018.08.016. vancomycin-resistant enterococci in the United States: systematic lit-
37. Goncalves A, Igrejas G, Radhouani H, Lopez M, Guerra A, Petrucci- erature review and meta-analysis. Infect Control Hosp Epidemiol 38:
Fonseca F, Alcaide E, Zorrilla I, Serra R, Torres C, Poeta P. 2011. Detec- 203–215. https://doi.org/10.1017/ice.2016.254.
tion of vancomycin-resistant enterococci from faecal samples of Iberian 54. DiazGranados CA, Jernigan JA. 2005. Impact of vancomycin resis-
wolf and Iberian lynx, including Enterococcus faecium strains of CC17 tance on mortality among patients with neutropenia and entero-
and the new singleton ST573. Sci Total Environ 410-411:266 –268. coccal bloodstream infection. J Infect Dis 191:588 –595. https://doi
https://doi.org/10.1016/j.scitotenv.2011.09.074. .org/10.1086/427512.
38. Marques C, Belas A, Franco A, Aboim C, Gama LT, Pomba C. 2018. 55. Eriksen KR. 1961. “Celbenin⬙-resistant staphylococci. Ugeskr Laeger
Increase in antimicrobial resistance and emergence of major interna- 123:384 –386. (In Danish.)
tional high-risk clonal lineages in dogs and cats with urinary tract
56. Kourtis AP, Hatfield K, Baggs J, Mu Y, See I, Epson E, Nadle J, Kainer MA,
infection: 16 year retrospective study. J Antimicrob Chemother 73:
Dumyati G, Petit S, Ray SM, Emerging Infections Program MRSA Author
377–384. https://doi.org/10.1093/jac/dkx401.
Group, Ham D, Capers C, Ewing H, Coffin N, McDonald LC, Jernigan J,
39. da Silva LP, Pitondo-Silva A, Martinez R, da Costa Darini AL. 2012.
Cardo D, Program EI. 2019. Vital signs: epidemiology and recent trends
Genetic features and molecular epidemiology of Enterococcus faecium
in methicillin-resistant and in methicillin-susceptible Staphylococcus
isolated in two university hospitals in Brazil. Diagn Microbiol Infect Dis
aureus bloodstream infections—United States. MMWR Morb Mortal
74:267–271. https://doi.org/10.1016/j.diagmicrobio.2012.07.012.
Wkly Rep 68:214 –219. https://doi.org/10.15585/mmwr.mm6809e1.
40. Kang M, Xie Y, He C, Chen ZX, Guo L, Yang Q, Liu JY, Du Y, Ou QS, Wang
57. Udo EE, Pearman JW, Grubb WB. 1993. Genetic analysis of commu-
LL. 2014. Molecular characteristics of vancomycin-resistant Enterococ-
nity isolates of methicillin-resistant Staphylococcus aureus in West-
cus faecium from a tertiary care hospital in Chengdu, China: molecular
ern Australia. J Hosp Infect 25:97–108. https://doi.org/10.1016/0195
characteristics of VRE in China. Eur J Clin Microbiol Infect Dis 33:
-6701(93)90100-e.
933–939. https://doi.org/10.1007/s10096-013-2029-z.
58. Diekema DJ, Pfaller MA, Schmitz FJ, Smayevsky J, Bell J, Jones RN, Beach
41. Strateva T, Sirakov I, Dimov S, Trifonova A, Savov E, Mitov I. 2018. Clonal
spread of vanA Enterococcus faecium sequence type 203 in Bulgarian M, SENTRY Participants Group. 2001. Survey of infections due to Staph-
hospitals. Infect Dis (Lond) 50:718–721. https://doi.org/10.1080/23744235 ylococcus species: frequency of occurrence and antimicrobial suscepti-
.2018.1453946. bility of isolates collected in the United States, Canada, Latin America,
42. Gao W, Howden BP, Stinear TP. 2018. Evolution of virulence in Entero- Europe, and the Western Pacific region for the SENTRY Antimicrobial
coccus faecium, a hospital-adapted opportunistic pathogen. Curr Opin Surveillance Program, 1997–1999. Clin Infect Dis 32:S114 –S132. https://
Microbiol 41:76 – 82. https://doi.org/10.1016/j.mib.2017.11.030. doi.org/10.1086/320184.
43. Satilmis L, Vanhems P, Benet T. 2016. Outbreaks of vancomycin- 59. Gilbert M, MacDonald J, Gregson D, Siushansian J, Zhang K, Elsayed S,
resistant enterococci in hospital settings: a systematic review and Laupland K, Louie T, Hope K, Mulvey M, Gillespie J, Nielsen D, Wheeler
calculation of the basic reproductive number. Infect Control Hosp V, Louie M, Honish A, Keays G, Conly J. 2006. Outbreak in Alberta of
Epidemiol 37:289 –294. https://doi.org/10.1017/ice.2015.301. community-acquired (USA300) methicillin-resistant Staphylococcus au-
44. Sood G, Perl TM. 2016. Outbreaks in health care settings. Infect Dis Clin reus in people with a history of drug use, homelessness or incarcera-
North Am 30:661– 687. https://doi.org/10.1016/j.idc.2016.04.003. tion. CMAJ 175:149 –154. https://doi.org/10.1503/cmaj.051565.
45. Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M, Viale A, Socci 60. Mulvey MR, MacDougall L, Cholin B, Horsman G, Fidyk M, Woods S,
ND, van den Brink MR, Kamboj M, Pamer EG. 2010. Vancomycin-resistant Saskatchewan CA-MRSA Study Group. 2005. Community-associated
Enterococcus domination of intestinal microbiota is enabled by antibiotic methicillin-resistant Staphylococcus aureus, Canada. Emerg Infect Dis
treatment in mice and precedes bloodstream invasion in humans. J Clin 11:844 – 850. https://doi.org/10.3201/eid1106.041146.
Invest 120:4332–4341. https://doi.org/10.1172/JCI43918. 61. David MZ, Rudolph KM, Hennessy TW, Boyle-Vavra S, Daum RS. 2008.
46. Buffie CG, Pamer EG. 2013. Microbiota-mediated colonization resis- Molecular epidemiology of methicillin-resistant Staphylococcus aureus,
tance against intestinal pathogens. Nat Rev Immunol 13:790 – 801. rural southwestern Alaska. Emerg Infect Dis 14:1693–1699. https://doi
https://doi.org/10.1038/nri3535. .org/10.3201/eid1411.080381.
47. Gouliouris T, Warne B, Cartwright EJP, Bedford L, Weerasuriya CK, Raven 62. Diep BA, Sensabaugh GF, Somboona NS, Carleton HA, Perdreau-
KE, Brown NM, Török ME, Limmathurotsakul D, Peacock SJ. 2018. Remington F. 2004. Widespread skin and soft-tissue infections due to
Duration of exposure to multiple antibiotics is associated with in- two methicillin-resistant Staphylococcus aureus strains harboring the
creased risk of VRE bacteraemia: a nested case-control study. J Antimi- genes for Panton-Valentine leucocidin. J Clin Microbiol 42:2080 –2084.
crob Chemother 73:1692–1699. https://doi.org/10.1093/jac/dky075. https://doi.org/10.1128/JCM.42.5.2080-2084.2004.
48. Coombs GW, Daley DA, Thin Y, Pang S, Collignon P, Bradbury S, 63. Lim WW, Wu P, Bond HS, Wong JY, Ni KW, Seto WH, Jit M, Cowling BJ.
Gottlieb T, Robertson G, Branley J, Barbaro D, Huntington P, van Hal S, 2019. Determinants of methicillin-resistant Staphylococcus aureus
Beukers A, Iredell J, Ginn A, Givney R, Winney I, Newton P, Hoddle M, (MRSA) prevalence in the Asia-Pacific region: a systematic review and
Baird R, Hennessy J, McLeod J, Binotto E, Thomsett B, Nimmo G, George meta-analysis. J Glob Antimicrob Resist 16:17–27. https://doi.org/10
N, Maloney S, Curtis C, Horvath R, Martin L, Runnegar N, Douglas J, .1016/j.jgar.2018.08.014.
64. European Antimicrobial Resistance Surveillance Network. 2019. Surveil- 81. Yigit H, Queenan AM, Anderson GJ, Domenech-Sanchez A, Biddle JW,
lance of antimicrobial resistance in Europe. Annual report of the Euro- Steward CD, Alberti S, Bush K, Tenover FC. 2001. Novel carbapenem-
pean Antimicrobial Resistance Surveillance Network (EARS-Net) 2018. hydrolyzing beta-lactamase, KPC-1, from a carbapenem-resistant strain
https://www.ecdc.europa.eu/sites/default/files/documents/surveillance of Klebsiella pneumoniae. Antimicrob Agents Chemother 45:1151–1161.
-antimicrobial-resistance-Europe-2018.pdf. Accessed 13 January 2020. https://doi.org/10.1128/AAC.45.4.1151-1161.2001.
65. Dai YX, Liu JL, Guo W, Meng HW, Huang Q, He L, Gao QQ, Lv HY, Liu Y, 82. David S, Reuter S, Harris SR, Glasner C, Feltwell T, Argimon S, Abudahab
Wang YN, Wang H, Liu Q, Li M. 2019. Decreasing methicillin-resistant K, Goater R, Giani T, Errico G, Aspbury M, Sjunnebo S, EuSCAPE Working
Staphylococcus aureus (MRSA) infections is attributable to the disap- Group, ESGEM Study Group, Feil EJ, Rossolini GM, Aanensen DM,
pearance of predominant MRSA ST239 clones, Shanghai, 2008 –2017. Grundmann H. 2019. Epidemic of carbapenem-resistant Klebsiella pneu-
Emerg Microbes Infect 8:471– 478. https://doi.org/10.1080/22221751 moniae in Europe is driven by nosocomial spread. Nat Microbiol
.2019.1595161. 4:1919 –1929. https://doi.org/10.1038/s41564-019-0492-8.
66. Duerden B, Fry C, Johnson AP, Wilcox MH. 2015. The control of 83. Hall JM, Corea E, Sanjeewani HD, Inglis TJ. 2014. Molecular mechanisms
methicillin-resistant Staphylococcus aureus blood stream infections in of beta-lactam resistance in carbapenemase-producing Klebsiella pneu-
England. Open Forum Infect Dis 2:ofv035. https://doi.org/10.1093/ofid/ moniae from Sri Lanka. J Med Microbiol 63:1087–1092. https://doi.org/
ofv035. 10.1099/jmm.0.076760-0.
67. Carlet J, Astagneau P, Brun-Buisson C, Coignard B, Salomon V, Tran B, 84. Hall JM, Ingram PR, O’Reilly LC, Inglis T. 2016. Temporal flux in beta-
Desenclos JC, Jarlier V, Schlemmer B, Parneix P, Regnier B, Fabry J, Hea F, lactam resistance among Klebsiella pneumoniae in Western Australia. J
French National Program for Prevention of Healthcare-Associated Infec- Med Microbiol 65:429 – 437. https://doi.org/10.1099/jmm.0.000242.
tions and Antimicrobial Resistance. 2009. French National Program for 85. Guh AY, Bulens SN, Mu Y, Jacob JT, Reno J, Scott J, Wilson LE, Vaeth E,
Prevention of Healthcare-Associated Infections and Antimicrobial Resis- Lynfield R, Shaw KM, Vagnone PMS, Bamberg WM, Janelle SJ, Dumyati
tance, 1992–2008: positive trends, but perseverance needed. Infect Con- G, Concannon C, Beldavs Z, Cunningham M, Cassidy PM, Phipps EC,
trol Hosp Epidemiol 30:737–745. https://doi.org/10.1086/598682. Kenslow N, Travis T, Lonsway D, Rasheed JK, Limbago BM, Kallen AJ.
68. DeLeo FR, Otto M, Kreiswirth BN, Chambers HF. 2010. Community- 2015. Epidemiology of carbapenem-resistant Enterobacteriaceae in 7 US
associated meticillin-resistant Staphylococcus aureus. Lancet 375: communities, 2012–2013. JAMA 314:1479 –1487. https://doi.org/10
1557–1568. https://doi.org/10.1016/S0140-6736(09)61999-1. .1001/jama.2015.12480.
69. Dermota U, Jurca T, Harlander T, Kosir M, Zajc U, Golob M, Zdovc I, 86. Zhang R, Chan EWC, Zhou HW, Chen S. 2017. Prevalence and genetic
Kosnik IG. 2016. Infections caused by community-associated characteristics of carbapenem-resistant Enterobacteriaceae strains in
methicillin-resistant Staphylococcus aureus European clone (St80) in China. Lancet Infect Dis 17:256 –257. https://doi.org/10.1016/S1473
Slovenia between 2006 and 2013. Zdr Varst 55:121–125. https://doi -3099(17)30072-5.
.org/10.1515/sjph-2016-0018. 87. Wei DD, Wan LG, Deng Q, Liu Y. 2016. Emergence of KPC-producing
70. Drougka E, Foka A, Liakopoulos A, Doudoulakakis A, Jelastopulu E, Chini V, Klebsiella pneumoniae hypervirulent clone of capsular serotype K1 that
Spiliopoulou A, Levidiotou S, Panagea T, Vogiatzi A, Lebessi E, Petinaki E, belongs to sequence type 11 in mainland China. Diagn Microbiol Infect
Spiliopoulou I. 2014. A 12-year survey of methicillin-resistant Staphylococ- Dis 85:192–194. https://doi.org/10.1016/j.diagmicrobio.2015.03.012.
cus aureus infections in Greece: ST80-IV epidemic? Clin Microbiol Infect 88. Wyres KL, Hawkey J, Hetland MAK, Fostervold A, Wick RR, Judd LM,
20:O796–O803. https://doi.org/10.1111/1469-0691.12624. Hamidian M, Howden BP, Lohr IH, Holt KE. 2019. Emergence and rapid
71. Fluit AC. 2012. Livestock-associated Staphylococcus aureus. Clin Microbiol global dissemination of CTX-M-15-associated Klebsiella pneumoniae
Infect 18:735–744. https://doi.org/10.1111/j.1469-0691.2012.03846.x. strain ST307. J Antimicrob Chemother 74:577–581. https://doi.org/10
72. van Alen S, Ballhausen B, Peters G, Friedrich AW, Mellmann A, Kock R, .1093/jac/dky492.
Becker K. 2017. In the centre of an epidemic: fifteen years of LA-MRSA 89. Lowe M, Kock MM, Coetzee J, Hoosien E, Peirano G, Strydom KA, Ehlers
CC398 at the university hospital Munster. Vet Microbiol 200:19 –24. MM, Mbelle NM, Shashkina E, Haslam DB, Dhawan P, Donnelly RJ, Chen
https://doi.org/10.1016/j.vetmic.2016.01.021. L, Kreiswirth BN, Pitout J. 2019. Klebsiella pneumoniae ST307 with
73. Hryniewicz MM, Garbacz K. 2017. Borderline oxacillin-resistant Staphy- blaOXA-181, South Africa, 2014 –2016. Emerg Infect Dis 25:739 –747.
lococcus aureus (BORSA)—a more common problem than expected? J https://doi.org/10.3201/eid2504.181482.
Med Microbiol 66:1367–1373. https://doi.org/10.1099/jmm.0.000585. 90. Cheng NC, Yu YC, Tai HC, Hsueh PR, Chang SC, Lai SY, Yi WC, Fang CT.
74. Skinner S, Murray M, Walus T, Karlowsky JA. 2009. Failure of cloxacillin 2012. Recent trend of necrotizing fasciitis in Taiwan: focus on mono-
in treatment of a patient with borderline oxacillin-resistant Staphylo- microbial Klebsiella pneumoniae necrotizing fasciitis. Clin Infect Dis
coccus aureus endocarditis. J Clin Microbiol 47:859 – 861. https://doi 55:930 –939. https://doi.org/10.1093/cid/cis565.
.org/10.1128/JCM.00571-08. 91. Bernal NP, Latenser BA, Born JM, Liao J. 2012. Trends in 393 necrotizing
75. Pada SK, Ding Y, Ling ML, Hsu LY, Earnest A, Lee TE, Yong HC, Jureen acute soft tissue infection patients 2000 –2008. Burns 38:252–260.
R, Fisher D. 2011. Economic and clinical impact of nosocomial https://doi.org/10.1016/j.burns.2011.07.008.
meticillin-resistant Staphylococcus aureus infections in Singapore: a 92. Krapp F, Morris AR, Ozer EA, Hauser AR. 2017. Virulence characteristics
matched case-control study. J Hosp Infect 78:36 – 40. https://doi.org/10 of carbapenem-resistant Klebsiella pneumoniae strains from patients
.1016/j.jhin.2010.10.016. with necrotizing skin and soft tissue infections. Sci Rep 7:13533. https://
76. Cosgrove SE, Sakoulas G, Perencevich EN, Schwaber MJ, Karchmer AW, doi.org/10.1038/s41598-017-13524-8.
Carmeli Y. 2003. Comparison of mortality associated with methicillin- 93. Li W, Sun GZ, Yu YH, Li N, Chen M, Jin RH, Jiao YM, Wu H. 2014.
resistant and methicillin-susceptible Staphylococcus aureus bacteremia: a Increasing occurrence of antimicrobial-resistant hypervirulent (hyper-
meta-analysis. Clin Infect Dis 36:53–59. https://doi.org/10.1086/345476. mucoviscous) Klebsiella pneumoniae isolates in China. Clin Infect Dis
77. Kaasch AJ, Barlow G, Edgeworth JD, Fowler VG, Hellmich M, Hopkins S, 58:225–232. https://doi.org/10.1093/cid/cit675.
Kern WV, Llewelyn MJ, Rieg S, Rodriguez-Baño J, Scarborough M, Seifert 94. Munoz-Price LS, Weinstein RA. 2008. Acinetobacter infection. N Engl J
H, Soriano A, Tilley R, Tőrők ME, Weiß V, Wilson APR, Thwaites GE. 2014. Med 358:1271–1281. https://doi.org/10.1056/NEJMra070741.
Staphylococcus aureus bloodstream infection: a pooled analysis of five 95. Villegas MV, Hartstein AI. 2003. Acinetobacter outbreaks, 1977–2000.
prospective, observational studies. J Infect 68:242–251. https://doi.org/ Infect Control Hosp Epidemiol 24:284 –295. https://doi.org/10.1086/
10.1016/j.jinf.2013.10.015. 502205.
78. Tzouvelekis LS, Markogiannakis A, Psichogiou M, Tassios PT, Daikos GL. 96. Ibrahim ME. 2019. Prevalence of Acinetobacter baumannii in Saudi
2012. Carbapenemases in Klebsiella pneumoniae and other Arabia: risk factors, antimicrobial resistance patterns and mechanisms
Enterobacteriaceae: an evolving crisis of global dimensions. Clin Micro- of carbapenem resistance. Ann Clin Microbiol Antimicrob 18:1. https://
biol Rev 25:682–707. https://doi.org/10.1128/CMR.05035-11. doi.org/10.1186/s12941-018-0301-x.
79. Kaewpoowat Q, Ostrosky-Zeichner L. 2015. Tigecycline: a critical safety 97. McDonald LC, Banerjee SN, Jarvis WR, National Nosocomial Infections
review. Expert Opin Drug Saf 14:335–342. https://doi.org/10.1517/ Surveillance System. 1999. Seasonal variation of Acinetobacter
14740338.2015.997206. infections: 1987–1996. Clin Infect Dis 29:1133–1137. https://doi.org/10
80. Centers for Disease Control and Prevention. 2013. Antibiotic resistance .1086/313441.
threats in the United States, 2013. https://www.cdc.gov/drugresistance/ 98. Harding CM, Hennon SW, Feldman MF. 2018. Uncovering the mecha-
threat-report-2013/pdf/ar-threats-2013-508.pdf. Accessed 23 October nisms of Acinetobacter baumannii virulence. Nat Rev Microbiol 16:
2019. 91–102. https://doi.org/10.1038/nrmicro.2017.148.
99. Davis JS, McMillan M, Swaminathan A, Kelly JA, Piera KE, Baird RW, drug-resistant ST292 clone in Kunming. Sci Rep 6:26522. https://doi
Currie BJ, Anstey NM. 2014. A 16-year prospective study of community- .org/10.1038/srep26522.
onset bacteremic Acinetobacter pneumonia: low mortality with appro- 115. Langan KM, Kotsimbos T, Peleg AY. 2015. Managing Pseudomonas
priate initial empirical antibiotic protocols. Chest 146:1038 –1045. aeruginosa respiratory infections in cystic fibrosis. Curr Opin Infect Dis
https://doi.org/10.1378/chest.13-3065. 28:547–556. https://doi.org/10.1097/QCO.0000000000000217.
100. Lob SH, Hoban DJ, Sahm DF, Badal RE. 2016. Regional differences 116. LaFayette SL, Houle D, Beaudoin T, Wojewodka G, Radzioch D, Hoffman
and trends in antimicrobial susceptibility of Acinetobacter bauman- LR, Burns JL, Dandekar AA, Smalley NE, Chandler JR, Zlosnik JE, Speert
nii. Int J Antimicrob Agents 47:317–323. https://doi.org/10.1016/j DP, Bernier J, Matouk E, Brochiero E, Rousseau S, Nguyen D. 2015.
.ijantimicag.2016.01.015. Cystic fibrosis-adapted Pseudomonas aeruginosa quorum sensing lasR
101. Magill SS, Edwards JR, Bamberg W, Beldavs ZG, Dumyati G, Kainer mutants cause hyperinflammatory responses. Sci Adv 1:e1500199.
MA, Lynfield R, Maloney M, McAllister-Hollod L, Nadle J, Ray SM, https://doi.org/10.1126/sciadv.1500199.
Thompson DL, Wilson LE, Fridkin SK, Emerging Infections Program 117. Newman JW, Floyd RV, Fothergill JL. 2017. The contribution of Pseu-
Healthcare-Associated Infections and Antimicrobial Use Prevalence domonas aeruginosa virulence factors and host factors in the establish-
Survey Team. 2014. Multistate point-prevalence survey of health ment of urinary tract infections. FEMS Microbiol Lett 364:fnx124.
care-associated infections. N Engl J Med 370:1198 –1208. https://doi https://doi.org/10.1093/femsle/fnx124.
.org/10.1056/NEJMoa1306801. 118. Hoiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. 2010. Antibiotic
102. Giammanco A, Cala C, Fasciana T, Dowzicky MJ. 2017. Global assess- resistance of bacterial biofilms. Int J Antimicrob Agents 35:322–332.
ment of the activity of tigecycline against multidrug-resistant Gram- https://doi.org/10.1016/j.ijantimicag.2009.12.011.
negative pathogens between 2004 and 2014 as part of the tigecycline 119. Davin-Regli A, Pagès J-M. 2015. Enterobacter aerogenes and Enterobac-
evaluation and surveillance trial. mSphere 2:e00310-16. https://doi.org/ ter cloacae; versatile bacterial pathogens confronting antibiotic treat-
10.1128/mSphere.00310-16. ment. Front Microbiol 6:392. https://doi.org/10.3389/fmicb.2015.00392.
103. Xie RQ, Zhang XD, Zhao Q, Peng B, Zheng J. 2018. Analysis of global 120. Georghiou PR, Hamill RJ, Wright CE, Versalovic J, Koeuth T, Watson DA,
prevalence of antibiotic resistance in Acinetobacter baumannii infec- Lupski JR. 1995. Molecular epidemiology of infections due to Entero-
tions disclosed a faster increase in OECD countries. Emerg Microbes bacter aerogenes—identification of hospital outbreak-associated strains
Infect 7:31. https://doi.org/10.1038/s41426-018-0038-9. by molecular techniques. Clin Infect Dis 20:84 –94. https://doi.org/10
104. Lee HY, Chang RC, Su LH, Liu SY, Wu SR, Chuang CH, Chen CL, Chiu CH. .1093/clinids/20.1.84.
2012. Wide spread of Tn2006 in an AbaR4-type resistance island among 121. Grattard F, Pozzetto B, Tabard L, Petit M, Ros A, Gaudin OG. 1995.
carbapenem-resistant Acinetobacter baumannii clinical isolates in Taiwan. Characterization of nosocomial strains of Enterobacter aerogenes by
Int J Antimicrob Agents 40:163–167. https://doi.org/10.1016/j.ijantimicag arbitrarily primed-PCR analysis and ribotyping. Infect Control Hosp
.2012.04.018. Epidemiol 16:224 –230. https://doi.org/10.1086/647094.
105. Zarrilli R, Pournaras S, Giannouli M, Tsakris A. 2013. Global evolution of 122. Bosi C, Davin-Regli A, Bornet C, Mallea M, Pages JM, Bollet C. 1999. Most
Enterobacter aerogenes strains in France belong to a prevalent clone. J
multidrug-resistant Acinetobacter baumannii clonal lineages. Int J An-
Clin Microbiol 37:2165–2169.
timicrob Agents 41:11–19. https://doi.org/10.1016/j.ijantimicag.2012.09
123. Chevalier J, Mulfinger C, Garnotel E, Nicolas P, Davin-Régli A, Pagès J-M.
.008.
2008. Identification and evolution of drug efflux pump in clinical
106. Tavares LCB, de Vasconcellos FM, de Sousa WV, Rocchetti TT, Mondelli
Enterobacter aerogenes strains isolated in 1995 and 2003. PLoS One
AL, Ferreira AM, Montelli AC, Sadatsune T, Tiba-Casas MR, Camargo CH.
3:e3203. https://doi.org/10.1371/journal.pone.0003203.
2019. Emergence and persistence of high-risk clones among MDR and
124. Mezzatesta ML, Gona F, Stefani S. 2012. Enterobacter cloacae complex:
XDR A. baumannii at a Brazilian teaching hospital. Front Microbiol
clinical impact and emerging antibiotic resistance. Future Microbiol
9:2898. https://doi.org/10.3389/fmicb.2018.02898.
7:887–902. https://doi.org/10.2217/fmb.12.61.
107. Escandón-Vargas K, Reyes S, Gutiérrez S, Villegas MV. 2017. The epide-
125. Paauw A, Caspers MPM, Schuren FHJ, Leverstein-van Hall MA, Deletoile
miology of carbapenemases in Latin America and the Caribbean. Ex-
A, Montijn RC, Verhoef J, Fluit AC. 2008. Genomic diversity within the
pert Rev Anti Infect Ther 15:277–297. https://doi.org/10.1080/14787210
Enterobacter cloacae complex. PLoS One 3:e3018. https://doi.org/10
.2017.1268918.
.1371/journal.pone.0003018.
108. Qureshi ZA, Hittle LE, O’Hara JA, Rivera JI, Syed A, Shields RK, Pasculle 126. Annavajhala MK, Gomez-Simmonds A, Uhlemann AC. 2019. Multidrug-
AW, Ernst RK, Doi Y. 2015. Colistin-resistant Acinetobacter baumannii: resistant Enterobacter cloacae complex emerging as a global, diversi-
beyond carbapenem resistance. Clin Infect Dis 60:1295–1303. https:// fying threat. Front Microbiol 10:44. https://doi.org/10.3389/fmicb.2019
doi.org/10.1093/cid/civ048. .00044.
109. Gellatly SL, Hancock R. 2013. Pseudomonas aeruginosa: new insights 127. Malek A, McGlynn K, Taffner S, Fine L, Tesini B, Wang J, Mostafa H, Petry
into pathogenesis and host defenses. Pathog Dis 67:159 –173. https:// S, Perkins A, Graman P, Hardy D, Pecora N. 2019. Next-generation
doi.org/10.1111/2049-632X.12033. sequencing based hospital outbreak investigation yields insight into
110. Yayan J, Ghebremedhin B, Rasche K. 2015. Antibiotic resistance of Klebsiella aerogenes population structure and determinants of carbap-
Pseudomonas aeruginosa in pneumonia at a single university hospital enem resistance and pathogenicity. Antimicrob Agents Chemother
center in Germany over a 10-year period. PLoS One 10:e0139836. 63:e02577-18. https://doi.org/10.1128/AAC.02577-18.
https://doi.org/10.1371/journal.pone.0139836. 128. Gomez-Simmonds A, Annavajhala MK, Wang Z, Macesic N, Hu Y, Gid-
111. Treepong P, Kos VN, Guyeux C, Blanc DS, Bertrand X, Valot B, Hocquet dins MJ, O’Malley A, Toussaint NC, Whittier S, Torres VJ, Uhlemann A-C.
D. 2018. Global emergence of the widespread Pseudomonas aeruginosa 2018. Genomic and geographic context for the evolution of high-risk
ST235 clone. Clin Microbiol Infect 24:258 –266. https://doi.org/10.1016/ carbapenem-resistant Enterobacter cloacae complex clones ST171 and
j.cmi.2017.06.018. ST78. mBio 9:e00542-18. https://doi.org/10.1128/mBio.00542-18.
112. Cabot G, Lopez-Causape C, Ocampo-Sosa AA, Sommer LM, Dominguez 129. Sader HS, Biedenbach DJ, Jones RN. 2003. Global patterns of suscep-
MA, Zamorano L, Juan C, Tubau F, Rodriguez C, Moya B, Pena C, tibility for 21 commonly utilized antimicrobial agents tested against
Martinez-Martinez L, Plesiat P, Oliver A, Spanish Network for Research 48,440 Enterobacteriaceae in the SENTRY Antimicrobial Surveillance
in Infectious Diseases. 2016. Deciphering the resistome of the wide- Program (1997–2001). Diagn Microbiol Infect Dis 47:361–364. https://
spread Pseudomonas aeruginosa sequence type 175 international high- doi.org/10.1016/S0732-8893(03)00052-X.
risk clone through whole-genome sequencing. Antimicrob Agents Che- 130. Thiolas A, Bollet C, La Scola B, Raoult D, Pagès J-M. 2005. Successive
mother 60:7415–7423. https://doi.org/10.1128/AAC.01720-16. emergence of Enterobacter aerogenes strains resistant to imipenem and
113. Xipell M, Bodro M, Marco F, Losno RA, Cardozo C, Soriano A. 2017. colistin in a patient. Antimicrob Agents Chemother 49:1354 –1358.
Clinical experience with ceftazidime/avibactam in patients with severe https://doi.org/10.1128/AAC.49.4.1354-1358.2005.
infections, including meningitis and lung abscesses, caused by exten- 131. Band VI, Crispell EK, Napier BA, Herrera CM, Tharp GK, Vavikolanu K,
sively drug-resistant Pseudomonas aeruginosa. Int J Antimicrob Agents Pohl J, Read TD, Bosinger SE, Trent MS, Burd EM, Weiss DS. 2016.
49:266 –268. https://doi.org/10.1016/j.ijantimicag.2016.11.005. Antibiotic failure mediated by a resistant subpopulation in Entero-
114. Fan X, Wu Y, Xiao M, Xu ZP, Kudinha T, Bazaj A, Kong F, Xu YC. 2016. bacter cloacae. Nat Microbiol 1:16053. https://doi.org/10.1038/
Diverse genetic background of multidrug-resistant Pseudomonas nmicrobiol.2016.53.
aeruginosa from mainland China, and emergence of an extensively 132. Schembri MA, Ben Zakour NL, Phan MD, Forde BM, Stanton-Cook M,
Beatson SA. 2015. Molecular characterization of the multidrug resistant Rossolini GM. 1999. Cloning and characterization of blaVIM, a new
Escherichia coli ST131 clone. Pathogens 4:422– 430. https://doi.org/10 integron-borne metallo-beta-lactamase gene from a Pseudomonas aerugi-
.3390/pathogens4030422. nosa clinical isolate. Antimicrob Agents Chemother 43:1584–1590.
133. Mathers AJ, Peirano G, Pitout J. 2015. The role of epidemic resistance 153. Watanabe M, Iyobe S, Inoue M, Mitsuhashi S. 1991. Transferable imi-
plasmids and international high-risk clones in the spread of multidrug- penem resistance in Pseudomonas aeruginosa. Antimicrob Agents Che-
resistant Enterobacteriaceae. Clin Microbiol Rev 28:565–591. https://doi mother 35:147–151. https://doi.org/10.1128/aac.35.1.147.
.org/10.1128/CMR.00116-14. 154. Matsumura Y, Peirano G, Devinney R, Bradford PA, Motyl MR, Adams
134. Brolund A, Lagerqvist N, Byfors S, Struelens MJ, Monnet DL, Albiger B, MD, Chen L, Kreiswirth B, Pitout J. 2017. Genomic epidemiology of
Kohlenberg A, European Antimicrobial Resistance Genes Surveillance global VIM-producing Enterobacteriaceae. J Antimicrob Chemother 72:
Network (EURGen-Net) Capacity Survey Group. 2019. Worsening epi- 2249 –2258. https://doi.org/10.1093/jac/dkx148.
demiological situation of carbapenemase-producing Enterobacteria- 155. Peirano G, Matsumura Y, Adams MD, Bradford P, Motyl M, Chen L,
ceae in Europe, assessment by national experts from 37 countries, July Kreiswirth BN, Pitout J. 2018. Genomic epidemiology of global
2018. Euro Surveill 24(9):pii⫽1900123. https://doi.org/10.2807/1560 carbapenemase-producing Enterobacter spp., 2008 –2014. Emerg Infect
-7917.ES.2019.24.9.1900123. Dis 24:1010 –1019. https://doi.org/10.3201/eid2406.171648.
135. Liu Y-Y, Wang Y, Walsh TR, Yi L-X, Zhang R, Spencer J, Doi Y, Tian G, 156. Lee K, Ha GY, Shin BM, Kim JJ, Kang JO, Jang SJ, Yong D, Chong Y,
Dong B, Huang X, Yu L-F, Gu D, Ren H, Chen X, Lv L, He D, Zhou H, Liang Korean Nationwide Surveillance of Antimicrobial Resistance (KONSAR)
Z, Liu J-H, Shen J. 2016. Emergence of plasmid-mediated colistin resis- Group. 2004. Metallo-beta-lactamase-producing Gram-negative bacilli
tance mechanism MCR-1 in animals and human beings in China: a in Korean nationwide surveillance of antimicrobial resistance group
microbiological and molecular biological study. Lancet Infect Dis 16: hospitals in 2003: continued prevalence of VIM-producing Pseudomo-
161–168. https://doi.org/10.1016/S1473-3099(15)00424-7. nas spp. and increase of IMP-producing Acinetobacter spp. Diagn Mi-
136. Abraham EP, Chain E. 1988. An enzyme from bacteria able to destroy crobiol Infect Dis 50:51–58. https://doi.org/10.1016/j.diagmicrobio
penicillin. 1940. Rev Infect Dis 10:677– 678. .2004.05.002.
137. Naas T, Oueslati S, Bonnin RA, Dabos ML, Zavala A, Dortet L, Retailleau 157. Cornaglia G, Giamarellou H, Rossolini GM. 2011. Metallo-beta-
P, Iorga BI. 2017. Beta-Lactamase Database (BLDB—structure and func- lactamases: a last frontier for beta-lactams? Lancet Infect Dis 11:
tion. J Enzyme Inhib Med Chem 32:917–919. https://doi.org/10.1080/ 381–393. https://doi.org/10.1016/S1473-3099(11)70056-1.
14756366.2017.1344235. 158. Jovcic B, Lepsanovic Z, Suljagic V, Rackov G, Begovic J, Topisirovic L,
138. Ambler RP. 1980. The structure of beta-lactamases. Philos Trans R Soc Kojic M. 2011. Emergence of NDM-1 metallo-beta-lactamase in Pseu-
Lond B Biol Sci 289:321–331. https://doi.org/10.1098/rstb.1980.0049. domonas aeruginosa clinical isolates from Serbia. Antimicrob Agents
139. Bush K, Jacoby GA. 2010. Updated functional classification of beta- Chemother 55:3929 –3931. https://doi.org/10.1128/AAC.00226-11.
lactamases. Antimicrob Agents Chemother 54:969 –976. https://doi 159. Jacoby GA. 2009. AmpC beta-lactamases. Clin Microbiol Rev 22:
.org/10.1128/AAC.01009-09. 161–182. https://doi.org/10.1128/CMR.00036-08.
140. Bush K, Bradford PA. 2019. Interplay between beta-lactamases and new 160. Pitout JDD, Peirano G, Kock MM, Strydom KA, Matsumura Y. 2020. The
beta-lactamase inhibitors. Nat Rev Microbiol 17:295–306. https://doi global ascendency of OXA-48-type carbapenemases. Clin Microbiol Rev
.org/10.1038/s41579-019-0159-8. 33:e00102-19. https://doi.org/10.1128/CMR.00102-19.
141. Richter SS, Doern GV, Heilmann KP, Miner S, Tendolkar S, Riahi F, 161. Poirel L, Bonnin RA, Nordmann P. 2012. Genetic features of the wide-
Diekema DJ. 2016. Detection and prevalence of penicillin-susceptible spread plasmid coding for the carbapenemase OXA-48. Antimicrob
Staphylococcus aureus in the United States in 2013. J Clin Microbiol Agents Chemother 56:559 –562. https://doi.org/10.1128/AAC.05289-11.
54:812– 814. https://doi.org/10.1128/JCM.03109-15. 162. Shaw KJ, Rather PN, Hare RS, Miller GH. 1993. Molecular genetics of
142. Sarti M, Campanile F, Sabia C, Santagati M, Gargiulo R, Stefani S. 2012. aminoglycoside resistance genes and familial relationships of the
Polyclonal diffusion of beta-lactamase-producing Enterococcus faecium. aminoglycoside-modifying enzymes. Microbiol Rev 57:138 –163.
J Clin Microbiol 50:169 –172. https://doi.org/10.1128/JCM.05640-11. 163. Kobayashi N, Alam M, Nishimoto Y, Urasawa S, Uehara N, Watanabe
143. Zscheck KK, Murray BE. 1991. Nucleotide sequence of the beta- N. 2001. Distribution of aminoglycoside resistance genes in recent
lactamase gene from Enterococcus faecalis HH22 and its similarity to clinical isolates of Enterococcus faecalis, Enterococcus faecium and
staphylococcal beta-lactamase genes. Antimicrob Agents Chemother Enterococcus avium. Epidemiol Infect 126:197–204. https://doi.org/
35:1736 –1740. https://doi.org/10.1128/aac.35.9.1736. 10.1017/s0950268801005271.
144. Wright G, McArthur AG. 2018. The comprehensive antibiotic resistance 164. Miller GH, Sabatelli FJ, Hare RS, Glupczynski Y, Mackey P, Shlaes D,
database. http://arpcard.mcmaster.ca/. Accessed 1 December 2019. Shimizu K, Shaw KJ. 1997. The most frequent aminoglycoside resistance
145. Gniadkowski M. 2008. Evolution of extended-spectrum beta-lactamases mechanisms— changes with time and geographic area: a reflection of
by mutation. Clin Microbiol Infect 14(Suppl 1):11–32. https://doi.org/ aminoglycoside usage patterns? Aminoglycoside Resistance Study
10.1111/j.1469-0691.2007.01854.x. Groups. Clin Infect Dis 24(Suppl 1):S46 –S62. https://doi.org/10.1093/
146. Canton R, Morosini MI, Martin O, de la Maza S, de la Pedrosa E. 2008. IRT clinids/24.supplement_1.s46.
and CMT beta-lactamases and inhibitor resistance. Clin Microbiol Infect 165. Ramirez MS, Tolmasky ME. 2010. Aminoglycoside modifying enzymes.
14:53– 62. https://doi.org/10.1111/j.1469-0691.2007.01849.x. Drug Resist Updat 13:151–171. https://doi.org/10.1016/j.drup.2010.08
147. Bradford PA, Bratu S, Urban C, Visalli M, Mariano N, Landman D, .003.
Rahal JJ, Brooks S, Cebular S, Quale J. 2004. Emergence of 166. Krause KM, Serio AW, Kane TR, Connolly LE. 2016. Aminoglycosides: an
carbapenem-resistant Klebsiella species possessing the class A overview. Cold Spring Harb Perspect Med 6:a027029. https://doi.org/
carbapenem-hydrolyzing KPC-2 and inhibitor-resistant TEM-30 beta- 10.1101/cshperspect.a027029.
lactamases in New York City. Clin Infect Dis 39:55– 60. https://doi 167. Castanheira M, Davis AP, Serio AW, Krause KM, Mendes RE. 2019. In vitro
.org/10.1086/421495. activity of plazomicin against Enterobacteriaceae isolates carrying
148. Nordmann P, Cuzon G, Naas T. 2009. The real threat of Klebsiella genes encoding aminoglycoside-modifying enzymes most common in
pneumoniae carbapenemase-producing bacteria. Lancet Infect Dis US Census divisions. Diagn Microbiol Infect Dis 94:73–77. https://doi
9:228 –236. https://doi.org/10.1016/S1473-3099(09)70054-4. .org/10.1016/j.diagmicrobio.2018.10.023.
149. Vena A, Castaldo N, Bassetti M. 2019. The role of new beta-lactamase 168. Costello SE, Deshpande LM, Davis AP, Mendes RE, Castanheira M. 2019.
inhibitors in Gram-negative infections. Curr Opin Infect Dis 32: Aminoglycoside-modifying enzyme and 16S ribosomal RNA methyl-
638 – 646. https://doi.org/10.1097/QCO.0000000000000600. transferase genes among a global collection of Gram-negative isolates.
150. Gottig S, Frank D, Mungo E, Nolte A, Hogardt M, Besier S, Wichelhaus J Glob Antimicrob Resist 16:278 –285. https://doi.org/10.1016/j.jgar
TA. 2019. Emergence of ceftazidime/avibactam resistance in KPC-3- .2018.10.020.
producing Klebsiella pneumoniae in vivo. J Antimicrob Chemother 74: 169. Almaghrabi R, Clancy CJ, Doi Y, Hao B, Chen L, Shields RK, Press EG,
3211–3216. https://doi.org/10.1093/jac/dkz330. Iovine NM, Townsend BM, Wagener MM, Kreiswirth B, Nguyen MH.
151. Zhang P, Shi Q, Hu H, Hong B, Wu X, Du X, Akova M, Yu Y. 2020. 2014. Carbapenem-resistant Klebsiella pneumoniae strains exhibit di-
Emergence of ceftazidime/avibactam resistance in carbapenem- versity in aminoglycoside-modifying enzymes, which exert differing
resistant Klebsiella pneumoniae in China. Clin Microbiol Infect 26: effects on plazomicin and other agents. Antimicrob Agents Chemother
124.e1–124.e4. https://doi.org/10.1016/j.cmi.2019.08.020. 58:4443– 4451. https://doi.org/10.1128/AAC.00099-14.
152. Lauretti L, Riccio ML, Mazzariol A, Cornaglia G, Amicosante G, Fontana R, 170. Hasani A, Sheikhalizadeh V, Ahangarzadeh Rezaee M, Rahmati-Yamchi
M, Hasani A, Ghotaslou R, Goli HR. 2016. Frequency of aminoglycoside- quinolone-DNA cleavage complex of type IIA topoisomerases. Nat
modifying enzymes and ArmA among different sequence groups of Struct Mol Biol 16:667– 669. https://doi.org/10.1038/nsmb.1604.
Acinetobacter baumannii in Iran. Microb Drug Resist 22:347–353. 189. Morais Cabral JH, Jackson AP, Smith CV, Shikotra N, Maxwell A, Lid-
https://doi.org/10.1089/mdr.2015.0254. dington RC. 1997. Crystal structure of the breakage-reunion domain of
171. Holbrook SYL, Garneau-Tsodikova S. 2018. Evaluation of aminoglyco- DNA gyrase. Nature 388:903–906. https://doi.org/10.1038/42294.
side and carbapenem resistance in a collection of drug-resistant Pseu- 190. Wohlkonig A, Chan PF, Fosberry AP, Homes P, Huang J, Kranz M,
domonas aeruginosa clinical isolates. Microb Drug Resist 24:1020 –1030. Leydon VR, Miles TJ, Pearson ND, Perera RL, Shillings AJ, Gwynn MN,
https://doi.org/10.1089/mdr.2017.0101. Bax BD. 2010. Structural basis of quinolone inhibition of type IIA
172. Libisch B, Poirel L, Lepsanovic Z, Mirovic V, Balogh B, Paszti J, Hunyadi topoisomerases and target-mediated resistance. Nat Struct Mol Biol
Z, Dobak A, Fuzi M, Nordmann P. 2008. Identification of PER-1 17:1152–1153. https://doi.org/10.1038/nsmb.1892.
extended-spectrum beta-lactamase producing Pseudomonas aerugi- 191. Schmitz FJ, Jones ME, Hofmann B, Hansen B, Scheuring S, Luckefahr M,
nosa clinical isolates of the international clonal complex CC11 from Fluit A, Verhoef J, Hadding U, Heinz HP, Kohrer K. 1998. Characteriza-
Hungary and Serbia. FEMS Immunol Med Microbiol 54:330 –338. tion of grlA, grlB, gyrA, and gyrB mutations in 116 unrelated isolates of
https://doi.org/10.1111/j.1574-695X.2008.00483.x. Staphylococcus aureus and effects of mutations on ciprofloxacin MIC.
173. Park CH, Robicsek A, Jacoby GA, Sahm D, Hooper DC. 2006. Prevalence Antimicrob Agents Chemother 42:1249 –1252.
in the United States of aac(6’)-Ib-cr encoding a ciprofloxacin-modifying 192. Yoshida H, Bogaki M, Nakamura M, Yamanaka LM, Nakamura S. 1991.
enzyme. Antimicrob Agents Chemother 50:3953–3955. https://doi.org/ Quinolone resistance-determining region in the DNA gyrase gyrB
10.1128/AAC.00915-06. gene of Escherichia coli. Antimicrob Agents Chemother 35:
174. Perilli M, Sabatini A, Pontieri E, Celenza G, Segatore B, Bottoni C, Bellio 1647–1650. https://doi.org/10.1128/aac.35.8.1647.
P, Mancini A, Marcoccia F, Brisdelli F, Amicosante G. 2015. OXA-23 193. Komp Lindgren P, Karlsson A, Hughes D. 2003. Mutation rate and
carbapenemase in multidrug-resistant Acinetobacter baumannii ST2 evolution of fluoroquinolone resistance in Escherichia coli isolates from
type: first identification in L’Aquila Hospital (Italy). Microb Drug Resist patients with urinary tract infections. Antimicrob Agents Chemother
21:97–101. https://doi.org/10.1089/mdr.2014.0056. 47:3222–3232. https://doi.org/10.1128/aac.47.10.3222-3232.2003.
175. Robicsek A, Strahilevitz J, Jacoby GA, Macielag M, Abbanat D, Park CH, 194. Robicsek A, Jacoby GA, Hooper DC. 2006. The worldwide emergence of
Bush K, Hooper DC. 2006. Fluoroquinolone-modifying enzyme: a new plasmid-mediated quinolone resistance. Lancet Infect Dis 6:629 – 640.
adaptation of a common aminoglycoside acetyltransferase. Nat Med https://doi.org/10.1016/S1473-3099(06)70599-0.
12:83– 88. https://doi.org/10.1038/nm1347. 195. Ruiz J. 2019. Transferable mechanisms of quinolone resistance from
176. Lakhundi S, Zhang K. 2018. Methicillin-resistant Staphylococcus aureus: 1998 onward. Clin Microbiol Rev 32:e00007-19. https://doi.org/10.1128/
molecular characterization, evolution, and epidemiology. Clin Microbiol CMR.00007-19.
Rev 31:e00020-18. https://doi.org/10.1128/CMR.00020-18. 196. Horinouchi S, Weisblum B. 1980. Posttranscriptional modification of
177. Chen FJ, Huang IW, Wang CH, Chen PC, Wang HY, Lai JF, Shiau YR, mRNA conformation: mechanism that regulates erythromycin-induced
resistance. Proc Natl Acad Sci U S A 77:7079 –7083. https://doi.org/10
Lauderdale TLY, TSAR Hospitals. 2012. mecA-positive Staphylococcus
.1073/pnas.77.12.7079.
aureus with low-level oxacillin MIC in Taiwan. J Clin Microbiol 50:
197. Weisblum B. 1995. Erythromycin resistance by ribosome modification.
1679 –1683. https://doi.org/10.1128/JCM.06711-11.
Antimicrob Agents Chemother 39:577–585. https://doi.org/10.1128/aac
178. Pu WX, Su Y, Li JX, Li CH, Yang ZQ, Deng HP, Ni CX. 2014. High
.39.3.577.
incidence of oxacillin-susceptible mecA-positive Staphylococcus aureus
198. Munita JM, Arias CA. 2016. Mechanisms of antibiotic resistance. Micro-
(OS-MRSA) associated with bovine mastitis in China. PLoS One
biol Spectr 4:VMBF-0016-2015. https://doi.org/10.1128/microbiolspec
9:e88134. https://doi.org/10.1371/journal.pone.0088134.
.VMBF-0016-2015.
179. Baig S, Johannesen TB, Overballe-Petersen S, Larsen J, Larsen AR,
199. Murphy E, Huwyler L, de Freire Bastos MDC. 1985. Transposon Tn554:
Stegger M. 2018. Novel SCCmec type XIII (9A) identified in an ST152
complete nucleotide sequence and isolation of transposition-defective
methicillin-resistant Staphylococcus aureus. Infect Genet Evol 61:74 –76.
and antibiotic-sensitive mutants. EMBO J 4:3357–3365.
https://doi.org/10.1016/j.meegid.2018.03.013.
200. Shaw KJ, Barbachyn MR. 2011. The oxazolidinones: past, present, and
180. Becker K, van Alen S, Idelevich EA, Schleimer N, Seggewiß J, Mellmann
future. Ann N Y Acad Sci 1241:48 –70. https://doi.org/10.1111/j.1749
A, Kaspar U, Peters G. 2018. Plasmid-encoded transferable mecB- -6632.2011.06330.x.
mediated methicillin resistance in Staphylococcus aureus. Emerg Infect 201. Locke JB, Zuill DE, Scharn CR, Deane J, Sahm DF, Denys GA, Goering RV,
Dis 24:242–248. https://doi.org/10.3201/eid2402.171074. Shaw KJ. 2014. Linezolid-resistant Staphylococcus aureus strain
181. Garcia-Alvarez L, Holden MT, Lindsay H, Webb CR, Brown DF, Curran 1128105, the first known clinical isolate possessing the cfr multidrug
MD, Walpole E, Brooks K, Pickard DJ, Teale C, Parkhill J, Bentley SD, resistance gene. Antimicrob Agents Chemother 58:6592– 6598. https://
Edwards GF, Girvan EK, Kearns AM, Pichon B, Hill RL, Larsen AR, Skov RL, doi.org/10.1128/AAC.03493-14.
Peacock SJ, Maskell DJ, Holmes MA. 2011. Meticillin-resistant Staphylo- 202. Mendes RE, Deshpande LM, Castanheira M, DiPersio J, Saubolle MA,
coccus aureus with a novel mecA homologue in human and bovine Jones RN. 2008. First report of cfr-mediated resistance to linezolid in
populations in the UK and Denmark: a descriptive study. Lancet Infect human staphylococcal clinical isolates recovered in the United States.
Dis 11:595– 603. https://doi.org/10.1016/S1473-3099(11)70126-8. Antimicrob Agents Chemother 52:2244 –2246. https://doi.org/10.1128/
182. Paterson GK. 13 January 2020. Low prevalence of livestock-associated AAC.00231-08.
methicillin-resistant Staphylococcus aureus clonal complex 398 and 203. Locke JB, Morales G, Hilgers M, Kedar GC, Rahawi S, Jose Picazo J, Shaw
mecC MRSA among human isolates in North-West England. J Appl KJ, Stein JL. 2010. Elevated linezolid resistance in clinical cfr-positive
Microbiol https://doi.org/10.1111/jam.14578. Staphylococcus aureus isolates is associated with co-occurring muta-
183. Paterson GK, Harrison EM, Holmes MA. 2014. The emergence of mecC tions in ribosomal protein L3. Antimicrob Agents Chemother 54:
methicillin-resistant Staphylococcus aureus. Trends Microbiol 22:42– 47. 5352–5355. https://doi.org/10.1128/AAC.00714-10.
https://doi.org/10.1016/j.tim.2013.11.003. 204. Cox G, Ejim L, Stogios PJ, Koteva K, Bordeleau E, Evdokimova E, Sieron
184. Arias CA, Murray BE. 2012. The rise of the Enterococcus: beyond van- AO, Savchenko A, Serio AW, Krause KM, Wright GD. 2018. Plazomicin
comycin resistance. Nat Rev Microbiol 10:266 –278. https://doi.org/10 retains antibiotic activity against most aminoglycoside modifying en-
.1038/nrmicro2761. zymes. ACS Infect Dis 4:980 –987. https://doi.org/10.1021/acsinfecdis
185. Zapun A, Contreras-Martel C, Vernet T. 2008. Penicillin-binding proteins .8b00001.
and beta-lactam resistance. FEMS Microbiol Rev 32:361–385. https:// 205. Doi Y, Wachino J-I, Arakawa Y. 2016. Aminoglycoside resistance: the
doi.org/10.1111/j.1574-6976.2007.00095.x. emergence of acquired 16S ribosomal RNA methyltransferases. Infect
186. Poirel L, Nordmann P. 2006. Carbapenem resistance in Acinetobacter Dis Clin North Am 30:523–537. https://doi.org/10.1016/j.idc.2016.02
baumannii: mechanisms and epidemiology. Clin Microbiol Infect 12: .011.
826 – 836. https://doi.org/10.1111/j.1469-0691.2006.01456.x. 206. Kristich CJ, Rice LB, Arias CA. 2014. Enterococcal infection—treatment
187. Hooper DC. 2001. Emerging mechanisms of fluoroquinolone resistance. and antibiotic resistance. In Gilmore MS, Clewell DB, Ike Y, Shankar N
Emerg Infect Dis 7:337–341. https://doi.org/10.3201/eid0702.010239. (ed), Enterococci: from commensals to leading causes of drug resistant
188. Laponogov I, Sohi MK, Veselkov DA, Pan XS, Sawhney R, Thompson AW, infection. Massachusetts Eye and Ear Infirmary, Boston, MA.
McAuley KE, Fisher LM, Sanderson MR. 2009. Structural insight into the 207. Arthur M, Molinas C, Depardieu F, Courvalin P. 1993. Characterization of
Tn1546, a Tn3-related transposon conferring glycopeptide resistance crob Agents Chemother 50:428 – 438. https://doi.org/10.1128/AAC
by synthesis of depsipeptide peptidoglycan precursors in Enterococcus .50.2.428-438.2006.
faecium BM4147. J Bacteriol 175:117–127. https://doi.org/10.1128/jb 223. Cui L, Ma X, Sato K, Okuma K, Tenover FC, Mamizuka EM, Gemmell CG,
.175.1.117-127.1993. Kim MN, Ploy MC, El-Solh N, Ferraz V, Hiramatsu K. 2003. Cell wall
208. Carias LL, Rudin SD, Donskey CJ, Rice LB. 1998. Genetic linkage and thickening is a common feature of vancomycin resistance in Staphylo-
cotransfer of a novel, vanB-containing transposon (Tn5382) and a coccus aureus. J Clin Microbiol 41:5–14. https://doi.org/10.1128/JCM.41
low-affinity penicillin-binding protein 5 gene in a clinical vancomycin- .1.5-14.2003.
resistant Enterococcus faecium isolate. J Bacteriol 180:4426 – 4434. 224. Renzoni A, Barras C, Francois P, Charbonnier Y, Huggler E, Garzoni C,
https://doi.org/10.1128/JB.180.17.4426-4434.1998. Kelley WL, Majcherczyk P, Schrenzel J, Lew DP, Vaudaux P. 2006.
209. Quintiliani R, Jr, Courvalin P. 1996. Characterization of Tn1547, a com- Transcriptomic and functional analysis of an autolysis-deficient,
posite transposon flanked by the IS16 and IS256-like elements, that teicoplanin-resistant derivative of methicillin-resistant Staphylococcus
confers vancomycin resistance in Enterococcus faecalis BM4281. Gene aureus. Antimicrob Agents Chemother 50:3048 –3061. https://doi.org/
172:1– 8. https://doi.org/10.1016/0378-1119(96)00110-2. 10.1128/AAC.00113-06.
210. Perichon B, Courvalin P. 2009. VanA-type vancomycin-resistant Staph- 225. Howden BP, Peleg AY, Stinear TP. 2014. The evolution of vancomycin
ylococcus aureus. Antimicrob Agents Chemother 53:4580–4587. https:// intermediate Staphylococcus aureus (VISA) and heterogenous-VISA. In-
doi.org/10.1128/AAC.00346-09. fect Genet Evol 21:575–582. https://doi.org/10.1016/j.meegid.2013.03
211. McGuinness WA, Malachowa N, DeLeo FR. 2017. Vancomycin resistance .047.
in Staphylococcus aureus. Yale J Biol Med 90:269 –281. 226. Zhang SS, Sun XX, Chang WJ, Dai YY, Ma XL. 2015. Systematic review
212. Melo-Cristino J, Resina C, Manuel V, Lito L, Ramirez M. 2013. First case and meta-analysis of the epidemiology of vancomycin-intermediate
of infection with vancomycin-resistant Staphylococcus aureus in Eu- and heterogeneous vancomycin-intermediate Staphylococcus aureus
rope. Lancet 382:205. https://doi.org/10.1016/S0140-6736(13)61219-2. isolates. PLoS One 10:e0136082. https://doi.org/10.1371/journal.pone
213. Panesso D, Planet PJ, Diaz L, Hugonnet JE, Tran TT, Narechania A, .0136082.
Munita JM, Rincon S, Carvajal LP, Reyes J, Londono A, Smith H, Sebra R, 227. Miller WR, Bayer AS, Arias CA. 2016. Mechanism of action and
Deikus G, Weinstock GM, Murray BE, Rossi F, Arthur M, Arias CA. 2015. resistance to daptomycin in Staphylococcus aureus and enterococci.
Methicillin-susceptible, vancomycin-resistant Staphylococcus aureus, Cold Spring Harb Perspect Med 6:a026997. https://doi.org/10.1101/
Brazil. Emerg Infect Dis 21:1844 –1848. https://doi.org/10.3201/eid2110 cshperspect.a026997.
.141914. 228. Kidd TJ, Mills G, Sa-Pessoa J, Dumigan A, Frank CG, Insua JL, Ingram R,
214. Rossi F, Diaz L, Wollam A, Panesso D, Zhou Y, Rincon S, Narechania A, Hobley L, Bengoechea JA. 2017. A Klebsiella pneumoniae antibiotic
Xing G, Di Gioia TSR, Doi A, Tran TT, Reyes J, Munita JM, Carvajal LP, resistance mechanism that subdues host defences and promotes vir-
Hernandez-Roldan A, Brandão D, van der Heijden IM, Murray BE, Planet ulence. EMBO Mol Med 9:430 – 447. https://doi.org/10.15252/emmm
PJ, Weinstock GM, Arias CA. 2014. Transferable vancomycin resistance .201607336.
229. Poirel L, Jayol A, Nordmann P. 2017. Polymyxins: antibacterial activity,
in a community-associated MRSA lineage. N Engl J Med 370:
susceptibility testing, and resistance mechanisms encoded by plasmids
1524 –1531. https://doi.org/10.1056/NEJMoa1303359.
or chromosomes. Clin Microbiol Rev 30:557–596. https://doi.org/10
215. Division of Healthcare Quality Promotion, Centers for Disease Control
.1128/CMR.00064-16.
and Prevention. 2015. Investigation and control of vancomycin-
230. Wright MS, Suzuki Y, Jones MB, Marshall SH, Rudin SD, van Duin D, Kaye
resistant Staphylococcus aureus (VRSA): 2015 update. https://www.cdc
K, Jacobs MR, Bonomo RA, Adams MD. 2015. Genomic and transcrip-
.gov/hai/pdfs/VRSA-Investigation-Guide-05_12_2015.pdf. Accessed 5
tomic analyses of colistin-resistant clinical isolates of Klebsiella pneu-
February 2020.
moniae reveal multiple pathways of resistance. Antimicrob Agents
216. Foucault ML, Courvalin P, Grillot-Courvalin C. 2009. Fitness cost of
Chemother 59:536 –543. https://doi.org/10.1128/AAC.04037-14.
VanA-type vancomycin resistance in methicillin-resistant Staphylococ-
231. Quinn JP, Dudek EJ, DiVincenzo CA, Lucks DA, Lerner SA. 1986. Emer-
cus aureus. Antimicrob Agents Chemother 53:2354 –2359. https://doi
gence of resistance to imipenem during therapy for Pseudomonas
.org/10.1128/AAC.01702-08.
aeruginosa infections. J Infect Dis 154:289 –294. https://doi.org/10
217. Zhu WM, Clark N, Patel JB. 2013. pSK41-like plasmid is necessary for
.1093/infdis/154.2.289.
Inc18-like vanA plasmid transfer from Enterococcus faecalis to Staphy- 232. Hasdemir UO, Chevalier J, Nordmann P, Pages JM. 2004. Detection and
lococcus aureus in vitro. Antimicrob Agents Chemother 57:212–219. prevalence of active drug efflux mechanism in various multidrug-
https://doi.org/10.1128/AAC.01587-12. resistant Klebsiella pneumoniae strains from Turkey. J Clin Microbiol
218. Zhu W, Murray PR, Huskins WC, Jernigan JA, McDonald LC, Clark NC, 42:2701–2706. https://doi.org/10.1128/JCM.42.6.2701-2706.2004.
Anderson KF, McDougal LK, Hageman JC, Olsen-Rasmussen M, Frace M, 233. Pages JM, James CE, Winterhalter M. 2008. The porin and the perme-
Alangaden GJ, Chenoweth C, Zervos MJ, Robinson-Dunn B, Schrecken- ating antibiotic: a selective diffusion barrier in Gram-negative bacteria.
berger PC, Reller LB, Rudrik JT, Patel JB. 2010. Dissemination of an Nat Rev Microbiol 6:893–903. https://doi.org/10.1038/nrmicro1994.
Enterococcus Inc18-like vanA plasmid associated with vancomycin- 234. Li XZ, Plesiat P, Nikaido H. 2015. The challenge of efflux-mediated
resistant Staphylococcus aureus. Antimicrob Agents Chemother 54: antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev
4314 – 4320. https://doi.org/10.1128/AAC.00185-10. 28:337– 418. https://doi.org/10.1128/CMR.00117-14.
219. Kos VN, Desjardins CA, Griggs A, Cerqueira G, Van Tonder A, Holden 235. Davin-Regli A, Lavigne JP, Pages JM. 2019. Enterobacter spp.: update on
MTG, Godfrey P, Palmer KL, Bodi K, Mongodin EF, Wortman J, Feldgar- taxonomy, clinical aspect, and emerging antimicrobial resistance. Clin
den M, Lawley T, Gill SR, Haas BJ, Birren B, Gilmore MS. 2012. Compar- Microbiol Rev 32:e00002-19. https://doi.org/10.1128/CMR.00002-19.
ative genomics of vancomycin-resistant Staphylococcus aureus strains 236. Masi M, Winterhalter M, Pages JM. 2019. Outer membrane porins.
and their positions within the clade most commonly associated with Subcell Biochem 92:79 –123. https://doi.org/10.1007/978-3-030-18768
methicillin-resistant S. aureus hospital-acquired infection in the United -2_4.
States. mBio 3:e00112-12. https://doi.org/10.1128/mBio.00112-12. 237. Hassan KA, Liu Q, Henderson PJ, Paulsen IT. 2015. Homologs of the
220. Howden BP, Davies JK, Johnson PD, Stinear TP, Grayson ML. 2010. Acinetobacter baumannii AceI transporter represent a new family of
Reduced vancomycin susceptibility in Staphylococcus aureus, including bacterial multidrug efflux systems. mBio 6:e01982-14. https://doi.org/
vancomycin-intermediate and heterogeneous vancomycin- 10.1128/mBio.01982-14.
intermediate strains: resistance mechanisms, laboratory detection, and 238. Su CC, Morgan CE, Kambakam S, Rajavel M, Scott H, Huang W, Emerson
clinical implications. Clin Microbiol Rev 23:99 –139. https://doi.org/10 CC, Taylor DJ, Stewart PL, Bonomo RA, Yu EW. 2019. Cryo-electron
.1128/CMR.00042-09. microscopy structure of an Acinetobacter baumannii multidrug efflux
221. Boyle-Vavra S, Challapalli M, Daum RS. 2003. Resistance to autolysis in pump. mBio 10:e01295-19. https://doi.org/10.1128/mBio.01295-19.
vancomycin-selected Staphylococcus aureus isolates precedes 239. Leus IV, Weeks JW, Bonifay V, Smith L, Richardson S, Zgurskaya HI. 2018.
vancomycin-intermediate resistance. Antimicrob Agents Chemother Substrate specificities and efflux efficiencies of RND efflux pumps of
47:2036 –2039. https://doi.org/10.1128/aac.47.6.2036-2039.2003. Acinetobacter baumannii. J Bacteriol 200:e00049-18. https://doi.org/10
222. Cui L, Iwamoto A, Lian JQ, Neoh HM, Maruyama T, Horikawa Y, .1128/JB.00049-18.
Hiramatsu K. 2006. Novel mechanism of antibiotic resistance origi- 240. Pagdepanichkit S, Tribuddharat C, Chuanchuen R. 2016. Distribution
nating in vancomycin-intermediate Staphylococcus aureus. Antimi- and expression of the Ade multidrug efflux systems in Acinetobacter
baumannii clinical isolates. Can J Microbiol 62:794 – 801. https://doi intracellular survival in macrophages. Cell Microbiol 18:831– 843.
.org/10.1139/cjm-2015-0730. https://doi.org/10.1111/cmi.12556.
241. Wong MHY, Chan EWC, Chen S. 2015. Evolution and dissemination of 259. Fraunholz M, Sinha B. 2012. Intracellular Staphylococcus aureus: live-in
OqxAB-like efflux pumps, an emerging quinolone resistance determi- and let die. Front Cell Infect Microbiol 2:43. https://doi.org/10.3389/
nant among members of Enterobacteriaceae. Antimicrob Agents Che- fcimb.2012.00043.
mother 59:3290 –3297. https://doi.org/10.1128/AAC.00310-15. 260. Lowy FD. 2000. Is Staphylococcus aureus an intracellular pathogen? Trends
242. Veleba M, Schneiders T. 2012. Tigecycline resistance can occur inde- Microbiol 8:341–343. https://doi.org/10.1016/s0966-842x(00)01803-5.
pendently of the ramA gene in Klebsiella pneumoniae. Antimicrob 261. Lehar SM, Pillow T, Xu M, Staben L, Kajihara KK, Andlen RV, DePalatis L,
Agents Chemother 56:4466 – 4467. https://doi.org/10.1128/AAC.06224 Raab H, Hazenbos WL, Morisaki JH, Kim J, Park S, Darwish M, Lee BC,
-11. Hernandez H, Loyet KM, Lupardus P, Fong RN, Yan DH, Halouni CC, Luis
243. Hill D, Rose B, Pajkos A, Robinson M, Bye P, Bell S, Elkins M, Thompson E, Khalfin Y, Plise E, Heong JC, Lyssikatos JP, Strandh M, Koefoed K,
B, MacLeod C, Aaron SD, Harbour C. 2005. Antibiotic susceptibilities of Andersen PS, Flygare JA, Tan MW, Brown EJ, Ariathasan SM. 2015. Novel
Pseudomonas aeruginosa isolates derived from patients with cystic antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature
fibrosis under aerobic, anaerobic, and biofilm conditions. J Clin Micro- 527:323–328. https://doi.org/10.1038/nature16057.
biol 43:5085–5090. https://doi.org/10.1128/JCM.43.10.5085-5090.2005. 262. Partridge SR, Kwong SM, Firth N, Jensen SO. 2018. Mobile genetic
244. Nickel JC, Ruseska I, Wright JB, Costerton JW. 1985. Tobramycin resis- elements associated with antimicrobial resistance. Clin Microbiol Rev
tance of Pseudomonas aeruginosa cells growing as a biofilm on urinary 31:e00088-17. https://doi.org/10.1128/CMR.00088-17.
catheter material. Antimicrob Agents Chemother 27:619 – 624. https:// 263. Siguier P, Gourbeyre E, Varani A, Ton-Hoang B, Chandler M. 2015.
doi.org/10.1128/aac.27.4.619. Everyman’s guide to bacterial insertion sequences. Microbiol Spectr
245. Percival SL, Suleman L, Vuotto C, Donelli G. 2015. Healthcare-associated 3:MDNA3-0030-2014. https://doi.org/10.1128/microbiolspec.MDNA3
infections, medical devices and biofilms: risk, tolerance and control. J -0030-2014.
Med Microbiol 64:323–334. https://doi.org/10.1099/jmm.0.000032. 264. Vandecraen J, Chandler M, Aertsen A, Van Houdt R. 2017. The impact of
246. Hall CW, Mah TF. 2017. Molecular mechanisms of biofilm-based anti- insertion sequences on bacterial genome plasticity and adaptability.
biotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol Crit Rev Microbiol 43:709 –730. https://doi.org/10.1080/1040841X.2017
Rev 41:276 –301. https://doi.org/10.1093/femsre/fux010. .1303661.
247. Schembri MA, Hjerrild L, Gjermansen M, Klemm P. 2003. Differential 265. Haniford DB, Ellis MJ. 2015. Transposons Tn10 and Tn5. Microbiol Spectr
expression of the Escherichia coli autoaggregation factor antigen 43. 3:MDNA3-0002-2014. https://doi.org/10.1128/microbiolspec.MDNA3-0002
J Bacteriol 185:2236 –2242. https://doi.org/10.1128/jb.185.7.2236 -2014.
-2242.2003. 266. Bennett PM. 2008. Plasmid encoded antibiotic resistance: acquisition
248. Secor PR, Michaels LA, Ratjen A, Jennings LK, Singh PK. 2018. Entropi- and transfer of antibiotic resistance genes in bacteria. Br J Pharmacol
cally driven aggregation of bacteria by host polymers promotes anti- 153(Suppl 1):S347–S357. https://doi.org/10.1038/sj.bjp.0707607.
biotic tolerance in Pseudomonas aeruginosa. Proc Natl Acad Sci U S A 267. Weigel LM, Clewell DB, Gill SR, Clark NC, McDougal LK, Flannagan SE,
115:10780 –10785. https://doi.org/10.1073/pnas.1806005115. Kolonay JF, Shetty J, Killgore GE, Tenover FC. 2003. Genetic analysis of
249. Secor PR, Sweere JM, Michaels LA, Malkovskiy AV, Lazzareschi D, a high-level vancomycin-resistant isolate of Staphylococcus aureus. Sci-
Katznelson E, Rajadas J, Birnbaum ME, Arrigoni A, Braun KR, Evanko SP, ence 302:1569 –1571. https://doi.org/10.1126/science.1090956.
Stevens DA, Kaminsky W, Singh PK, Parks WC, Bollyky PL. 2015. Fila- 268. Partridge SR. 2015. What’s in a name? ISSwi1 corresponds to trans-
mentous bacteriophage promote biofilm assembly and function. Cell posons related to Tn2 and Tn3. mBio 6:e01344-15. https://doi.org/10
Host Microbe 18:549 –559. https://doi.org/10.1016/j.chom.2015.10.013. .1128/mBio.01344-15.
250. Weigel LM, Donlan RM, Shin DH, Jensen B, Clark NC, McDougal LK, Zhu 269. Nicolas E, Lambin M, Dandoy D, Galloy C, Nguyen N, Oger CA, Hallet
W, Musser KA, Thompson J, Kohlerschmidt D, Dumas N, Limberger RJ, B. 2015. The Tn3-family of replicative transposons. Microbiol Spectr
Patel JB. 2007. High-level vancomycin-resistant Staphylococcus aureus 3:MDNA3-0060-2014. https://doi.org/10.1128/microbiolspec.MDNA3
isolates associated with a polymicrobial biofilm. Antimicrob Agents -0060-2014.
Chemother 51:231–238. https://doi.org/10.1128/AAC.00576-06. 270. Post V, White PA, Hall RM. 2010. Evolution of AbaR-type genomic
251. Brauner A, Fridman O, Gefen O, Balaban NQ. 2016. Distinguishing resistance islands in multiply antibiotic-resistant Acinetobacter bau-
between resistance, tolerance and persistence to antibiotic treat- mannii. J Antimicrob Chemother 65:1162–1170. https://doi.org/10
ment. Nat Rev Microbiol 14:320 –330. https://doi.org/10.1038/nrmicro .1093/jac/dkq095.
.2016.34. 271. Hamidian M, Hall RM. 2017. Origin of the AbGRI1 antibiotic resistance
252. Balaban NQ, Helaine S, Lewis K, Ackermann M, Aldridge B, Andersson island found in the comM gene of Acinetobacter baumannii GC2 iso-
DI, Brynildsen MP, Bumann D, Camilli A, Collins JJ, Dehio C, Fortune S, lates. J Antimicrob Chemother 72:2944 –2947. https://doi.org/10.1093/
Ghigo JM, Hardt WD, Harms A, Heinemann M, Hung DT, Jenal U, Levin jac/dkx206.
BR, Michiels J, Storz G, Tan MW, Tenson T, Van Melderen L, Zinkernagel 272. Gregory PD, Lewis RA, Curnock SP, Dyke KG. 1997. Studies of the
A. 2019. Definitions and guidelines for research on antibiotic persis- repressor (BlaI) of beta-lactamase synthesis in Staphylococcus aureus.
tence. Nat Rev Microbiol 17:441– 448. https://doi.org/10.1038/s41579 Mol Microbiol 24:1025–1037. https://doi.org/10.1046/j.1365-2958.1997
-019-0196-3. .4051770.x.
253. Liu JF, Gefen O, Ronin I, Bar-Meir M, Balaban NQ. 2020. Effect of 273. Poirel L, Lartigue MF, Decousser JW, Nordmann P. 2005. ISEcp1B-
tolerance on the evolution of antibiotic resistance under drug combi- mediated transposition of blaCTX-M in Escherichia coli. Antimicrob
nations. Science 367:200 –204. https://doi.org/10.1126/science.aaj2191. Agents Chemother 49:447– 450. https://doi.org/10.1128/AAC.49.1.447
254. Brauner A, Shoresh N, Fridman O, Balaban NQ. 2017. An experimental -450.2005.
framework for quantifying bacterial tolerance. Biophys J 112: 274. Karim A, Poirel L, Nagarajan S, Nordmann P. 2001. Plasmid-mediated
2664 –2671. https://doi.org/10.1016/j.bpj.2017.05.014. extended-spectrum beta-lactamase (CTX-M-3 like) from India and gene
255. Yan J, Bassler BL. 2019. Surviving as a community: antibiotic tolerance association with insertion sequence ISEcp1. FEMS Microbiol Lett 201:
and persistence in bacterial biofilms. Cell Host Microbe 26:15–21. 237–241. https://doi.org/10.1111/j.1574-6968.2001.tb10762.x.
https://doi.org/10.1016/j.chom.2019.06.002. 275. Bauernfeind A, Schneider I, Jungwirth R, Sahly H, Ullmann U. 1999. A
256. Cohen NR, Lobritz MA, Collins JJ. 2013. Microbial persistence and the novel type of AmpC beta-lactamase, ACC-1, produced by a Klebsiella
road to drug resistance. Cell Host Microbe 13:632– 642. https://doi.org/ pneumoniae strain causing nosocomial pneumonia. Antimicrob Agents
10.1016/j.chom.2013.05.009. Chemother 43:1924 –1931.
257. Cano V, March C, Insua JL, Aguilo N, Llobet E, Moranta D, Regueiro V, 276. Zowawi HM, Forde BM, Alfaresi M, Alzarouni A, Farahat Y, Chong TM,
Brennan GP, Millan-Lou MI, Martin C, Garmendia J, Bengoechea JA. Yin WF, Chan KG, Li J, Schembri MA, Beatson SA, Paterson DL. 2015.
2015. Klebsiella pneumoniae survives within macrophages by avoiding Stepwise evolution of pandrug-resistance in Klebsiella pneumoniae. Sci
delivery to lysosomes. Cell Microbiol 17:1537–1560. https://doi.org/10 Rep 5:15082. https://doi.org/10.1038/srep15082.
.1111/cmi.12466. 277. Chen YG, Qu TT, Yu YS, Zhou JY, Li LJ. 2006. Insertion sequence
258. Zou J, Shankar N. 2016. The opportunistic pathogen Enterococcus ISEcp1-like element connected with a novel aph(2⬙) allele [aph(2⬙)-Ie]
faecalis resists phagosome acidification and autophagy to promote conferring high-level gentamicin resistance and a novel streptomycin
adenylyltransferase gene in Enterococcus. J Med Microbiol 55: a multidrug resistant Escherichia coli clone. Proc Natl Acad Sci U S A
1521–1525. https://doi.org/10.1099/jmm.0.46702-0. 111:5694 –5699. https://doi.org/10.1073/pnas.1322678111.
278. Partridge SR. 2016. Mobilization of blaBKC-1 by ISKpn23? Antimi- 297. Bauernfeind A, Chong Y, Schweighart S. 1989. Extended broad-spectrum
crob Agents Chemother 60:5102–5104. https://doi.org/10.1128/AAC beta-lactamase in Klebsiella pneumoniae including resistance to cephamy-
.00785-16. cins. Infection 17:316–321. https://doi.org/10.1007/bf01650718.
279. Harmer CJ, Hall RM. 2016. IS26-mediated formation of transposons 298. Philippon A, Arlet G, Jacoby GA. 2002. Plasmid-determined AmpC-type
carrying antibiotic resistance genes. mSphere 1:e00038-16. https://doi beta-lactamases. Antimicrob Agents Chemother 46:1–11. https://doi
.org/10.1128/mSphere.00038-16. .org/10.1128/aac.46.1.1-11.2002.
280. Toleman MA, Bennett PM, Walsh TR. 2006. ISCR elements: novel gene- 299. Hennequin C, Chlilek A, Beyrouthy R, Bonnet R, Robin F. 2018. Diversity
capturing systems of the 21st century? Microbiol Mol Biol Rev 70: of DHA-1-encoding plasmids in Klebsiella pneumoniae isolates from 16
296 –316. https://doi.org/10.1128/MMBR.00048-05. French hospitals. J Antimicrob Chemother 73:2981–2989. https://doi
281. Lee CH, Chu C, Liu JW, Chen YS, Chiu CJ, Su LH. 2007. Collateral damage .org/10.1093/jac/dky285.
of flomoxef therapy: in vivo development of porin deficiency and 300. Deshpande LM, Jones RN, Fritsche TR, Sader HS. 2006. Occurrence and
acquisition of blaDHA-1 leading to ertapenem resistance in a clinical characterization of carbapenemase-producing Enterobacteriaceae: re-
isolate of Klebsiella pneumoniae producing CTX-M-3 and SHV-5 beta- port from the SENTRY antimicrobial surveillance program (2000 –2004).
lactamases. J Antimicrob Chemother 60:410 – 413. https://doi.org/10 Microb Drug Resist 12:223–230. https://doi.org/10.1089/mdr.2006.12
.1093/jac/dkm215. .223.
282. Cannatelli A, Giani T, D’Andrea MM, Di Pilato V, Arena F, Conte V, 301. Walsh TR, Weeks J, Livermore DM, Toleman MA. 2011. Dissemination
Tryfinopoulou K, Vatopoulos A, Rossolini GM, COLGRIT Study Group. of NDM-1 positive bacteria in the New Delhi environment and its
2014. MgrB inactivation is a common mechanism of colistin resis- implications for human health: an environmental point prevalence
tance in KPC-producing Klebsiella pneumoniae of clinical origin. study. Lancet Infect Dis 11:355–362. https://doi.org/10.1016/S1473
Antimicrob Agents Chemother 58:5696 –5703. https://doi.org/10 -3099(11)70059-7.
.1128/AAC.03110-14. 302. Towner KJ, Evans B, Villa L, Levi K, Hamouda A, Amyes SG, Carattoli
283. Chen TL, Wu RC, Shaio MF, Fung CP, Cho WL. 2008. Acquisition of a A. 2011. Distribution of intrinsic plasmid replicase genes and their
plasmid-borne blaOXA-58 gene with an upstream IS1008 insertion association with carbapenem-hydrolyzing class D beta-lactamase
conferring a high level of carbapenem resistance to Acinetobacter genes in European clinical isolates of Acinetobacter baumannii. An-
baumannii. Antimicrob Agents Chemother 52:2573–2580. https://doi timicrob Agents Chemother 55:2154 –2159. https://doi.org/10.1128/
.org/10.1128/AAC.00393-08. AAC.01661-10.
284. Aubert D, Naas T, Heritier C, Poirel L, Nordmann P. 2006. Functional 303. Lean SS, Yeo CC. 2017. Small, enigmatic plasmids of the nosocomial
characterization of IS1999, an IS4 family element involved in mobiliza- pathogen, Acinetobacter baumannii: good, bad, who knows? Front
tion and expression of beta-lactam resistance genes. J Bacteriol 188: Microbiol 8:1547. https://doi.org/10.3389/fmicb.2017.01547.
304. Hamidian M, Nigro SJ, Hall RM. 2012. Variants of the gentamicin and
6506 – 6514. https://doi.org/10.1128/JB.00375-06.
tobramycin resistance plasmid pRAY are widely distributed in Acineto-
285. Aubert D, Naas T, Nordmann P. 2003. IS1999 increases expression of
bacter. J Antimicrob Chemother 67:2833–2836. https://doi.org/10.1093/
the extended-spectrum beta-lactamase VEB-1 in Pseudomonas
jac/dks318.
aeruginosa. J Bacteriol 185:5314 –5319. https://doi.org/10.1128/jb
305. Hamidian M, Holt KE, Pickard D, Dougan G, Hall RM. 2014. A GC1
.185.17.5314-5319.2003.
Acinetobacter baumannii isolate carrying AbaR3 and the aminoglyco-
286. Depardieu F, Podglajen I, Leclercq R, Collatz E, Courvalin P. 2007. Modes
side resistance transposon TnaphA6 in a conjugative plasmid. J Anti-
and modulations of antibiotic resistance gene expression. Clin Micro-
microb Chemother 69:955–958. https://doi.org/10.1093/jac/dkt454.
biol Rev 20:79 –114. https://doi.org/10.1128/CMR.00015-06.
306. Hamidian M, Hall RM. 2014. pACICU2 is a conjugative plasmid of
287. Naas T, Philippon L, Poirel L, Ronco E, Nordmann P. 1999. An SHV-
Acinetobacter carrying the aminoglycoside resistance transposon Tna-
derived extended-spectrum beta-lactamase in Pseudomonas aerugi-
phA6. J Antimicrob Chemother 69:1146 –1148. https://doi.org/10.1093/
nosa. Antimicrob Agents Chemother 43:1281–1284.
jac/dkt488.
288. Lee KY, Hopkins JD, Syvanen M. 1990. Direct involvement of IS26 in an
307. Hu H, Hu Y, Pan Y, Liang H, Wang H, Wang X, Hao Q, Yang X, Yang X,
antibiotic resistance operon. J Bacteriol 172:3229 –3236. https://doi Xiao X, Luan C, Yang Y, Cui Y, Yang R, Gao GF, Song Y, Zhu B. 2012.
.org/10.1128/jb.172.6.3229-3236.1990. Novel plasmid and its variant harboring both a bla(NDM-1) gene and
289. Aminov RI. 2011. Horizontal gene exchange in environmental microbi- type IV secretion system in clinical isolates of Acinetobacter lwoffii.
ota. Front Microbiol 2:158. https://doi.org/10.3389/fmicb.2011.00158. Antimicrob Agents Chemother 56:1698 –1702. https://doi.org/10.1128/
290. Shintani M, Sanchez ZK, Kimbara K. 2015. Genomics of microbial AAC.06199-11.
plasmids: classification and identification based on replication and 308. Jacoby GA, Sutton L, Knobel L, Mammen P. 1983. Properties of IncP-2
transfer systems and host taxonomy. Front Microbiol 6:242. https://doi plasmids of Pseudomonas spp. Antimicrob Agents Chemother 24:
.org/10.3389/fmicb.2015.00242. 168 –175. https://doi.org/10.1128/aac.24.2.168.
291. Guglielmini J, Quintais L, Garcillan-Barcia MP, de la Cruz F, Rocha EP. 309. Liu J, Yang L, Chen D, Peters BM, Li L, Li B, Xu Z, Shirtliff ME. 2018.
2011. The repertoire of ICE in prokaryotes underscores the unity, Complete sequence of pBM413, a novel multidrug resistance mega-
diversity, and ubiquity of conjugation. PLoS Genet 7:e1002222. https:// plasmid carrying qnrVC6 and blaIMP-45 from Pseudomonas aerugi-
doi.org/10.1371/journal.pgen.1002222. nosa. Int J Antimicrob Agents 51:145–150. https://doi.org/10.1016/
292. Thomas CM, Nielsen KM. 2005. Mechanisms of, and barriers to, hori- j.ijantimicag.2017.09.008.
zontal gene transfer between bacteria. Nat Rev Microbiol 3:711–721. 310. Xiong J, Alexander DC, Ma JH, Deraspe M, Low DE, Jamieson FB, Roy
https://doi.org/10.1038/nrmicro1234. PH. 2013. Complete sequence of pOZ176, a 500-kilobase IncP-2 plas-
293. Carattoli A. 2009. Resistance plasmid families in Enterobacteriaceae. mid encoding IMP-9-mediated carbapenem resistance, from outbreak
Antimicrob Agents Chemother 53:2227–2238. https://doi.org/10.1128/ isolate Pseudomonas aeruginosa 96. Antimicrob Agents Chemother
AAC.01707-08. 57:3775–3782. https://doi.org/10.1128/AAC.00423-13.
294. Rozwandowicz M, Brouwer MSM, Fischer J, Wagenaar JA, Gonzalez- 311. Botelho J, Grosso F, Quinteira S, Mabrouk A, Peixe L. 2017. The com-
Zorn B, Guerra B, Mevius DJ, Hordijk J. 2018. Plasmids carrying antimi- plete nucleotide sequence of an IncP-2 megaplasmid unveils a mosaic
crobial resistance genes in Enterobacteriaceae. J Antimicrob Chemother architecture comprising a putative novel blaVIM-2-harbouring trans-
73:1121–1137. https://doi.org/10.1093/jac/dkx488. poson in Pseudomonas aeruginosa. J Antimicrob Chemother 72:
295. Pitout J. 2010. Infections with extended-spectrum beta-lactamase- 2225–2229. https://doi.org/10.1093/jac/dkx143.
producing Enterobacteriaceae changing epidemiology and drug treat- 312. Shearer JE, Wireman J, Hostetler J, Forberger H, Borman J, Gill J,
ment choices. Drugs 70:313–333. https://doi.org/10.2165/11533040 Sanchez S, Mankin A, Lamarre J, Lindsay JA, Bayles K, Nicholson A,
-000000000-00000. O’Brien F, Jensen SO, Firth N, Skurray RA, Summers AO. 2011. Major
296. Petty NK, Ben Zakour NL, Stanton-Cook M, Skippington E, Totsika M, families of multiresistant plasmids from geographically and epidemio-
Forde BM, Phan MD, Moriel DG, Peters KM, Davies M, Rogers BA, logically diverse staphylococci. G3 (Bethesda) 1:581–591. https://doi
Dougan G, Rodriguez-Bano J, Pascual A, Pitout JDD, Upton M, Paterson .org/10.1534/g3.111.000760.
DL, Walsh TR, Schembri MA, Beatson SA. 2014. Global dissemination of 313. Malachowa N, DeLeo FR. 2010. Mobile genetic elements of Staphylo-
coccus aureus. Cell Mol Life Sci 67:3057–3071. https://doi.org/10.1007/ 2 carry multiple antibiotic resistance genes in Pseudomonas aeruginosa
s00018-010-0389-4. ST235, ST253, ST111 and ST175 and are globally dispersed. J Antimi-
314. Mojumdar M, Khan SA. 1988. Characterization of the tetracycline resis- crob Chemother 72:620 – 622. https://doi.org/10.1093/jac/dkw471.
tance gene of plasmid pT181 of Staphylococcus aureus. J Bacteriol 334. Cochetti I, Tili E, Mingoia M, Varaldo PE, Montanari MP. 2008. erm(B)-
170:5522–5528. https://doi.org/10.1128/jb.170.12.5522-5528.1988. carrying elements in tetracycline-resistant pneumococci and corre-
315. Horinouchi S, Weisblum B. 1982. Nucleotide sequence and functional spondence between Tn1545 and Tn6003. Antimicrob Agents Che-
map of pC194, a plasmid that specifies inducible chloramphenicol mother 52:1285–1290. https://doi.org/10.1128/AAC.01457-07.
resistance. J Bacteriol 150:815– 825. 335. Launay A, Ballard SA, Johnson PD, Grayson ML, Lambert T. 2006.
316. Horinouchi S, Weisblum B. 1982. Nucleotide sequence and functional Transfer of vancomycin resistance transposon Tn1549 from Clostridium
map of pE194, a plasmid that specifies inducible resistance to macro- symbiosum to Enterococcus spp. in the gut of gnotobiotic mice. Anti-
lide, lincosamide, and streptogramin type B antibodies. J Bacteriol microb Agents Chemother 50:1054 –1062. https://doi.org/10.1128/AAC
150:804 – 814. .50.3.1054-1062.2006.
317. Jensen SO, Apisiridej S, Kwong SM, Yang YH, Skurray RA, Firth N. 2010. 336. Katayama Y, Takeuchi F, Ito T, Ma XX, Ui-Mizutani Y, Kobayashi I,
Analysis of the prototypical Staphylococcus aureus multiresistance plas- Hiramatsu K. 2003. Identification in methicillin-susceptible Staphylococ-
mid pSK1. Plasmid 64:135–142. https://doi.org/10.1016/j.plasmid.2010 cus hominis of an active primordial mobile genetic element for the
.06.001. staphylococcal cassette chromosome mec of methicillin-resistant
318. Morton TM, Eaton DM, Johnston JL, Archer GL. 1993. DNA sequence Staphylococcus aureus. J Bacteriol 185:2711–2722. https://doi.org/10
and units of transcription of the conjugative transfer gene complex (trs) .1128/jb.185.9.2711-2722.2003.
of Staphylococcus aureus plasmid pGO1. J Bacteriol 175:4436 – 4447. 337. Enright MC, Robinson DA, Randle G, Feil EJ, Grundmann H, Spratt BG.
https://doi.org/10.1128/jb.175.14.4436-4447.1993. 2002. The evolutionary history of methicillin-resistant Staphylococcus
319. Liu MA, Kwong SM, Jensen SO, Brzoska AJ, Firth N. 2013. Biology of the aureus (MRSA). Proc Natl Acad Sci U S A 99:7687–7692. https://doi.org/
staphylococcal conjugative multiresistance plasmid pSK41. Plasmid 70: 10.1073/pnas.122108599.
42–51. https://doi.org/10.1016/j.plasmid.2013.02.001. 338. Katayama Y, Zhang HZ, Hong D, Chambers HF. 2003. Jumping the
320. Archer GL, Johnston JL. 1983. Self-transmissible plasmids in staphylo- barrier to beta-lactam resistance in Staphylococcus aureus. J Bacteriol
cocci that encode resistance to aminoglycosides. Antimicrob Agents 185:5465–5472. https://doi.org/10.1128/jb.185.18.5465-5472.2003.
Chemother 24:70 –77. https://doi.org/10.1128/aac.24.1.70. 339. von Wintersdorff CJH, Penders J, van Niekerk JM, Mills ND, Majum-
321. Goering RV, Ruff EA. 1983. Comparative analysis of conjugative plas- der S, van Alphen LB, Savelkoul PHM, Wolffs P. 2016. Dissemination
mids mediating gentamicin resistance in Staphylococcus aureus. Anti- of antimicrobial resistance in microbial ecosystems through hori-
microb Agents Chemother 24:450 – 452. https://doi.org/10.1128/aac.24 zontal gene transfer. Front Microbiol 7:173. https://doi.org/10.3389/
.3.450. fmicb.2016.00173.
322. Littlejohn TG, DiBerardino D, Messerotti LJ, Spiers SJ, Skurray RA. 1991. 340. Conlan S, Thomas PJ, Deming C, Park M, Lau AF, Dekker JP, Snitkin ES,
Structure and evolution of a family of genes encoding antiseptic and Clark TA, Luong K, Song Y, Tsai YC, Boitano M, Dayal J, Brooks SY,
disinfectant resistance in Staphylococcus aureus. Gene 101:59 – 66. Schmidt B, Young AC, Thomas JW, Bouffard GG, Blakesley RW, NISC
https://doi.org/10.1016/0378-1119(91)90224-y. Comparative Sequencing Program, Mullikin JC, Korlach J, Henderson
323. Morton TM, Johnston JL, Patterson J, Archer GL. 1995. Characterization DK, Frank KM, Palmore TN, Segre JA. 2014. Single-molecule sequencing
of a conjugative staphylococcal mupirocin resistance plasmid. Antimi- to track plasmid diversity of hospital-associated carbapenemase-
crob Agents Chemother 39:1272–1280. https://doi.org/10.1128/aac.39 producing Enterobacteriaceae. Sci Transl Med 6:254ra126. https://doi
.6.1272. .org/10.1126/scitranslmed.3009845.
324. Diep BA, Gill SR, Chang RF, Phan TH, Chen JH, Davidson MG, Lin F, Lin 341. Weingarten RA, Johnson RC, Conlan S, Ramsburg AM, Dekker JP, Lau
J, Carleton HA, Mongodin EF, Sensabaugh GF, Perdreau-Remington F. AF, Khil P, Odom RT, Deming C, Park M, Thomas PJ, NISC Compar-
2006. Complete genome sequence of USA300, an epidemic clone of ative Sequencing Program, Henderson DK, Palmore TN, Segre JA,
community-acquired meticillin-resistant Staphylococcus aureus. Lancet Frank KM. 2018. Genomic analysis of hospital plumbing reveals diverse
367:731–739. https://doi.org/10.1016/S0140-6736(06)68231-7. reservoir of bacterial plasmids conferring carbapenem resistance. mBio
325. Evans J, Dyke KG. 1988. Characterization of the conjugation system 9:e02011-17. https://doi.org/10.1128/mBio.02011-17.
associated with the Staphylococcus aureus plasmid pJE1. J Gen Micro- 342. Grundmann H, Aires-De-Sousa M, Boyce J, Tiemersma E. 2006. Emer-
biol 134:1– 8. https://doi.org/10.1099/00221287-134-1-1. gence and resurgence of meticillin-resistant Staphylococcus aureus
326. Bender J, Strommenger B, Steglich M, Zimmermann O, Fenner I, as a public-health threat. Lancet 368:874 – 885. https://doi.org/10
Lensing C, Dagwadordsch U, Kekule AS, Werner G, Layer F. 2015. .1016/S0140-6736(06)68853-3.
Linezolid resistance in clinical isolates of Staphylococcus epidermidis 343. Gardete S, Tomasz A. 2014. Mechanisms of vancomycin resistance in
from German hospitals and characterization of two cfr-carrying Staphylococcus aureus. J Clin Invest 124:2836 –2840. https://doi.org/10
plasmids. J Antimicrob Chemother 70:1630 –1638. https://doi.org/10 .1172/JCI68834.
.1093/jac/dkv025. 344. Meyer R. 2009. Replication and conjugative mobilization of broad
327. Juhas M, van der Meer JR, Gaillard M, Harding RM, Hood DW, Crook DW. host-range IncQ plasmids. Plasmid 62:57–70. https://doi.org/10.1016/j
2009. Genomic islands: tools of bacterial horizontal gene transfer and .plasmid.2009.05.001.
evolution. FEMS Microbiol Rev 33:376 –393. https://doi.org/10.1111/j 345. Maslanova I, Stribna S, Doskar J, Pantucek R. 2016. Efficient plasmid
.1574-6976.2008.00136.x. transduction to Staphylococcus aureus strains insensitive to the lytic
328. Bellanger X, Payot S, Leblond-Bourget N, Guédon G. 2014. Conjugative action of transducing phage. FEMS Microbiol Lett 363:fnw211. https://
and mobilizable genomic islands in bacteria: evolution and diversity. doi.org/10.1093/femsle/fnw211.
FEMS Microbiol Rev 38:720 –760. https://doi.org/10.1111/1574-6976 346. Chlebowicz MA, Mašlaňová I, Kuntová L, Grundmann H, Pantůček R,
.12058. Doškař J, van Dijl JM, Buist G. 2014. The staphylococcal cassette chro-
329. Mantri Y, Williams KP. 2004. Islander: a database of integrative islands mosome mec type V from Staphylococcus aureus ST398 is packaged
in prokaryotic genomes, the associated integrases and their DNA site into bacteriophage capsids. Int J Med Microbiol 304:764 –774. https://
specificities. Nucleic Acids Res 32:D55–D58. https://doi.org/10.1093/ doi.org/10.1016/j.ijmm.2014.05.010.
nar/gkh059. 347. Blahova J, Kralikova K, Krcmery V, Jezek P. 2000. Low-frequency trans-
330. Dobrindt U, Hochhut B, Hentschel U, Hacker J. 2004. Genomic islands in duction of imipenem resistance and high-frequency transduction of
pathogenic and environmental microorganisms. Nat Rev Microbiol ceftazidime and aztreonam resistance by the bacteriophage AP-151
2:414 – 424. https://doi.org/10.1038/nrmicro884. isolated from a Pseudomonas aeruginosa strain. J Chemother 12:
331. Burrus V, Pavlovic G, Decaris B, Guedon G. 2002. Conjugative 482– 486. https://doi.org/10.1179/joc.2000.12.6.482.
transposons: the tip of the iceberg. Mol Microbiol 46:601– 610. https:// 348. Krahn T, Wibberg D, Maus I, Winkler A, Bontron S, Sczyrba A, Nordmann
doi.org/10.1046/j.1365-2958.2002.03191.x. P, Puhler A, Poirel L, Schluter A. 2016. Intraspecies transfer of the
332. Delavat F, Miyazaki R, Carraro N, Pradervand N, van der Meer JR. 2017. chromosomal Acinetobacter baumannii bla(NDM-1) carbapenemase
The hidden life of integrative and conjugative elements. FEMS Micro- gene. Antimicrob Agents Chemother 60:3032–3040. https://doi.org/10
biol Rev 41:512–537. https://doi.org/10.1093/femsre/fux008. .1128/AAC.00124-16.
333. Roy Chowdhury P, Scott MJ, Djordjevic SP. 2017. Genomic islands 1 and 349. Johnston C, Martin B, Fichant G, Polard P, Claverys JP. 2014. Bacterial
transformation: distribution, shared mechanisms and divergent control. 2017. Frequency of antiseptic resistance genes and reduced suscepti-
Nat Rev Microbiol 12:181–196. https://doi.org/10.1038/nrmicro3199. bility to biocides in carbapenem-resistant Acinetobacter baumannii. J
350. Cameron ADS, Redfield RJ. 2006. Non-canonical CRP sites control com- Med Microbiol 66:13–17. https://doi.org/10.1099/jmm.0.000403.
petence regulons in Escherichia coli and many other gamma- 370. Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL. 2007. Drugs for bad
proteobacteria. Nucleic Acids Res 34:6001– 6014. https://doi.org/10 bugs: confronting the challenges of antibacterial discovery. Nat Rev
.1093/nar/gkl734. Drug Discov 6:29 – 40. https://doi.org/10.1038/nrd2201.
351. Palchevskiy V, Finkel SE. 2006. Escherichia coli competence gene 371. Fischbach MA, Walsh CT. 2009. Antibiotics for emerging pathogens.
homologs are essential for competitive fitness and the use of DNA Science 325:1089 –1093. https://doi.org/10.1126/science.1176667.
as a nutrient. J Bacteriol 188:3902–3910. https://doi.org/10.1128/JB 372. Renwick M, Mossialos E. 2018. What are the economic barriers of
.01974-05. antibiotic R&D and how can we overcome them? Expert Opin Drug
352. Domingues S, Nielsen KM, da Silva GJ. 2012. Various pathways leading Discov 13:889 – 892. https://doi.org/10.1080/17460441.2018.1515908.
to the acquisition of antibiotic resistance by natural transformation. 373. Sertkaya A, Eyraud J, Birkenbach A, Franz C, Ackerley N, Overton V,
Mob Genet Elements 2:257–260. https://doi.org/10.4161/mge.23089. Outterson K. 2014. Analytical framework for examining the value of
353. Baltrus DA. 2013. Exploring the costs of horizontal gene transfer. Trends antibacterial products. https://aspe.hhs.gov/system/files/pdf/76891/rpt
Ecol Evol 28:489 – 495. https://doi.org/10.1016/j.tree.2013.04.002. _antibacterials.pdf. Accessed 20 January 2020.
354. Vogwill T, MacLean RC. 2015. The genetic basis of the fitness costs of 374. Theuretzbacher U, Outterson K, Engel A, Karlen A. 19 November 2019.
antimicrobial resistance: a meta-analysis approach. Evol Appl The global preclinical antibacterial pipeline. Nat Rev Microbiol https://
8:284 –295. https://doi.org/10.1111/eva.12202. doi.org/10.1038/s41579-019-0288-0.
355. Wiggins AG, LaVoie SP, Wireman J, Summers AO. 2018. Thinking out- 375. Sharma P, Towse A. 2011. New drugs to tackle antimicrobial resistance:
side the (pill) box: does toxic metal exposure thwart antibiotic stew- analysis of EU policy options. https://www.ohe.org/publications/new
ardship best practices? Plasmid 99:68 –71. https://doi.org/10.1016/j -drugs-tackle-antimicrobial-resistance-analysis-eu-policy-options. Ac-
.plasmid.2018.08.003. cessed 8 January 2020.
356. Lin F, Xu Y, Chang Y, Liu C, Jia X, Ling B. 2017. Molecular charac- 376. Access to Medicine Foundation. 2018. Antimicrobial resistance bench-
terization of reduced susceptibility to biocides in clinical isolates of mark 2018. https://accesstomedicinefoundation.org/media/uploads/
Acinetobacter baumannii. Front Microbiol 8:1836. https://doi.org/10 downloads/5bc5edd8367eb_Antimicrobial-Resistance
.3389/fmicb.2017.01836. -Benchmark-2018.pdf. Accessed 8 January 2020.
357. Pal C, Bengtsson-Palme J, Kristiansson E, Larsson D. 2015. Co- 377. Innovative Medicines Initiative (IMI). 2017. New drugs for bad bugs. The
occurrence of resistance genes to antibiotics, biocides and metals Innovative Medicines Initiative response to antimicrobial resistance.
reveals novel insights into their co-selection potential. BMC Genomics https://www.imi.europa.eu/sites/default/files/uploads/documents/
16:964. https://doi.org/10.1186/s12864-015-2153-5. projects/IMI_AMR_2017_LR.pdf. Accessed 7 January 2020.
358. Canton R, Ruiz-Garbajosa P. 2011. Co-resistance: an opportunity for the 378. Innovative Medicines Initiative (IMI). 2019. VALUE-Dx. The value of
bacteria and resistance genes. Curr Opin Pharmacol 11:477– 485. diagnostics to combat antimicrobial resistance by optimising anti-
https://doi.org/10.1016/j.coph.2011.07.007. biotic use. https://www.imi.europa.eu/projects-results/project-factsheets/
359. McDonnell G, Russell AD. 2001. Antiseptics and disinfectants: activity, value-dx. Accessed 7 January 2020.
action, and resistance. Clin Microbiol Rev 14:227–228. https://doi.org/ 379. COMBACTE. 2019. COMBACTE-CARE: combatting bacterial resistance in
10.1128/CMR.12.1.147. Europe— carbapenem-resistance. https://www.combacte.com/about/
360. Schweizer HP. 1998. Intrinsic resistance to inhibitors of fatty acid about-combacte-care-detail/. Accessed 7 January 2020.
biosynthesis in Pseudomonas aeruginosa is due to efflux: application of 380. DRIVE AB. 2014. Driving reinvestment in R&D for antibiotics and their
a novel technique for generation of unmarked chromosomal mutations responsible use. http://drive-ab.eu/. Accessed 8 January 2020.
for the study of efflux systems. Antimicrob Agents Chemother 42: 381. Ardal C, Baraldi E, Theuretzbacher U, Outterson K, Plahte J, Ciabuschi F,
394 –398. https://doi.org/10.1128/AAC.42.2.394. Rottingen JA. 2018. Insights into early stage of antibiotic development
361. Nikaido H. 1998. Antibiotic resistance caused by Gram-negative multi- in small- and medium-sized enterprises: a survey of targets, costs, and
drug efflux pumps. Clin Infect Dis 27:S32–S41. https://doi.org/10.1086/ durations. J Pharm Policy Pract 11:8. https://doi.org/10.1186/s40545
514920. -018-0135-0.
362. Paulsen IT, Littlejohn TG, Radstrom P, Sundstrom L, Skold O, Swedberg 382. Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP,
G, Skurray RA. 1993. The 3’ conserved segment of integrons contains a Mueller A, Schaberle TF, Hughes DE, Epstein S, Jones M, Lazarides L,
gene associated with multidrug resistance to antiseptics and disinfec- Steadman VA, Cohen DR, Felix CR, Fetterman KA, Millett WP, Nitti AG,
tants. Antimicrob Agents Chemother 37:761–768. https://doi.org/10 Zullo AM, Chen C, Lewis K. 2015. A new antibiotic kills pathogens
.1128/aac.37.4.761. without detectable resistance. Nature 517:455– 459. https://doi.org/10
363. Kazama H, Hamashima H, Sasatsu M, Arai T. 1999. Characterization of .1038/nature14098.
the antiseptic-resistance gene qacE delta 1 isolated from clinical and 383. Summers WC. 2001. Bacteriophage therapy. Annu Rev Microbiol 55:
environmental isolates of Vibrio parahaemolyticus and Vibrio cholerae 437– 451. https://doi.org/10.1146/annurev.micro.55.1.437.
non-O1. FEMS Microbiol Lett 174:379 –384. https://doi.org/10.1111/j 384. Liu CI, Liu GY, Song Y, Yin F, Hensler ME, Jeng WY, Nizet V, Wang AH,
.1574-6968.1999.tb13593.x. Oldfield E. 2008. A cholesterol biosynthesis inhibitor blocks Staphylo-
364. Gillings MR, Duan XJ, Hardwick SA, Holley MP, Stokes HW. 2009. Gene coccus aureus virulence. Science 319:1391–1394. https://doi.org/10
cassettes encoding resistance to quaternary ammonium compounds: a .1126/science.1153018.
role in the origin of clinical class 1 integrons? ISME J 3:209 –215. 385. Vaara M, Vaara T. 1983. Sensitization of Gram-negative bacteria to
https://doi.org/10.1038/ismej.2008.98. antibiotics and complement by a nontoxic oligopeptide. Nature 303:
365. Gaze WH, Abdouslam N, Hawkey PM, Wellington E. 2005. Incidence of 526 –528. https://doi.org/10.1038/303526a0.
class 1 integrons in a quaternary ammonium compound-polluted en- 386. Zinkernagel AS, Peyssonnaux C, Johnson RS, Nizet V. 2008. Pharmaco-
vironment. Antimicrob Agents Chemother 49:1802–1807. https://doi logic augmentation of hypoxia-inducible factor-1alpha with mimosine
.org/10.1128/AAC.49.5.1802-1807.2005. boosts the bactericidal capacity of phagocytes. J Infect Dis 197:
366. Sidhu MS, Heir E, Sorum H, Holck A. 2001. Genetic linkage between 214 –217. https://doi.org/10.1086/524843.
resistance to quaternary ammonium compounds and beta-lactam an- 387. Gill EE, Franco OL, Hancock RE. 2015. Antibiotic adjuvants: diverse
tibiotics in food-related Staphylococcus spp. Microb Drug Resist strategies for controlling drug-resistant pathogens. Chem Biol Drug
7:363–371. https://doi.org/10.1089/10766290152773374. Des 85:56 –78. https://doi.org/10.1111/cbdd.12478.
367. Baker-Austin C, Wright MS, Stepanauskas R, McArthur JV. 2006. Co- 388. Theuretzbacher U, Gottwalt S, Beyer P, Butler M, Czaplewski L, Lien-
selection of antibiotic and metal resistance. Trends Microbiol 14: hardt C, Moja L, Paul M, Paulin S, Rex JH, Silver LL, Spigelman M,
176 –182. https://doi.org/10.1016/j.tim.2006.02.006. Thwaites GE, Paccaud JP, Harbarth S. 2019. Analysis of the clinical
368. Kazama H, Hamashima H, Sasatsu M, Arai T. 1998. Distribution of the antibacterial and antituberculosis pipeline. Lancet Infect Dis 19:
antiseptic-resistance gene qacE delta 1 in Gram-positive bacteria. FEMS e40 – e50. https://doi.org/10.1016/S1473-3099(18)30513-9.
Microbiol Lett 165:295–299. https://doi.org/10.1111/j.1574-6968.1998 389. Bohlmann L, De Oliveira DMP, El-Deeb IM, Brazel EB, Harbison-Price N,
.tb13160.x. Ong CY, Rivera-Hernandez T, Ferguson SA, Cork AJ, Phan MD, Soder-
369. Liu WJ, Fu L, Huang M, Zhang JP, Wu Y, Zhou YS, Zeng J, Wang GX. holm AT, Davies MR, Nimmo GR, Dougan G, Schembri MA, Cook GM,
McEwan AG, von Itzstein M, McDevitt CA, Walker MJ. 2018. Chemical novel fluoroquinolone lascufloxacin (KRP-AM1977). Antimicrob Agents
synergy between ionophore PBT2 and zinc reverses antibiotic resis- Chemother 61:e00120-17. https://doi.org/10.1128/AAC.00120-17.
tance. mBio 9:e02391-18. https://doi.org/10.1128/mBio.02391-18. 408. Furuie H, Tanioka S, Shimizu K, Manita S, Nishimura M, Yoshida H. 2018.
390. The PEW Charitable Trusts. 2019. Antibiotics currently in global clinical Intrapulmonary pharmacokinetics of lascufloxacin in healthy adult vol-
development. https://www.pewtrusts.org/-/media/assets/2019/08/arp unteers. Antimicrob Agents Chemother 62:e02169-17. https://doi.org/
_antibiotics_currently_in_global_clinical_development_data_table_v2 10.1128/AAC.02169-17.
.pdf. Accessed 18 November 2019. 409. Kato T, Odajima M, Kohno S. 2017. Phase II clinical study of oral
391. Shaeer KM, Zmarlicka MT, Chahine EB, Piccicacco N, Cho JC. 2019. lascufloxacin in community-acquired pneumonia, abstr P-1362. Abstr
Plazomicin: a next-generation aminoglycoside. Pharmacotherapy 39: 27th Congr ECCMID.
77–93. https://doi.org/10.1002/phar.2203. 410. Kyorin Pharmaceutical Co. Ltd. 2019. NHI drug price listing and release of
392. U.S. Food and Drug Administration. 2018. Centre for drug evaluation and oral quinolone antibacterial agent “Lasvic tablets 75mg.” https://www
research. Application number: 210303Orig1s000. Summary review. https:// .kyorin-pharm.co.jp/en/news/a329f0ae64024c1173f40660eede0efb37f1cbb0
www.accessdata.fda.gov/drugsatfda_docs/nda/2018/210303Orig .pdf. Accessed 12 March 2020.
1s000SumR.pdf. Accessed 14 January 2020. 411. Bonomo RA. 2019. Cefiderocol: a novel siderophore cephalosporin
393. Wagenlehner FME, Cloutier DJ, Komirenko AS, Cebrik DS, Krause KM, defeating carbapenem-resistant pathogens. Clin Infect Dis 69:
Keepers TR, Connolly LE, Miller LG, Friedland I, Dwyer JP, EPIC Study S519 –S520. https://doi.org/10.1093/cid/ciz823.
Group. 2019. Once-daily plazomicin for complicated urinary tract 412. Zhanel GG, Golden AR, Zelenitsky S, Wiebe K, Lawrence CK, Adam HJ,
infections. N Engl J Med 380:729 –740. https://doi.org/10.1056/ Idowu T, Domalaon R, Schweizer F, Zhanel MA, Lagace-Wiens PRS, Walkty
NEJMoa1801467. AJ, Noreddin A, Lynch JP, Karlowsky JA. 2019. Cefiderocol: a siderophore
394. Snydman DR, McDermott LA, Jacobus NV, Kerstein K, Grossman TH, cephalosporin with activity against carbapenem-resistant and multidrug-
Sutcliffe JA. 2018. Evaluation of the in vitro activity of eravacycline resistant Gram-negative bacilli. Drugs 79:271–289. https://doi.org/10.1007/
against a broad spectrum of recent clinical anaerobic isolates. Antimi- s40265-019-1055-2.
crob Agents Chemother 62:e02206-17. https://doi.org/10.1128/AAC 413. Martin-Loeches I, Dale GE, Torres A. 2018. Murepavadin: a new antibi-
.02206-17. otic class in the pipeline. Expert Rev Anti Infect Ther 16:259 –268.
395. Solomkin JS, Gardovskis J, Lawrence K, Montravers P, Sway A, Evans D, https://doi.org/10.1080/14787210.2018.1441024.
Tsai L. 2019. IGNITE4: results of a phase 3, randomized, multicenter, 414. Bernardini F, Dale GE, Wach A, Obrecht D. 2019. WS01-4 pharmacoki-
prospective trial of eravacycline vs. meropenem in the treatment of netics and pharmacodynamics of murepavadin (POL7080) in neutro-
complicated intra-abdominal infections. Clin Infect Dis 69:921–929. penic lung infection models when evaluated by aerosol administration.
https://doi.org/10.1093/cid/ciy1029. J Cyst Fibros 18(Suppl 1):S2. https://doi.org/10.1016/S1569
396. Dougherty JA, Sucher AJ, Chahine EB, Shihadeh KC. 2019. -1993(19)30120-1.
Omadacycline: a new tetracycline antibiotic. Ann Pharmacother 53: 415. Perazella MA. 2018. Pharmacology behind common drug nephrotoxic-
486 –500. https://doi.org/10.1177/1060028018818094. ities. Clin J Am Soc Nephrol 13:1897–1908. https://doi.org/10.2215/CJN
397. Stets R, Popescu M, Gonong JR, Mitha I, Nseir W, Madej A, Kirsch C, Das .00150118.
AF, Garrity-Ryan L, Steenbergen JN, Manley A, Eckburg PB, Tzanis E, 416. Spero Therapeutics Inc. 2018. Spero Therapeutics announces positive
McGovern PC, Loh E. 2019. Omadacycline for community-acquired top-line data for two product candidates from its potentiator platform.
bacterial pneumonia. N Engl J Med 380:517–527. https://doi.org/10 https://investors.sperotherapeutics.com/node/7091/pdf. Accessed 21 April
.1056/NEJMoa1800201. 2020.
398. European Medicines Agency. 2019. Withdrawal of application for the 417. Corbett D, Wise A, Langley T, Skinner K, Trimby E, Birchall S, Dorali A,
marketing authorisation of Nuzyra (omadacycline). https://www.ema Sandiford S, Williams J, Warn P, Vaara M, Lister T. 2017. Potentiation of
.europa.eu/en/documents/medicine-qa/questions-answers-withdrawal antibiotic activity by a novel cationic peptide: potency and spectrum of
-application-marketing-authorisation-nuzyra-omadacycline_en.pdf. Ac- activity of SPR741. Antimicrob Agents Chemother 61:e00200-17. https://
cessed 11 March 2020. doi.org/10.1128/AAC.00200-17.
399. DRIVE AB. 2018. DRIVE AB report: revitalizing the antibiotic pipeline: 418. Vaara M, Siikanen O, Apajalahti J, Fox J, Frimodt-Moller N, He H,
stimulatinh innovation while driving sustainable used and global Poudyal A, Li JA, Nation RL, Vaara T. 2010. A novel polymyxin derivative
access. http://drive-ab.eu/wp-content/uploads/2018/01/DRIVE-AB-Final that lacks the fatty acid tail and carries only three positive charges has
-Report-Jan2018.pdf. Accessed 12 March 2020. strong synergism with agents excluded by the intact outer membrane.
400. Merck & Co., Inc. 2019. Recarbrio prescribing information. http://bit.ly/ Antimicrob Agents Chemother 54:3341–3346. https://doi.org/10.1128/
2U4x4bP. Accessed 23 November 2019. AAC.01439-09.
401. Papp-Wallace KM, Barnes MD, Alsop J, Taracila MA, Bethel CR, Becka SA, 419. Harris PNA, Peleg AY, Iredell J, Ingram PR, Miyakis S, Stewardson AJ,
van Duin D, Kreiswirth BN, Kaye KS, Bonomo RA. 2018. Relebactam is a Rogers BA, McBryde ES, Roberts JA, Lipman J, Athan E, Paul SK, Baker
potent inhibitor of the KPC-2 beta-lactamase and restores imipenem P, Harris-Brown T, Paterson DL. 2015. Meropenem versus piperacillin-
susceptibility in KPC-producing Enterobacteriaceae. Antimicrob Agents tazobactam for definitive treatment of bloodstream infections due to
Chemother 62:e00174-18. https://doi.org/10.1128/AAC.00174-18. ceftriaxone non-susceptible Escherichia coli and Klebsiella spp (the
402. U.S. Food and Drug Administration. 2019. FDA approves new antibiotic MERINO trial): study protocol for a randomised controlled trial. Trials
to treat community-acquired bacterial pneumonia. https://www.fda 16:24. https://doi.org/10.1186/s13063-014-0541-9.
.gov/news-events/press-announcements/fda-approves-new-antibiotic 420. Harris PNA, Tambyah PA, Lye DC, Mo Y, Lee TH, Yilmaz M, Alenazi TH,
-treat-community-acquired-bacterial-pneumonia. Accessed 3 Novem- Arabi Y, Falcone M, Bassetti M, Righi E, Rogers BA, Kanj S, Bhally H,
ber 2019. Iredell J, Mendelson M, Boyles TH, Looke D, Miyakis S, Walls G, Al
403. Alexander E, Goldberg L, Das AF, Moran GJ, Sandrock C, Gasink LB, Khamis M, Zikri A, Crowe A, Ingram P, Daneman N, Griffin P, Athan E,
Spera P, Sweeney C, Paukner S, Wicha WW, Gelone SP, Schranz J. 2019. Lorenc P, Baker P, Roberts L, Beatson SA, Peleg AY, Harris-Brown T,
Oral lefamulin vs moxifloxacin for early clinical response among adults Paterson DL, MERINO Trial Investigators and the Australasian Society
with community-acquired bacterial pneumonia: the LEAP 2 random- for Infectious Disease Clinical Research Network (ASID-CRN). 2018.
ized clinical trial. JAMA 322:1661–1671. https://doi.org/10.1001/jama Effect of piperacillin-tazobactam vs meropenem on 30-day mortality
.2019.15468. for patients with E. coli or Klebsiella pneumoniae bloodstream infection
404. Nabriva Therapeutics AG. 2020. Lefamulin. https://www.nabriva.com/ and ceftriaxone resistance: a randomized clinical trial. JAMA 320:
pipeline-research. Accessed 11 March 2020. 984 –994. https://doi.org/10.1001/jama.2018.12163.
405. European Medicines Agency. 2020. EMEA-002075-PIP01-16-M01. https:// 421. Kehlet H. 2008. Fast-track colorectal surgery. Lancet 371:791–793.
www.ema.europa.eu/en/medicines/human/paediatric-investigation https://doi.org/10.1016/S0140-6736(08)60357-8.
-plans/emea-002075-pip01-16-m01. Accessed 11 March 2020. 422. Khoo CK, Vickery CJ, Forsyth N, Vinall NS, Eyre-Brook IA. 2007. A
406. Kyorin Pharmaceutical Co. Ltd. 2019. Kyorin pharmaceutical receives prospective randomized controlled trial of multimodal perioperative
marketing approval for oral quinolone antibacterial agent “Lasvic management protocol in patients undergoing elective colorectal re-
tablets 75mg.” https://www.kyorin-pharm.co.jp/en/news/691640 section for cancer. Ann Surg 245:867– 872.
e4c4142c191c8d4061a71c02e299076a4a.pdf. Accessed 11 March 2020. 423. Iversen K, Ihlemann N, Gill SU, Madsen T, Elming H, Jensen KT, Bruun
407. Kishii R, Yamaguchi Y, Takei M. 2017. In vitro activities and spectrum of the NE, Høfsten DE, Fursted K, Christensen JJ, Schultz M, Klein CF, Fosbøll
EL, Rosenvinge F, Schønheyder HC, Køber L, Torp-Pedersen C, Helweg- resistant bacteria. Nature 546:587–588. https://doi.org/10.1038/
Larsen J, Tønder N, Moser C, Bundgaard H. 2019. Partial oral versus nature.2017.22173.
intravenous antibiotic treatment of endocarditis. N Engl J Med 380: 443. UC San Diego Health. 2019. With OK from FDA, UC San Diego research-
415– 424. https://doi.org/10.1056/NEJMoa1808312. ers prepare to launch novel phage study. https://health.ucsd.edu/
424. Jewetz E, Gunnison JB, Bruff JB, Coleman VR. 1952. Studies on antibiotic news/releases/Pages/2019-01-08-FDA-okays-uc-san-diego-to-launch
synergism and antagonism. Synergism among seven antibiotics -novel-phage-study.aspx. Accessed 13 August 2019.
against various bacteria in vitro. J Bacteriol 64:29 –39. 444. Lehman SM, Mearns G, Rankin D, Cole RA, Smrekar F, Branston SD,
425. Bushby SR, Hitchings GH. 1968. Trimethoprim, a sulphonamide poten- Morales S. 2019. Design and preclinical development of a phage prod-
tiator. Br J Pharmacol Chemother 33:72–90. https://doi.org/10.1111/j uct for the treatment of antibiotic-resistant Staphylococcus aureus in-
.1476-5381.1968.tb00475.x. fections. Viruses 11:88. https://doi.org/10.3390/v11010088.
426. Tyers M, Wright GD. 2019. Drug combinations: a strategy to extend the 445. Pirnay JP, Verbeken G, Ceyssens PJ, Huys I, De Vos D, Ameloot C,
life of antibiotics in the 21st century. Nat Rev Microbiol 17:141–155. Fauconnier A. 2018. The magistral phage. Viruses 10:64. https://doi.org/
https://doi.org/10.1038/s41579-018-0141-x. 10.3390/v10020064.
427. Neu HC, Fu KP. 1978. Clavulanic acid, a novel inhibitor of beta- 446. Chong CR, Sullivan DJ. 2007. New uses for old drugs. Nature 448:
lactamases. Antimicrob Agents Chemother 14:650 – 655. https://doi 645– 646. https://doi.org/10.1038/448645a.
.org/10.1128/aac.14.5.650. 447. Christiansen SH, Murphy RA, Juul-Madsen K, Fredborg M, Hvam ML,
428. Wong DR, van Duin D. 2017. Novel beta-lactamase inhibitors: unlocking Axelgaard E, Skovdal SM, Meyer RL, Sorensen U, Moller A, Nyengaard
their potential in therapy. Drugs 77:615– 628. https://doi.org/10.1007/ JR, Norskov-Lauritsen N, Wang M, Gadjeva M, Howard KA, Davies JC,
s40265-017-0725-1. Petersen E, Vorup-Jensen T. 2017. The immunomodulatory drug glati-
429. Wright GD. 2016. Antibiotic adjuvants: rescuing antibiotics from resis- ramer acetate is also an effective antimicrobial agent that kills Gram-
tance. Trends Microbiol 24:862– 871. https://doi.org/10.1016/j.tim.2016 negative bacteria. Sci Rep 7:15653. https://doi.org/10.1038/s41598-017
.06.009. -15969-3.
430. King AM, Reid-Yu SA, Wang WL, King DT, De Pascale G, Strynadka NC, 448. Kim W, Zhu WP, Hendricks GL, Van Tyne D, Steele AD, Keohane CE, Fricke
Walsh TR, Coombes BK, Wright GD. 2014. Aspergillomarasmine A over- N, Conery AL, Shen S, Pan W, Lee K, Rajamuthiah R, Fuchs BB, Vlahovska
comes metallo-beta-lactamase antibiotic resistance. Nature 510: PM, Wuest WM, Gilmore MS, Gao HJ, Ausubel FM, Mylonakis E. 2018. A
503–506. https://doi.org/10.1038/nature13445. new class of synthetic retinoid antibiotics effective against bacterial per-
431. Thompson RJ, Bobay BG, Stowe SD, Olson AL, Peng LL, Su ZM, Actis LA, sisters. Nature 556:103–107. https://doi.org/10.1038/nature26157.
Melander C, Cavanagh J. 2012. Identification of BfmR, a response 449. Younis W, Thangamani S, Seleem MN. 2015. Repurposing non-antimicrobial
regulator involved in biofilm development, as a target for a drugs and clinical molecules to treat bacterial infections. Curr Pharm Des
2-aminoimidazole-based antibiofilm agent. Biochemistry 51: 21:4106–4111. https://doi.org/10.2174/1381612821666150506154434.
9776 –9778. https://doi.org/10.1021/bi3015289. 450. Brown D. 2015. Antibiotic resistance breakers: can repurposed drugs fill
the antibiotic discovery void? Nat Rev Drug Discov 14:821– 832. https://
432. Cox G, Koteva K, Wright GD. 2014. An unusual class of anthracyclines
doi.org/10.1038/nrd4675.
potentiate Gram-positive antibiotics in intrinsically resistant Gram-
451. McConnell MJ. 2019. Where are we with monoclonal antibodies for
negative bacteria. J Antimicrob Chemother 69:1844 –1855. https://doi
multidrug-resistant infections? Drug Discov Today 24:1132–1138.
.org/10.1093/jac/dku057.
https://doi.org/10.1016/j.drudis.2019.03.002.
433. Zurawski DV, Reinhart AA, Alamneh YA, Pucci MJ, Si YZ, Abu-Taleb R,
452. Tsai CW, Morris S. 2015. Approval of raxibacumab for the treatment
Shearer JP, Demons ST, Tyner SD, Lister T. 2017. SPR741, an antibiotic
of inhalation anthrax under the US Food and Drug Administration
adjuvant, potentiates the in vitro and in vivo activity of rifampin against
“animal rule.” Front Microbiol 6:1320. https://doi.org/10.3389/fmicb
clinically relevant extensively drug-resistant Acinetobacter baumannii.
.2015.01320.
Antimicrob Agents Chemother 61:e01239-17. https://doi.org/10.1128/
453. Hou AW, Morrill AM. 2017. Obiltoxaximab: adding to the treatment
AAC.01239-17.
arsenal for Bacillus anthracis infection. Ann Pharmacother 51:908 –913.
434. Jen FE, Everest-Dass AV, El-Deeb IM, Singh S, Haselhorst T, Walker MJ,
https://doi.org/10.1177/1060028017713029.
von Itzstein M, Jennings MP. 2020. Neisseria gonorrhoeae becomes
454. Kufel WD, Devanathan AS, Marx AH, Weber DJ, Daniels LM. 2017.
susceptible to polymyxin B and colistin in the presence of PBT2. ACS Bezlotoxumab: a novel agent for the prevention of recurrent Clostrid-
Infect Dis 6:50 –55. https://doi.org/10.1021/acsinfecdis.9b00307. ium difficile infection. Pharmacotherapy 37:1298 –1308. https://doi.org/
435. Hancock REW, Nijnik A, Philpott DJ. 2012. Modulating immunity as a 10.1002/phar.1990.
therapy for bacterial infections. Nat Rev Microbiol 10:243–254. https:// 455. Adis Insight Drugs. 2019. Panobacumab—Aridis Pharmaceuticals.
doi.org/10.1038/nrmicro2745. https://adisinsight.springer.com/drugs/800024401. Accessed 10 January
436. Feigman MS, Kim S, Pidgeon SE, Yu Y, Ongwae GM, Patel DS, Regen S, 2020.
Im W, Pires MM. 2018. Synthetic immunotherapeutics against Gram- 456. Secher T, Fauconnier L, Szade A, Rutschi O, Fas SC, Ryffel B, Rudolf MP.
negative pathogens. Cell Chem Biol 25:1185–1194. https://doi.org/10 2011. Anti-Pseudomonas aeruginosa serotype O11 LPS immunoglobulin
.1016/j.chembiol.2018.05.019. M monoclonal antibody panobacumab (KBPA101) confers protection
437. Bhandari T, Nizet V. 2014. Hypoxia-inducible factor (HIF) as a pharma- in a murine model of acute lung infection. J Antimicrob Chemother
cological target for prevention and treatment of infectious diseases. 66:1100 –1109. https://doi.org/10.1093/jac/dkr038.
Infect Dis Ther 3:159 –174. https://doi.org/10.1007/s40121-014-0030-1. 457. Milla CE, Chmiel JF, Accurso FJ, VanDevanter DR, Konstan MW,
438. Zarember KA, Malech HL. 2005. HIF-1 alpha: a master regulator of Yarranton G, Geller DE, for the KB001 Study Group. 2014. Anti-PcrV
innate host defenses? J Clin Invest 115:1702–1704. https://doi.org/10 antibody in cystic fibrosis: a novel approach targeting Pseudomonas
.1172/JCI25740. aeruginosa airway infection. Pediatr Pulmonol 49:650 – 658. https://
439. Peyssonnaux C, Datta V, Cramer T, Doedens A, Theodorakis EA, Gallo doi.org/10.1002/ppul.22890.
RL, Hurtado-Ziola N, Nizet V, Johnson RS. 2005. HIF-1 alpha expression 458. Jain R, Beckett VV, Konstan MW, Accurso FJ, Burns JL, Mayer-
regulates the bactericidal capacity of phagocytes. J Clin Invest 115: Hamblett N, Milla C, VanDevanter DR, Chmiel JF, KB001-A Study
1806 –1815. https://doi.org/10.1172/JCI23865. Group. 2018. KB001-A, a novel anti-inflammatory, found to be safe
440. Berger EA, McClellan SA, Vistisen KS, Hazlett LD. 2013. HIF-1 alpha is and well-tolerated in cystic fibrosis patients infected with Pseu-
essential for effective PMN bacterial killing, antimicrobial peptide pro- domonas aeruginosa. J Cyst Fibros 17:484 – 491. https://doi.org/10
duction and apoptosis in Pseudomonas aeruginosa keratitis. PLoS Pat- .1016/j.jcf.2017.12.006.
hog 9:e1003457. https://doi.org/10.1371/journal.ppat.1003457. 459. DiGiandomenico A, Warrener P, Hamilton M, Guillard S, Ravn P, Minter
441. Okumura CYM, Hollands A, Tran DN, Olson J, Dahesh S, von Kockritz- R, Camara MM, Venkatraman V, MacGill RS, Lin J, Wang Q, Keller AE,
Blickwede M, Thienphrapa W, Corle C, Jeung SN, Kotsakis A, Shalwitz Bonnell JC, Tomich M, Jermutus L, McCarthy MP, Melnick DA, Suzich JA,
RA, Johnson RS, Nizet V. 2012. A new pharmacological agent (AKB- Stover CK. 2012. Identification of broadly protective human antibodies
4924) stabilizes hypoxia inducible factor-1 (HIF-1) and increases skin to Pseudomonas aeruginosa exopolysaccharide Psl by phenotypic
innate defenses against bacterial infection. J Mol Med (Berl) 90: screening. J Exp Med 209:1273–1287. https://doi.org/10.1084/jem
1079 –1089. https://doi.org/10.1007/s00109-012-0882-3. .20120033.
442. Reardon S. 2017. Modified viruses deliver death to antibiotic- 460. Ali SO, Yu XQ, Robbie GJ, Wu Y, Shoemaker K, Yu L, DiGiandomenico A,
Keller AE, Anude C, Hernandez-Illas M, Bellamy T, Falloon J, Dubovsky gence of vanB E. faecium sequence type 203. J Infect Dis 202:
F, Jafri HS. 2019. Phase 1 study of MEDI3902, an investigational anti- 1278 –1286. https://doi.org/10.1086/656319.
Pseudomonas aeruginosa PcrV and Psl bispecific human monoclonal 476. Lister DM, Kotsanas D, Ballard SA, Howden BP, Carse E, Tan K, Scott C,
antibody, in healthy adults. Clin Microbiol Infect 25:629.e1– 629.e6. Gillespie EE, Mahony AA, Doherty R, Korman TM, Johnson PDR, Stuart
https://doi.org/10.1016/j.cmi.2018.08.004. RL. 2015. Outbreak of vanB vancomycin-resistant Enterococcus faecium
461. EVADE. 2019. Effort to prevent nosocomial pneumonia caused by colonization in a neonatal service. Am J Infect Control 43:1061–1065.
Pseudomonas aeruginosa in mechanically ventilated subjects. https:// https://doi.org/10.1016/j.ajic.2015.05.047.
www.combacte.com/trials/evade/. Accessed 10 January 2020. 477. Ruiz-Garbajosa P, Bonten MJM, Robinson DA, Top J, Nallapareddy SR,
462. Hoggarth A, Weaver A, Pu QQ, Huang T, Schettler J, Chen F, Yuan XF, Torres C, Coque TM, Canton R, Baquero F, Murray BE, del Campo R,
Wu M. 2019. Mechanistic research holds promise for bacterial vaccines Willems R. 2006. Multilocus sequence typing scheme for Enterococcus
and phage therapies for Pseudomonas aeruginosa. Drug Des Devel Ther faecalis reveals hospital-adapted genetic complexes in a background of
13:909 –924. https://doi.org/10.2147/DDDT.S189847. high rates of recombination. J Clin Microbiol 44:2220 –2228. https://doi
463. Rello J, Krenn CG, Locker G, Pilger E, Madl C, Balica L, Dugernier T, .org/10.1128/JCM.02596-05.
Laterre PF, Spapen H, Depuydt P, Vincent JL, Bogar L, Szabo Z, Volgyes 478. McBride SM, Fischetti VA, LeBlanc DJ, Moellering RC, Gilmore MS. 2007.
B, Manez R, Cakar N, Ramazanoglu A, Topeli A, Mastruzzo MA, Jasovich Genetic diversity among Enterococcus faecalis. PLoS One 2:e582.
A, Remolif CG, Soria LD, Hernandez MAA, Balart CR, Kremer I, Molnar Z, https://doi.org/10.1371/journal.pone.0000582.
von Sonnenburg F, Lyons A, Joannidis M, Burgmann H, Welte T, Klingler 479. Freitas AR, Novais C, Ruiz-Garbajosa P, Coque TM, Peixe L. 2009. Clonal
A, Hochreiter R, Westritschnig K. 2017. A randomized placebo- expansion within clonal complex 2 and spread of vancomycin-resistant
controlled phase II study of a Pseudomonas vaccine in ventilated ICU plasmids among different genetic lineages of Enterococcus faecalis
patients. Crit Care 21:22. https://doi.org/10.1186/s13054-017-1601-9. from Portugal. J Antimicrob Chemother 63:1104 –1111. https://doi.org/
464. Jansen KU, Knirsch C, Anderson AS. 2018. The role of vaccines in 10.1093/jac/dkp103.
preventing bacterial antimicrobial resistance. Nat Med 24:10 –19. 480. Erlandson KM, Sun J, Iwen PC, Rupp ME. 2008. Impact of the more-
https://doi.org/10.1038/nm.4465. potent antibiotics quinupristin-dalfopristin and linezolid on outcome
465. Creech CB, Frenck RW, Sheldon EA, Seiden DJ, Kankam MK, Zito ET, measure of patients with vancomycin-resistant Enterococcus bactere-
Girgenti D, Severs JM, Immermann FW, McNeil LK, Cooper D, Jansen KU, mia. Clin Infect Dis 46:30 –36. https://doi.org/10.1086/523588.
Gruber W, Eiden J, Anderson AS, Baber J. 2017. Safety, tolerability, and 481. Zhanel GG, Hoban DJ, Karlowsky JA. 2001. Nitrofurantoin is active
immunogenicity of a single dose 4-antigen or 3-antigen Staphylococcus against vancomycin-resistant enterococci. Antimicrob Agents Che-
aureus vaccine in healthy older adults: results of a randomised trial. mother 45:324 –326. https://doi.org/10.1128/AAC.45.1.324-326.2001.
Vaccine 35:385–394. https://doi.org/10.1016/j.vaccine.2016.11.032. 482. Falagas ME, Vouloumanou EK, Samonis G, Vardakas KZ. 2016. Fosfo-
466. Creech CB, Frenck RW, Fiquet A, Feldman R, Kankam MK, Pathirana S, mycin. Clin Microbiol Rev 29:321–347. https://doi.org/10.1128/CMR
Baber J, Radley D, Cooper D, Eiden J, Gruber WC, Jansen KU, Anderson .00068-15.
AS, Gurtman A. 2020. Persistence of immune responses through 36 483. Linden PK. 2002. Treatment options for vancomycin-resistant entero-
months in healthy adults after vaccination with a novel Staphylococcus coccal infections. Drugs 62:425– 441. https://doi.org/10.2165/00003495
aureus 4-antigen vaccine (SA4Ag). Open Forum Infect Dis 7:ofz532. -200262030-00002.
https://doi.org/10.1093/ofid/ofz532. 484. Wisplinghoff H, Bischoff T, Tallent SM, Seifert H, Wenzel RP, Edmond
467. Seeberger PH, Pereira CL, Khan N, Xiao GZ, Diago-Navarro E, Reppe K, MB. 2004. Nosocomial bloodstream infections in US hospitals: analysis
Opitz B, Fries BC, Witzenrath M. 2017. A semi-synthetic glycoconjugate of 24,179 cases from a prospective nationwide surveillance study. Clin
vaccine candidate for carbapenem-resistant Klebsiella pneumoniae. An- Infect Dis 39:309 –317. https://doi.org/10.1086/421946.
gew Chem Int Ed Engl 56:13973–13978. https://doi.org/10.1002/anie 485. Pinholt M, Ostergaard C, Arpi M, Bruun NE, Schonheyder HC, Gradel KO,
.201700964. Sogaard M, Knudsen JD, Danish Collaborative Bacteraemia Network
468. Ainsworth S, Ketter PM, Yu JJ, Grimm RC, May HC, Cap AP, Chambers JP, (DACOBAN). 2014. Incidence, clinical characteristics and 30-day mor-
Guentzel MN, Arulanandam BP. 2017. Vaccination with a live attenu- tality of enterococcal bacteraemia in Denmark 2006 –2009: a
ated Acinetobacter baumannii deficient in thioredoxin provides protec- population-based cohort study. Clin Microbiol Infect 20:145–151.
tion against systemic Acinetobacter infection. Vaccine 35:3387–3394. https://doi.org/10.1111/1469-0691.12236.
https://doi.org/10.1016/j.vaccine.2017.05.017. 486. Kramer A, Schwebke I, Kampf G. 2006. How long do nosocomial
469. Wang TZ, Kodiyanplakkal RPL, Calfee DP. 2019. Antimicrobial resistance pathogens persist on inanimate surfaces? A systematic review. BMC
in nephrology. Nat Rev Nephrol 15:463– 481. https://doi.org/10.1038/ Infect Dis 6:130. https://doi.org/10.1186/1471-2334-6-130.
s41581-019-0150-7. 487. Santajit S, Indrawattana N. 2016. Mechanisms of antimicrobial resis-
470. Bilinski J, Grzesiowski P, Sorensen N, Madry K, Muszynski J, Robak K, tance in ESKAPE pathogens. Biomed Res Int 2016:2475067. https://doi
Wroblewska M, Dzieciatkowski T, Dulny G, Dwilewicz-Trojaczek J, .org/10.1155/2016/2475067.
Wiktor-Jedrzejczak W, Basak GW. 2017. Fecal microbiota transplanta- 488. Turner NA, Sharma-Kuinkel BK, Maskarinec SA, Eichenberger EM, Shah
tion in patients with blood disorders inhibits gut colonization with PP, Carugati M, Holland TL, Fowler VG. 2019. Methicillin-resistant Staph-
antibiotic-resistant bacteria: results of a prospective, single-center ylococcus aureus: an overview of basic and clinical research. Nat Rev
study. Clin Infect Dis 65:364 –370. https://doi.org/10.1093/cid/cix252. Microbiol 17:203–218. https://doi.org/10.1038/s41579-018-0147-4.
471. Remschmidt C, Schroder C, Behnke M, Gastmeier P, Geffers C, Kramer 489. Harkins CP, Pichon B, Doumith M, Parkhill J, Westh H, Tomasz A, De
TS. 2018. Continuous increase of vancomycin resistance in enterococci Lencastre H, Bentley SD, Kearns AM, Holden M. 2017. Methicillin-
causing nosocomial infections in Germany—10 years of surveillance. resistant Staphylococcus aureus emerged long before the introduction
Antimicrob Resist Infect Control 7:54. https://doi.org/10.1186/s13756 of methicillin into clinical practice. Genome Biol 18:130. https://doi.org/
-018-0353-x. 10.1186/s13059-017-1252-9.
472. Arias CA, Murray BE. 2008. Emergence and management of drug- 490. Corey GR, Arhin FF, Wikler MA, Sahm DF, Kreiswirth BN, Mediavilla JR,
resistant enterococcal infections. Expert Rev Anti Infect Ther Good S, Fiset C, Jiang H, Moeck G, Kabler H, Green S, O’Riordan W,
6:637– 655. https://doi.org/10.1586/14787210.6.5.637. SOLO I, SOLO II Investigators. 2016. Pooled analysis of single-dose
473. O’Driscoll T, Crank CW. 2015. Vancomycin-resistant enterococcal oritavancin in the treatment of acute bacterial skin and skin-structure
infections: epidemiology, clinical manifestations, and optimal man- infections caused by Gram-positive pathogens, including a large pa-
agement. Infect Drug Resist 8:217–230. https://doi.org/10.2147/IDR tient subset with methicillin-resistant Staphylococcus aureus. Int J An-
.S54125. timicrob Agents 48:528 –534. https://doi.org/10.1016/j.ijantimicag.2016
474. Mioljević V, Marković-Denić L, Vidović A, Jovanović M, Tosić T, Tomin D. .07.019.
2013. Risk factors for vancomycin-resistant Enterococcus colonization in 491. Rubinstein E, Kollef MH, Nathwani D. 2008. Pneumonia caused by
hematologic patients. Vojnosanit Pregl 70:1109 –1116. https://doi.org/ methicillin-resistant Staphylococcus aureus. Clin Infect Dis 46(Suppl
10.2298/vsp1312109m. 5):S378 –S385. https://doi.org/10.1086/533594.
475. Johnson PDR, Ballard SA, Grabsch EA, Stinear TP, Seemann T, Young HL, 492. McNeil JC, Vallejo JG, Hulten KG, Kaplan SL. 2018. Osteoarticular infec-
Grayson ML, Howden BP. 2010. A sustained hospital outbreak of tions following open or penetrating trauma in children in the post-
vancomycin-resistant Enterococcus faecium bacteremia due to emer- community-acquired methicillin-resistant Staphylococcus aureus era:
the impact of Enterobacter cloacae. Pediatr Infect Dis J 37:1204 –1210. Fridkin S, Emerging Infections Program-Active Bacterial Core Sur-
https://doi.org/10.1097/INF.0000000000001991. veillance MRSA Surveillance Investigators. 2013. National burden of
493. Chuang YY, Huang YC. 2013. Molecular epidemiology of community- invasive methicillin-resistant Staphylococcus aureus infections,
associated meticillin-resistant Staphylococcus aureus in Asia. Lancet United States, 2011. JAMA Intern Med 173:1970 –1978. https://doi
Infect Dis 13:698 –708. https://doi.org/10.1016/S1473-3099(13)70136-1. .org/10.1001/jamainternmed.2013.10423.
494. Liu YD, Wang H, Du N, Shen EH, Chen HB, Niu JQ, Ye HF, Chen MJ. 2009. 509. Tzouvelekis LS, Markogiannakis A, Piperaki E, Souli M, Daikos GL. 2014.
Molecular evidence for spread of two major methicillin-resistant Staph- Treating infections caused by carbapenemase-producing Enterobacte-
ylococcus aureus clones with a unique geographic distribution in Chi- riaceae. Clin Microbiol Infect 20:862– 872. https://doi.org/10.1111/1469
nese hospitals. Antimicrob Agents Chemother 53:512–518. https://doi -0691.12697.
.org/10.1128/AAC.00804-08. 510. Gu DX, Dong N, Zheng ZW, Lin D, Huang M, Wang LH, Chan EWC, Shu
495. Zhao CJ, Liu YM, Zhao MZ, Liu YL, Yu Y, Chen HB, Sun QN, Chen HW, LB, Yu J, Zhang R, Chen S. 2018. A fatal outbreak of ST11 carbapenem-
Jiang W, Liu YD, Han SM, Xu YC, Chen MJ, Cao B, Wang H. 2012. resistant hypervirulent Klebsiella pneumoniae in a Chinese hospital: a
Characterization of community acquired Staphylococcus aureus associ- molecular epidemiological study. Lancet Infect Dis 18:37– 46. https://
ated with skin and soft tissue infection in Beijing: high prevalence of doi.org/10.1016/S1473-3099(17)30489-9.
PVL⫹ ST398. PLoS One 7:e38577. https://doi.org/10.1371/journal.pone 511. Hsieh PF, Lin TL, Lee CZ, Tsai SF, Wang JT. 2008. Serum-induced
.0038577. iron-acquisition systems and TonB contribute to virulence in Klebsiella
496. Song JH, Hsueh PR, Chung DR, Ko KS, Kang CI, Peck KR, Yeom JS, Kim SW, pneumoniae causing primary pyogenic liver abscess. J Infect Dis 197:
Chang HH, Kim YS, Jung SI, Son JS, So TMK, Lalitha MK, Yang YH, Huang 1717–1727. https://doi.org/10.1086/588383.
SG, Wang H, Lu QA, Carlos CC, Perera JA, Chiu CH, Liu JW, Chongthaleong 512. Siu LK, Yeh KM, Lin JC, Fung CP, Chang FY. 2012. Klebsiella pneumoniae
A, Thamlikitkul V, Van PH, ANSORP Study Group. 2011. Spread of liver abscess: a new invasive syndrome. Lancet Infect Dis 12:881– 887.
methicillin-resistant Staphylococcus aureus between the community and https://doi.org/10.1016/S1473-3099(12)70205-0.
the hospitals in Asian countries: an ANSORP study. J Antimicrob Che- 513. Quan JJ, Zhao DD, Liu LL, Chen Y, Zhou JC, Jiang Y, Du XX, Zhou ZH,
mother 66:1061–1069. https://doi.org/10.1093/jac/dkr024. Akova M, Yu YS. 2017. High prevalence of ESBL-producing Escherichia
497. Sahibzada S, Abraham S, Coombs GW, Pang S, Hernandez-Jover M, coli and Klebsiella pneumoniae in community-onset bloodstream infec-
Jordan D, Heller J. 2017. Transmission of highly virulent community- tions in China. J Antimicrob Chemother 72:273–280. https://doi.org/10
associated MRSA ST93 and livestock-associated MRSA ST398 between .1093/jac/dkw372.
humans and pigs in Australia. Sci Rep 7:5273. https://doi.org/10.1038/ 514. Hu L, Liu Y, Deng L, Zhong Q, Hang Y, Wang Z, Zhan L, Wang L, Yu F.
s41598-017-04789-0. 2016. Outbreak by ventilator-associated ST11 K. pneumoniae with co-
498. van Hal SJ, Jensen SO, Vaska VL, Espedido BA, Paterson DL, Gosbell IB. production of CTX-M-24 and KPC-2 in a SICU of a tertiary teaching
2012. Predictors of mortality in Staphylococcus aureus bacteremia. Clin hospital in central China. Front Microbiol 7:1190. https://doi.org/10
Microbiol Rev 25:362–386. https://doi.org/10.1128/CMR.05022-11. .3389/fmicb.2016.01190.
515. Ewers C, Stamm I, Pfeifer Y, Wieler LH, Kopp PA, Schønning K, Prenger-
499. Dillon HC, Derrick CW. 1975. Clinical experience with clindamycin
Berninghoff E, Scheufen S, Stolle I, Günther S, Bethe A. 2014. Clonal
hydrochloride: 1. Treatment of streptococcal and mixed streptococcal-
spread of highly successful ST15-CTX-M-15 Klebsiella pneumoniae in
staphylococcal skin infections. Pediatrics 55:205–212.
companion animals and horses. J Antimicrob Chemother 69:
500. Fowler VG, Boucher HW, Corey GR, Abrutyn E, Karchmer AW, Rupp ME,
2676 –2680. https://doi.org/10.1093/jac/dku217.
Levine DP, Chambers HF, Tally FP, Vigliani GA, Cabell CH, Link AS,
516. Navon-Venezia S, Kondratyeva K, Carattoli A. 2017. Klebsiella
DeMeyer I, Filler SG, Zervos M, Cook P, Parsonnet J, Bernstein JM, Price
pneumoniae: a major worldwide source and shuttle for antibiotic resis-
CS, Forrest GN, Fätkenheuer G, Gareca M, Rehm SJ, Brodt HR, Tice A,
tance. FEMS Microbiol Rev 41:252–275. https://doi.org/10.1093/femsre/
Cosgrove SE, S. aureus Endocarditis and Bacteremia Study Group. 2006.
fux013.
Daptomycin versus standard therapy for bacteremia and endocarditis
517. Frasson I, Lavezzo E, Franchin E, Toppo S, Barzon L, Cavallaro A, Richter
caused by Staphylococcus aureus. N Engl J Med 355:653– 665. https://
SN, Palu G. 2012. Antimicrobial treatment and containment measures
doi.org/10.1056/NEJMoa053783.
for an extremely drug-resistant Klebsiella pneumoniae ST101 isolate
501. Weigelt J, Itani K, Stevens D, Lau W, Dryden M, Knirsch C, Linezolid CSSTI
carrying pKPN101-IT, a novel fully sequenced bla(KPC-2) plasmid. J Clin
Study Group. 2005. Linezolid versus vancomycin in treatment of compli- Microbiol 50:3768 –3772. https://doi.org/10.1128/JCM.01892-12.
cated skin and soft tissue infections. Antimicrob Agents Chemother 49: 518. D’Andrea MM, Arena F, Pallecchi L, Rossolini GM. 2013. CTX-M-type
2260–2266. https://doi.org/10.1128/AAC.49.6.2260-2266.2005. beta-lactamases: a successful story of antibiotic resistance. Int J Med
502. Shorr AF, Lodise TP, Corey GR, De Anda C, Fang E, Das AF, Prokocimer P. Microbiol 303:305–317. https://doi.org/10.1016/j.ijmm.2013.02.008.
2015. Analysis of the phase 3 ESTABLISH trials of tedizolid versus linezolid 519. Wyres KL, Holt KE. 2016. Klebsiella pneumoniae population genomics
in acute bacterial skin and skin structure infections. Antimicrob Agents and antimicrobial-resistant clones. Trends Microbiol 24:944 –956.
Chemother 59:864–871. https://doi.org/10.1128/AAC.03688-14. https://doi.org/10.1016/j.tim.2016.09.007.
503. Boucher HW, Wilcox M, Talbot GH, Puttagunta S, Das AF, Dunne MW. 520. López-Camacho E, Paño-Pardo JR, Ruiz-Carrascoso G, Wesselink J-J,
2014. Once-weekly dalbavancin versus daily conventional therapy for Lusa-Bernal S, Ramos-Ruiz R, Ovalle S, Gómez-Gil R, Pérez-Blanco V,
skin infection. N Engl J Med 370:2169 –2179. https://doi.org/10.1056/ Pérez-Vázquez M, Gómez-Puertas P, Mingorance J. 2018. Population
NEJMoa1310480. structure of OXA-48-producing Klebsiella pneumoniae ST405 isolates
504. Tanaseannu C, Bergallo C, Teglia O, Jasovich A, Oliva ME, Dukart G, during a hospital outbreak characterised by genomic typing. J Glob
Dartois N, Cooper CA, Gandjini H, Mallick R, 303 and 318 Study Groups. Antimicrob Resist 15:48 –54. https://doi.org/10.1016/j.jgar.2018.06.008.
2008. Integrated results of 2 phase 3 studies comparing tigecycline and 521. Tumbarello M, Trecarichi EM, De Rosa FG, Giannella M, Giacobbe DR,
levofloxacin in community-acquired pneumonia. Diagn Microbiol Infect Bassetti M, Losito AR, Bartoletti M, Del Bono V, Corcione S, Maiuro G,
Dis 61:329 –338. https://doi.org/10.1016/j.diagmicrobio.2008.04.009. Tedeschi S, Celani L, Cardellino CS, Spanu T, Marchese A, Ambretti S,
505. Paul M, Bishara J, Yahav D, Goldberg E, Neuberger A, Ghanem-Zoubi N, Cauda R, Viscoli C, Viale P, ISGRI-SITA (Italian Study Group on Resistant
Dickstein Y, Nseir W, Dan M, Leibovici L. 2015. Trimethoprim- Infections of the Società Italiana Terapia Antinfettiva). 2015. Infections
sulfamethoxazole versus vancomycin for severe infections caused by caused by KPC-producing Klebsiella pneumoniae: differences in therapy
meticillin resistant Staphylococcus aureus: randomised controlled trial. and mortality in a multicentre study. J Antimicrob Chemother 70:
BMJ 350:h2219. https://doi.org/10.1136/bmj.h2219. 2133–2143. https://doi.org/10.1093/jac/dkv086.
506. Ng J, Gosbell IB. 2005. Successful oral pristinamycin therapy for osteo- 522. Hauck C, Cober E, Richter SS, Perez F, Salata RA, Kalayjian RC, Watkins
articular infections due to methicillin-resistant Staphylococcus aureus RR, Scalera NM, Doi Y, Kaye KS, Evans S, Fowler VG, Bonomo RA, van
(MRSA) and other Staphylococcus spp. J Antimicrob Chemother 55: Duin D. 2016. Spectrum of excess mortality due to carbapenem-
1008 –1012. https://doi.org/10.1093/jac/dki108. resistant Klebsiella pneumoniae infections. Clin Microbiol Infect 22:
507. Chen CJ, Huang YC. 2014. New epidemiology of Staphylococcus aureus 513–519. https://doi.org/10.1016/j.cmi.2016.01.023.
infection in Asia. Clin Microbiol Infect 20:605– 623. https://doi.org/10 523. Paterson DL, Ko WC, Von Gottberg A, Mohapatra S, Casellas JM, Goos-
.1111/1469-0691.12705. sens H, Mulazimoglu L, Trenholme G, Klugman KP, Bonomo RA, Rice LB,
508. Dantes R, Mu Y, Belflower R, Aragon D, Dumyati G, Harrison LH, Wagener MM, McCormack JG, Yu VL. 2004. Antibiotic therapy for
Lessa FC, Lynfield R, Nadle J, Petit S, Ray SM, Schaffner W, Townes J, Klebsiella pneumoniae bacteremia: implications of production of
extended-spectrum beta-lactamases. Clin Infect Dis 39:31–37. https:// stream infections: analysis of trends in prevalence and epidemiology.
doi.org/10.1086/420816. Emerg Infect Dis 8:220 –221. https://doi.org/10.3201/eid0802.010121.
524. Souli M, Galani I, Antoniadou A, Papadomichelakis E, Poulakou G, 540. Lamas Ferreiro JL, Álvarez Otero J, González González L, Novoa Lamaza-
Panagea T, Vourli S, Zerva L, Armaganidis A, Kanellakopoulou K, Gia- res L, Arca Blanco A, Bermúdez Sanjurjo JR, Rodríguez Conde I, Fernán-
marellou H. 2010. An outbreak of infection due to beta-lactamase dez Soneira M, de la Fuente Aguado J. 2017. Pseudomonas aeruginosa
Klebsiella pneumoniae carbapenemase 2-producing K. pneumoniae in a urinary tract infections in hospitalized patients: mortality and prognos-
Greek university hospital: molecular characterization, epidemiology, tic factors. PLoS One 12:e0178178. https://doi.org/10.1371/journal
and outcomes. Clin Infect Dis 50:364 –373. https://doi.org/10.1086/ .pone.0178178.
649865. 541. Marvig RL, Sommer LM, Molin S, Johansen HK. 2015. Convergent
525. Petty LA, Henig O, Patel TS, Pogue JM, Kaye KS. 2018. Overview of evolution and adaptation of Pseudomonas aeruginosa within patients
meropenem-vaborbactam and newer antimicrobial agents for the with cystic fibrosis. Nat Genet 47:57– 64. https://doi.org/10.1038/ng
treatment of carbapenem-resistant Enterobacteriaceae. Infect Drug Re- .3148.
sist 11:1461–1472. https://doi.org/10.2147/IDR.S150447. 542. Dou Y, Huan JN, Guo F, Zhou ZD, Shi Y. 2017. Pseudomonas aeruginosa
526. El Nekidy WS, Mooty MY, Attallah N, Cardona L, Bonilla MF, Ghazi IM. prevalence, antibiotic resistance and antimicrobial use in Chinese burn
2017. Successful treatment of multidrug resistant Klebsiella pneumoniae wards from 2007 to 2014. J Int Med Res 45:1124 –1137. https://doi.org/
using dual carbapenem regimen in immunocompromised patient. 10.1177/0300060517703573.
IDCases 9:53–55. https://doi.org/10.1016/j.idcr.2017.06.005. 543. Dapas JI, Rivero C, Burgos P, Vila A. 2016. Pseudomonas aeruginosa
527. Shirley M. 2018. Ceftazidime-avibactam: a review in the treatment of infective endocarditis following aortic valve implantation: a note of
serious Gram-negative bacterial infections. Drugs 78:675– 692. https:// caution. Open Cardiovasc Med J 10:28 –34. https://doi.org/10.2174/
doi.org/10.1007/s40265-018-0902-x. 1874192401610010028.
528. Weiner LM, Webb AK, Limbago B, Dudeck MA, Patel J, Kallen AJ, 544. Aguilar-Rodea P, Zúñiga G, Rodríguez-Espino BA, Olivares Cervantes AL,
Edwards JR, Sievert DM. 2016. Antimicrobial-resistant pathogens asso- Gamiño Arroyo AE, Moreno-Espinosa S, de la Rosa Zamboni D, López
ciated with healthcare-associated infections: summary of data reported Martínez B, Castellanos-Cruz MDC, Parra-Ortega I, Jiménez Rojas VL,
to the National Healthcare Safety Network at the Centers for Disease Vigueras Galindo JC, Velázquez-Guadarrama N. 2017. Identification of
Control and Prevention, 2011–2014. Infect Control Hosp Epidemiol extensive drug resistant Pseudomonas aeruginosa strains: new clone
37:1288 –1301. https://doi.org/10.1017/ice.2016.174. ST1725 and high-risk clone ST233. PLoS One 12:e0172882. https://doi
529. Kim B-N, Peleg AY, Lodise TP, Lipman J, Li J, Nation R, Paterson DL. .org/10.1371/journal.pone.0172882.
2009. Management of meningitis due to antibiotic-resistant Acineto- 545. Maatallah M, Cheriaa J, Backhrouf A, Iversen A, Grundmann H, Do T,
bacter species. Lancet Infect Dis 9:245–255. https://doi.org/10.1016/ Lanotte P, Mastouri M, Elghmati MS, Rojo F, Mejdi S, Giske CG. 2011.
S1473-3099(09)70055-6. Population structure of Pseudomonas aeruginosa from five Mediterra-
530. Sievert DM, Ricks P, Edwards JR, Schneider A, Patel J, Srinivasan A, nean countries: evidence for frequent recombination and epidemic
occurrence of CC235. PLoS One 6:e25617. https://doi.org/10.1371/
Kallen A, Limbago B, Fridkin S, National Healthcare Safety Network
journal.pone.0025617.
(NHSN) Team and Participating NHSN Facilities. 2013. Antimicrobial-
546. Bassetti M, Castaldo N, Cattelan A, Mussini C, Righi E, Tascini C,
resistant pathogens associated with healthcare-associated infections:
Menichetti F, Mastroianni CM, Tumbarello M, Grossi P, Artioli S, Caran-
summary of data reported to the National Healthcare Safety Network at
nante N, Cipriani L, Coletto D, Russo A, Digaetano M, Losito AR, Peghin
the Centers for Disease Control and Prevention, 2009 –2010. Infect
M, Capone A, Nicole S, Vena A, CEFTABUSE Study Group. 2019.
Control Hosp Epidemiol 34:1–14. https://doi.org/10.1086/668770.
Ceftolozane/tazobactam for the treatment of serious Pseudomonas
531. Zhou K, Tang X, Wang L, Guo Z, Xiao S, Wang Q, Zhuo C. 2018. An
aeruginosa infections: a multicentre nationwide clinical experience. Int
emerging clone (ST457) of Acinetobacter baumannii clonal complex 92
J Antimicrob Agents 53:408 – 415. https://doi.org/10.1016/j.ijantimicag
with enhanced virulence and increasing endemicity in south China.
.2018.11.001.
Clin Infect Dis 67:S179 –S188. https://doi.org/10.1093/cid/ciy691.
547. Fujii A, Seki M, Higashiguchi M, Tachibana I, Kumanogoh A, Tomono K.
532. van Dessel H, Dijkshoorn L, van der Reijden T, Bakker N, Paauw A, van
2014. Community-acquired, hospital-acquired, and healthcare-
den Broek P, Verhoef J, Brisse S. 2004. Identification of a new geo-
associated pneumonia caused by Pseudomonas aeruginosa. Respir Med
graphically widespread multiresistant Acinetobacter baumannii clone Case Rep 12:30 –33. https://doi.org/10.1016/j.rmcr.2014.03.002.
from European hospitals. Res Microbiol 155:105–112. https://doi.org/ 548. Sligl WI, Dragan T, Smith SW. 2015. Nosocomial Gram-negative bacte-
10.1016/j.resmic.2003.10.003. remia in intensive care: epidemiology, antimicrobial susceptibilities,
533. Antunes LCS, Visca P, Towner KJ. 2014. Acinetobacter baumannii: evo- and outcomes. Int J Infect Dis 37:129 –134. https://doi.org/10.1016/j.ijid
lution of a global pathogen. Pathog Dis 71:292–301. https://doi.org/10 .2015.06.024.
.1111/2049-632X.12125. 549. Oncul O, Oksuz S, Acar A, Ulkur E, Turhan V, Uygur F, Ulcay A, Erdem H,
534. Lăzureanu V, Porosnicu
, M, Gândac C, Moisil T, Bădit,oiu L, Laza R, Musta Ozyurt M, Gorenek L. 2014. Nosocomial infection characteristics in a
V, Crisan
, A, Marinescu A-R. 2016. Infection with Acinetobacter bauman- burn intensive care unit: analysis of an eleven-year active surveillance.
nii in an intensive care unit in the western part of Romania. BMC Infect Burns 40:835– 841. https://doi.org/10.1016/j.burns.2013.11.003.
Dis 16(Suppl 1):95. https://doi.org/10.1186/s12879-016-1399-0. 550. Lund-Palau H, Turnbull AR, Bush A, Bardin E, Cameron L, Soren O,
535. Norton MD, Spilkia AJ, Godoy VG. 2013. Antibiotic resistance acquired Wierre-Gore N, Alton EWFW, Bundy JG, Connett G, Faust SN, Filloux A,
through a DNA damage-inducible response in Acinetobacter bauman- Freemont P, Jones A, Khoo V, Morales S, Murphy R, Pabary R, Simbo A,
nii. J Bacteriol 195:1335–1345. https://doi.org/10.1128/JB.02176-12. Schelenz S, Takats Z, Webb J, Williams HD, Davies JC. 2016. Pseudomo-
536. Thompson MG, Black CC, Pavlicek RL, Honnold CL, Wise MC, Alamneh nas aeruginosa infection in cystic fibrosis: pathophysiological mecha-
YA, Moon JK, Kessler JL, Si YZ, Williams R, Yildirim S, Kirkup BC, Green nisms and therapeutic approaches. Expert Rev Respir Med 10:685– 697.
RK, Hall ER, Palys TJ, Zurawski DV. 2014. Validation of a novel murine https://doi.org/10.1080/17476348.2016.1177460.
wound model of Acinetobacter baumannii infection. Antimicrob Agents 551. Edlin R, Shapiro D, Hersh A, Copp H. 2013. Antibiotic resistance patterns
Chemother 58:1332–1342. https://doi.org/10.1128/AAC.01944-13. in outpatient pediatric urinary tract infections. J Urol 190:222–227.
537. Nwugo CC, Arivett BA, Zimbler DL, Gaddy JA, Richards AM, Actis LA. https://doi.org/10.1016/j.juro.2013.01.069.
2012. Effect of ethanol on differential protein production and expres- 552. Chen CH, Huang CC. 2013. Risk factor analysis for extended-spectrum
sion of potential virulence functions in the opportunistic pathogen beta-lactamase-producing Enterobacter cloacae bloodstream infections
Acinetobacter baumannii. PLoS One 7:e51936. https://doi.org/10.1371/ in central Taiwan. BMC Infect Dis 13:417. https://doi.org/10.1186/1471
journal.pone.0051936. -2334-13-417.
538. Hassan KA, Jackson SM, Penesyan A, Patching SG, Tetu SG, Eijkelkamp 553. Wang HJ, Shi H, Zhou W, Hu ZQ, Mu LY, Su M, Jiang YM. 2012. Common
BA, Brown MH, Henderson PJF, Paulsen IT. 2013. Transcriptomic and pathogens and clinical characteristics of neonatal pneumonia. Zhong-
biochemical analyses identify a family of chlorhexidine efflux proteins. guo Dang Dai Er Ke Za Zhi 14:898 –902.
Proc Natl Acad Sci U S A 110:20254 –20259. https://doi.org/10.1073/ 554. Múñez E, Ramos A, Alvarez de Espejo T, Vaqué J, Sánchez-Payá J, Pastor
pnas.1317052110. V, Asensio A. 2012. Aetiology of surgical infections in patients under-
539. Tacconelli E, Tumbarello M, Bertagnolio S, Citton R, Spanu T, Fadda G, going craniotomy. Neurocirugia (Astur) 23:54 –59. https://doi.org/10
Cauda R. 2002. Multidrug-resistant Pseudomonas aeruginosa blood- .1016/j.neucir.2011.08.002.
555. Fata F, Chittivelu S, Tessler S, Kupfer Y. 1996. Gas gangrene of the arm genetic elements and their contribution to the emergence of antimi-
due to Enterobacter cloacae in a neutropenic patient. South Med J crobial resistant Enterococcus faecalis and Enterococcus faecium. Clin
89:1095–1096. Microbiol Infect 16:541–554. https://doi.org/10.1111/j.1469-0691.2010
556. Sanders WE, Sanders CC. 1997. Enterobacter spp.: pathogens poised to .03226.x.
flourish at the turn of the century. Clin Microbiol Rev 10:220 –241. 566. Fujimura T, Murakami K. 2008. Staphylococcus aureus clinical isolate
557. Izdebski R, Baraniak A, Herda M, Fiett J, Bonten MJ, Carmeli Y, Goossens with high-level methicillin resistance with an lytH mutation caused by
H, Hryniewicz W, Brun-Buisson C, Gniadkowski M, MOSAR WP2, WP3 IS1182 insertion. Antimicrob Agents Chemother 52:643– 647. https://
and WP5 Study Groups. 2015. MLST reveals potentially high-risk inter- doi.org/10.1128/AAC.00395-07.
national clones of Enterobacter cloacae. J Antimicrob Chemother 70: 567. Maki H, McCallum N, Bischoff M, Wada A, Berger-Bächi B. 2004. tcaA
48 –56. https://doi.org/10.1093/jac/dku359. inactivation increases glycopeptide resistance in Staphylococcus aureus.
558. Lin YC, Chen TL, Ju HL, Chen HS, Wang FD, Yu KW, Liu CY. 2006. Clinical Antimicrob Agents Chemother 48:1953–1959. https://doi.org/10.1128/
characteristics and risk factors for attributable mortality in Enterobacter AAC.48.6.1953-1959.2004.
cloacae bacteremia. J Microbiol Immunol Infect 39:67–72. 568. Partridge SR, Hall RM. 2003. In34, a complex In5 family class 1 integron
559. Hunter CJ, Petrosyan M, Ford HR, Prasadarao NV. 2008. Enterobacter containing orf513 and dfrA10. Antimicrob Agents Chemother 47:
sakazakii: an emerging pathogen in infants and neonates. Surg Infect 342–349. https://doi.org/10.1128/aac.47.1.342-349.2003.
(Larchmt) 9:533–539. https://doi.org/10.1089/sur.2008.006. 569. Toleman MA, Walsh TR. 2008. Evolution of the ISCR3 group of ISCR
560. Robredo B, Torres C, Singh KV, Murray BE. 2000. Molecular analysis of
elements. Antimicrob Agents Chemother 52:3789 –3791. https://doi
Tn1546 in vanA-containing Enterococcus spp. isolated from humans
.org/10.1128/AAC.00479-08.
and poultry. Antimicrob Agents Chemother 44:2588 –2589. https://doi
570. Mussi MA, Limansky AS, Viale AM. 2005. Acquisition of resistance to
.org/10.1128/aac.44.9.2588-2589.2000.
carbapenems in multidrug-resistant clinical strains of Acinetobacter
561. Simjee S, White DG, McDermott PF, Wagner DD, Zervos MJ, Donabe-
baumannii: natural insertional inactivation of a gene encoding a mem-
dian SM, English LL, Hayes JR, Walker RD. 2002. Characterization of
ber of a novel family of beta-barrel outer membrane proteins. Antimi-
Tn1546 in vancomycin-resistant Enterococcus faecium isolated from
canine urinary tract infections: evidence of gene exchange between crob Agents Chemother 49:1432–1440. https://doi.org/10.1128/AAC.49
human and animal enterococci. J Clin Microbiol 40:4659 – 4665. https:// .4.1432-1440.2005.
doi.org/10.1128/jcm.40.12.4659-4665.2002. 571. Wang Y, Tong MK, Chow KH, Cheng VCC, Tse CWS, Wu AKL, Lai RWM,
562. Schwarz FV, Perreten V, Teuber M. 2001. Sequence of the 50-kb con- Luk WK, Tsang DNC, Ho PL. 2018. Occurrence of highly conjugative
jugative multiresistance plasmid pRE25 from Enterococcus faecalis IncX3 epidemic plasmid carrying bla(NDM) in Enterobacteriaceae iso-
RE25. Plasmid 46:170 –187. https://doi.org/10.1006/plas.2001.1544. lates in geographically widespread areas. Front Microbiol 9:2272.
563. Liu Y, Wang Y, Wu C, Shen Z, Schwarz S, Du XD, Dai L, Zhang W, Zhang 572. Boutoille D, Corvec S, Caroff N, Giraudeau C, Espaze E, Caillon J, Plesiat
Q, Shen J. 2012. First report of the multidrug resistance gene cfr in P, Reynaud A. 2004. Detection of an IS21 insertion sequence in the
Enterococcus faecalis of animal origin. Antimicrob Agents Chemother mexR gene of Pseudomonas aeruginosa increasing beta-lactam resis-
56:1650 –1654. https://doi.org/10.1128/AAC.06091-11. tance. FEMS Microbiol Lett 230:143–146. https://doi.org/10.1016/S0378
564. Lim SK, Tanimoto K, Tomita H, Ike Y. 2006. Pheromone-responsive -1097(03)00882-6.
conjugative vancomycin resistance plasmids in Enterococcus faecalis 573. Evans JC, Segal H. 2007. A novel insertion sequence, ISPa26, in oprD of
isolates from humans and chicken feces. Appl Environ Microbiol 72: Pseudomonas aeruginosa is associated with carbapenem resistance.
6544 – 6553. https://doi.org/10.1128/AEM.00749-06. Antimicrob Agents Chemother 51:3776 –3777. https://doi.org/10.1128/
565. Hegstad K, Mikalsen T, Coque TM, Werner G, Sundsfjord A. 2010. Mobile AAC.00837-07.