RuizJ ClinMicrobiolRev201932e00007-1
RuizJ ClinMicrobiolRev201932e00007-1
RuizJ ClinMicrobiolRev201932e00007-1
net/publication/335170533
CITATIONS READS
71 1,360
1 author:
Joaquim Ruiz
Universidad Científica del Sur
308 PUBLICATIONS 9,208 CITATIONS
SEE PROFILE
All content following this page was uploaded by Joaquim Ruiz on 28 August 2020.
crossm
a
Universidad Continental, Lima, Peru
SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Quinolones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Utility of quinolones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Adverse events . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Evolution of Quinolone Resistance over Time . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Quinolone Resistance Mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
GENERAL OVERVIEW OF TMQR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Nomenclature of the Transferable Mechanisms of Quinolone Resistance . . . . . . . . . . . . . . . . 9
Is it correct to use the term plasmid-mediated quinolone resistance? . . . . . . . . . . . . . . . . 10
TMQR misidentification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Changing Resistance Paradigms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Clinical Relevance of TMQR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Molecular Epidemiology of TMQR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Qnr . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Qnr Classification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Qnr Families . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
QnrA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
QnrB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
QnrC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
QnrD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
QnrE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
QnrS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
QnrVC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Chromosomal Qnr . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Qnr Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Original Qnr Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Mechanisms of Qnr Action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
In vitro mutations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
QUINOLONE MODIFICATION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
AAC(6=)Ib-cr . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
AAC(6=)Ib-cr subtypes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
Other Quinolone Modification Enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
TRANSFERABLE EFFLUX PUMPS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
OqxAB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
QepA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
QacA and QacB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Other Transferable Efflux Pumps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
TMQR AND THE FUTURE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
CONCLUSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
ACKNOWLEDGMENTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
AUTHOR BIO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
INTRODUCTION
Research on transferable mechanisms of quinolone resistance (TMQR) (see “Is it correct
to use the term plasmid-mediated quinolone resistance?” below, for an explanation
about the use of this term and acronym) is almost as old as quinolones themselves
(1–3). Nonetheless, despite some sporadic unconfirmed descriptions of TMQR (2–6) and
the presence of transferable mechanisms of resistance related to a slowdown of the
bacterial duplication time associated with a 2.2-kb region of the plasmid pKM101
containing the korB, traL, korA, and traM genes (7), TMQR remained undetected. In most
of the early studies in which quinolone resistance transfer was claimed, transconjugants
were selected with nalidixic acid, which possesses a relatively high frequency of
mutation (8, 9). Furthermore, some studies used low MICs to select nalidixic acid-
resistant transconjugants as nalidixic acid resistant (e.g., 8 g/ml in a study by Jonsson
[2]). These findings suggest that the selection of spontaneous mutants rather than the
presence of true transconjugants is the most feasible scenario. This led to a debate
about the feasibility of TMQR development (10, 11). Indeed, some studies on TMQR
were further reanalyzed, and it was proposed that plasmids act as a mutator factor able
to induce the development of nalidixic acid resistance (12), and the presence of
spontaneous quinolone target mutations (10) showed a lack of transfer of nalidixic acid
resistance (13).
Thus, for 36 years after the first nalidixic acid description (14), TMQR remained a
unicorn or a vanishing hitchhiker, a myth or urban legend, until 1998, when the
presence of TMQR was first unequivocally demonstrated (15).
Quinolones
Quinolones are synthetic products that were first synthetized in 1949 (16). There-
after, a high number of derivatives and related substances were developed, some of
which showed antibacterial properties. Although some quinolone derivative molecules
were patented in the late 1950s, it is largely considered that the quinolone era began
in 1962, with the synthesis of nalidixic acid (14, 17–19). The first clinical trial reports on
the use of nalidixic acid are from 1963 (20–23). Subsequently, nalidixic acid was
introduced into clinical practice as early as 1964 (18, 24, 25) albeit limited to the
treatment of urinary tract infections (26). Despite this limitation, this agent has played
a role in the treatment of other infections, such as those of the gastrointestinal tract,
especially in some developing areas (27–30). In subsequent years, the quinolone family
grew, and some of its members were introduced into the antibacterial clinical arma-
mentarium; these include oxolinic acid (31), piromidic acid (32), cinoxacin (33), and
pipemidic acid (34), among others. Although first proposed in 1960 (17), the next step
in the history of quinolones was the addition of a fluorine atom, which opened the door
to the fluoroquinolone era. Norfloxacin is considered the starting point of this era (35),
despite the fluorine atom first being present in another quinolone introduced into
clinical practice, flumequine (36). This fluorine atom was thereafter maintained in
almost all the quinolones introduced into clinical practice, largely expanding their
bacterial spectra and levels of activity (37). Nonetheless, some recent quinolones, a few
of which were introduced into clinical practice only a few years ago (e.g., garenoxacin,
nemonoxacin, and ozenoxacin), lack the fluorine atom substituent at position 6,
although they may present a fluorine atom(s) in other positions (for instance, the
garenoxacin molecule possesses an OCHF2 group in position 8), usually referred to as
“desfluoroquinolones” or “nonfluorinated quinolones” (38–42). Finally, some recently
developed molecules present antibiotic hybrid characteristics, such as cadazolid, a
quinoxolidinone (fluoroquinolone-oxazolidone hybrid molecule) that has been inves-
tigated to treat Clostridium difficile infections (43) (Fig. 1).
In summary, since the beginning of the quinolone era, more than 10,000 quinolones
have been synthetized, and their activities and properties have been explored (25, 44,
45), with more than 40 being approved for either human or veterinary applications.
Currently, 21 quinolones remain in human use in the European Union and/or the
United States (Fig. 2), while several others are used in other countries; for instance,
balofloxacin has been approved for human use in South Korea and India (46, 47).
Despite the fact that the classification of quinolones, or other antibacterial agents,
in “generations” is imprecise and subject to different interpretations, the quinolones
have traditionally been classified into 4 generations based on their spectrum of activity
(48, 49) (see Table 1 for definitions used in this article). In addition, the quinolone ring
may be subdivided into 4 subclasses (cinnolines, naphthyridines, pyridopyrimidines,
and quinolines) related to the position of the nitrogen atoms within the molecule. Of
these, the quinoline subclass has by far the largest number of quinolones that have
been introduced into clinical/veterinary practice. Furthermore, other structurally related
molecules, such as 2-pyridones, quinazoline-2,4-diones, and isothiazoloquinolones, also
have antibacterial activity. Of these, 2-pyridones have probably been the most exten-
sively studied. Although usually classified as a different antibiotic family, the
2-pyridones might be structurally considered a fifth quinolone subclass. In fact, they
differ from quinolones only in the position of the N atom, which is located at the ring
juncture, and the subsequent loss of a double bond (50) (Table 1 and Fig. 3). Never-
theless, to date, no 2-pyridone has been introduced into clinical practice.
Utility of quinolones. Quinolones have been used to treat a great variety of
bacterial infections by either Gram-positive or Gram-negative microorganisms, includ-
ing intracellular pathogens (37, 51–56). Quinolones have also been used or proposed
for prophylactic treatment of specific at-risk populations, such as patients with cirrhosis
or neoplasms, and in posttransplant or presurgery/postsurgery patients (57–62). None-
theless, in several cases, the benefits have been controversial or unsatisfactory, and the
addition of new factors, such as increasing levels of antibiotic resistance, has led to the
restriction or avoidance of some of these uses (60, 63–65).
The utility of quinolones has also been explored in the treatment of parasitic
infections such as malaria, toxoplasmosis, and leishmaniosis; fungal infections (e.g.,
Candida albicans or Aspergillus fumigatus); or viral infections such as those by BK
polyomavirus, rhinovirus, and hepatitis C virus (HCV) (66–75). Meanwhile, the structur-
ally related quinolone molecules elvitegravir and ivacaftor are currently in use as an
integrase inhibitor in HIV therapy and in the treatment of cystic fibrosis, respectively
(76–78). Moreover, in addition to the antineoplastic potential of some established
quinolones such as gemifloxacin (79), new quinolone derivatives are being explored as
specific antineoplastic agents (71, 80).
Beyond human uses, fluoroquinolones have also been included in the veterinary
armamentarium, in livestock as a growth promoter (an application which is currently
forbidden in different countries, including those of the European Union) (81–83), and as a
prophylactic agent (84, 85) or in the treatment of infections (81, 85). Indeed, at the end of
the 20th century, quinolones ranked among the antibiotics most widely used worldwide,
with ciprofloxacin being considered the antibacterial agent most frequently used (81).
Adverse events. To our knowledge, adverse events were described in early clinical
trials of nalidixic acid (20, 21), and the first warning as to the possible occurrence of
adverse events related to the use of quinolones was reported as far back as 1965 (4).
FIG 1 The chronology of the quinolones (1949 to the present). The top side of the temporal line indicates the years of discovery/synthesis of a
series of representative quinolones. In all cases, the year of the most ancient report found in the literature is reported. Note that although several
thousand quinolones have been synthetized and their anti-infective potential has been explored, only a selection is presented. Furthermore, no
quinolone without a “specific” name has been included (because in the vast majority of cases, their development was discontinued), leading to
a lower number of quinolones from ⬃2010 onward, as most are in the first stages of development (for instance, DS-8587 or KPI-10 [both of which
are described in articles from 2013 {452, 453}]). The bottom side of the temporal line indicates a series of milestones in the history of quinolones.
1, fluoroquinolone-oxazolidone hybrid molecule (43).
FIG 2 Quinolones in use in human therapeutics in the United States and the European Union. In gray are
quinolones currently (as of March 2019) used in human health. In orange are quinolones that have been
discontinued (marked only when this information has been found). In yellow are quinolones proposed
for withdrawal from use on March 2019 (https://www.ema.europa.eu/en/documents/referral/quinolone
-fluoroquinolone-article-31-referral-annex-i_en.pdf). Note that discontinuation may be related to adverse
events (e.g., trovafloxacin) or to economical and market reasons. Some of these antibiotics (or other
quinolones that have not been approved or are in the investigational phase) may be considered in
special circumstances as last-resort treatment (454). Note that in all the cases, the data listed refer only
to the United States and the European Union (including the United Kingdom at the time of writing). The
introduction or current or past use/nonuse of these or other quinolones in other geographical areas may
not be inferred by this figure. a, extracted from https://www.accessdata.fda.gov/scripts/cder/ob/index
.cfm; b, in use in at least one European Union member country (including the United Kingdom at the time
of writing) (https://www.ema.europa.eu/documents/referral/quinolone-fluoroquinolone-article-31
-referral-annex-i_en.pdf). G, generation; Desf, desfluoroquinolone.
Since then, several adverse effects related to the use of quinolones have been de-
scribed, including blood disorders, central nervous system events (dizziness, sleep
disorders, and seizures, among others), gastrointestinal disturbances and C. difficile-
associated diarrhea, myasthenia gravis exacerbations, peripheral neuropathy, photo-
toxicity, rashes, and torsade de pointes, among others (25, 45, 86, 87). Of note, adverse
events are related to specific quinolone substituents. Thus, halogen atoms at position
8, such as a chloro atom in clinafloxacin or sitafloxacin or a fluor atom in sparfloxacin,
have been involved in phototoxicity reactions (45, 88–90). Interestingly, several adverse
events have been related to ethnic background (89). In this sense, ethnic differences in
phototoxicity reactions have led to the introduction of sitafloxacin into human clinical
practice in several Asian countries, such as Japan (https://www.pmda.go.jp/files/
000152974.pdf) and Thailand (91), while remaining absent from the antibiotic arma-
mentariums of the European Union, the United States, and other countries.
Cinnolines Cinoxacin
2-Pyridonesg
aV, frequent use (past or present) for animal health (livestock and/or companion animals). Note that the list of quinolones used in veterinary health include
quinolones with extended use for human health, such as ciprofloxacin, norfloxacin, and ofloxacin, some of which are included in the list of essential medicines
(http://www.who.int/medicines/publications/essentialmedicines/20th_EML2017_FINAL_amendedAug2017.pdf?ua⫽1). Most of the listed quinolones have not been in-
troduced into either human or veterinary medicine. Note that some quinolones may be found in the literature under different names. For instance, avarofloxacin,
benofloxacin, caderofloxacin, enoxacin, nadifloxacin, pirfloxacin, rosoxacin, and tioxic acid may also be found as acorafloxacin, vebufloxacin, cadrofloxacin, enofloxa-
cin, jinofloxacin, irloxacin, acrosoxacin, and tioxacin, respectively.
bNote that in some classifications, several quinolones may be considered to belong to another generation. The criteria followed in the present scheme are as follows.
First generation indicates quinolones presenting activity against some Gram-negative microorganisms (e.g., not against P. aeruginosa), almost all of which are
nonfluorinated and with a limited spectrum of clinical indications. Second generation indicates quinolones with an expanded spectrum of activity (includes most
Gram-negative microorganisms, e.g., P. aeruginosa, and some Gram-positive organisms), with expanded indications (which may include systemic infections), and
marked with the stable introduction of a fluor atom in position 6. Third generation is similar to second generation, with expanded Gram-positive coverage; members of this
class may possess activity against some atypical pathogens. Fourth generation is similar to third generation but with activity against anaerobic microorganisms.
Desfluoroquinolones are new quinolones in which the fluor atom in position 6 has been removed, usually not included in the fourth-generation scheme.
cCarries a fluor atom in position 6.
dMetioxate has almost the same structure as tioxic acid but with COOH in position 3 modified by the presence of 4-methylpiperidine (https://pubchem.ncbi.nlm.nih
fThose with the prefix “levo” (i.e., levofloxacin and levonadifloxacin) are isomers of previously described quinolones which exhibit enhanced activity.
gAt present, no 2-pyridone has been introduced as an antibacterial agent in clinical practice. Examples of this antibacterial agent group are ABT-719 (50) and KRQ-
10018 (459). The 2-pyridone ABT-719 should not be confused with modimelanotide, which also receives the same ABT-719 code. Modimelanotide is an unrelated
molecule developed later and designed to prevent acute kidney injury (460).
The most commonly known adverse events are considered to be antibiotic class related
and include arthralgias, cartilage affectations, tendinitis, and tendon ruptures. Due to
known teratogenic and mutagenic effects, the use of quinolones has classically been
avoided in pregnant women and children (88, 92). It is of note that different reports have
reanalyzed the use of quinolones in children, including neonates, and they have been
considered to be safe and beneficial in specific circumstances, such as severe or life-
threating infections by quinolone-susceptible microorganisms; furthermore, secondary ef-
fects were considered reversible (88, 93–96). In this line, several fluoroquinolones have been
introduced in established pediatric antibacterial armamentariums, such as tosufloxacin in
Japan for treating respiratory infections (97, 98). Meanwhile, a recent meta-analysis and
systematic review on the use of quinolones in pregnant women highlighted the safety of
using fluoroquinolones during the first trimester of pregnancy (99).
FIG 3 General structure and subclasses of quinolones. Quinolones have a bicyclic structure. At present,
4 quinolone subclasses, differing in the positions and numbers of the nitrogen atoms present in the basal
bicyclic structure, have been developed and introduced into clinical and/or veterinary settings (A to D).
In these 4 quinolone subclasses, the atom numeration is usually described using the quinoline subclass
as a general model (A). Position 1 is considered the N atom, and the subsequent positions are numbered
anticlockwise. Note that no atoms present in the ring junctions are numbered. Fluoroquinolones present
a fluoridine as a substituent in position 6. The radicals present in positions 1 and 7 are critical for
quinolone-target interactions. Thus, it has been proposed that radical 1 interacts with amino acid 83 of
GyrA (numeration of E. coli) or its equivalent in ParC by means of Van der Waals forces, while radical 7
interacts with amino acid 87 by charge attraction (455). (A) Quinoline. This molecule presents a nitrogen
atom only in position 1 (e.g., oxolinic acid, ciprofloxacin, and norfloxacin). (B) Cinnoline. This molecule
presents nitrogen atoms in positions 1 and 2 (e.g., cinoxacin). (C) Pyridopyrimidine. The molecule
presents nitrogen atoms in positions 1, 6, and 8 (e.g., pipemidic acid). (D) Naphthyridine. This molecule
presents nitrogen atoms in positions 1 and 8 (e.g., nalidixic acid and trovafloxacin). (E) 2-Pyridone. This
molecule is usually not considered a member of the quinolone antibiotic family. The atom numeration
of this molecule differs from those in panels A to D (the nitrogen atom in the upper ring juncture is
numbered as atom 5).
There have been recent warnings regarding the possible development of aortic
aneurysms and dissection in patients at risk as antibiotic-class-related severe adverse
events (100, 101). In this line, mice challenged with a high-fat diet and a low-dose
angiotensin infusion exposed to ciprofloxacin were more prone to developing aortic
destruction and aneurysms (102). Other unexpected severe adverse events are shown
in Table 2.
Immediate quinolone-induced hypersensitivity, mediated by quinolone-specific IgE
(103), has been on the rise in the last years, with quinolones currently likely ranking
second to -lactams as the antimicrobial agents most frequently involved in allergic
reactions (104), with an especially high incidence among patients treated with moxi-
floxacin (105–107). The severity of these reactions ranges from anaphylaxis and urticaria
to life-threatening anaphylactic shock (87, 105), with a few fatal cases being reported
in the literature (108).
resulted in patient death (45, 87, 88, 90, 101, 461). In several cases, these findings have led to the
discontinuation of research, withdrawal from clinical practice, and strong restrictions to specific nonsystemic
applications (such as topical or ophthalmic), to serious life- or limb-threatening infections, or to last-resort
applications, such as compassionate use of several of these quinolones (45, 100, 454, 462, 463). Note that
these regulations may differ among different countries. In addition, ethnic background may play a role in
the frequency of these adverse events (89). For information about systemic quinolones in use in the
European Union and the United States, see Fig. 2.
cMost relevant quinolones involved in the specific adverse event. Note that other quinolones may also be
101).
are deposited. However, this database may have misidentified or erroneous sequences,
which compromises the utility of the tool (209).
Regarding TMQR, the clearest example of misidentification is related to the expo-
nential growth which qnr scientific literature has undergone in the last years. This
phenomenon has favored the publication of partial qnr sequences to which allele
numeration has been “assigned” or, more seriously, full qnr sequences which have been
erroneously assigned to either an allele or a qnr family (209, 210). The possibility of
posting sequences in GenBank without validation (see “Qnr Classification,” below), in
order to be verified and correctly named and numbered, contributes to the perpetu-
ation and amplification of nomenclature errors because they are included in the most
relevant gene database worldwide (209, 210). Indeed, upon analyzing 1,657 Qnr
sequences recorded in GenBank (209), it was observed that 340 (20.5%) sequences
presented a major error. These errors included 105 Qnr sequences introduced in
GenBank as “PipB2,” a type III effector protein which has been associated with the
formation of vesicles (211); 145 sequences classified within an erroneous Qnr family; 16
sequences with an erroneous allele assignation; 24 partial sequences with an allele
assignation; and 50 sequences with a nonnormative initial ATG codon assigned (209).
Moreover, 449 (27.1%) of the sequences were only partially or not identified, and 9
unreported transferable alleles were detected, which were later allele numbered within
GenBank records (209). Another finding is the nondetection of qnr genes when
sequences are submitted to GenBank, leading to “hidden” qnr genes which can be
detected only after a direct DNA BLAST search (210). This may lead to unnoticed
mistakes in the discussion of results obtained in field studies.
Although this problem is less frequent because of the lower number of variants, it
also affects the remaining TMQR. Thus, different qepA variants are recorded in GenBank
as “qac” variants, while a series of GenBank-recorded qepA variants remain undescribed
(see “QepA,” below) (179). Regarding OqxAB, it is of note that a series of allelic variants
of OqxA and OqxB, which are either transferable or indigenous to K. pneumoniae, have
received further numeration; for instance, “OqxB20” and “OqxB29” were first reported
in the same article (212).
In an effort to minimize these problems, GenBank has developed “RefSeq” to name
proteins as consistently and as correctly as possible (for instance, a search for qnrA
alleles may be performed at https://www.ncbi.nlm.nih.gov/pathogens/isolates#/ref-
gene/gene_family:qnrA; for other determinants, all that is needed is a change from
“qnrA” to the desired gene). Nonetheless, it is the responsibility of all researchers to
facilitate this effort, providing the most correct and normative nomenclature, submit-
ting newly described sequences to the respective repositories, when available, in order
to be numbered in a rational manner, and providing these data in GenBank submis-
sions, manuscripts, and presentations.
The most serious problem with AAC(6=)Ib-cr is the assertion of new allelic variants
in the development of quinolone resistance. Since a single amino acid change may alter
the functionality of an antibiotic-modifying enzyme by amplifying, limiting, or modi-
fying its spectrum of activity, it is important to determine the effect of any new allele
on different quinolones prior to asserting its role as a quinolone resistance determinant.
with the resistance breakpoint being surpassed in the presence of a single target
mutation (37, 217–219). Thus, the bacterial phenotype of resistance to nalidixic acid and
susceptibility (or diminished susceptibility) to fluoroquinolones has frequently been
described and associated with the presence of at least one target mutation (37, 115,
118, 119, 217, 218), being considered a risk factor for the development of full resistance
to fluoroquinolones (219, 220). These findings have led to the modification of several
CLSI quinolone resistance breakpoints in 2016 (221, 222). In fact, in several reports, the
use of nalidixic acid was suggested as a predictor of fluoroquinolone resistance (219,
223). Along this line, natural resistance to nalidixic acid and at least diminished
susceptibility to fluoroquinolones related to the presence of a specific wild-type GyrA
amino acid in position 83 and/or position 87 (E. coli numeration) have been described
in microorganisms such as Bartonella spp. and Brevundimonas spp., among others
(224–226).
The eruption of TMQR has altered this scenario, increasing the isolation of
quinolone-resistant microorganisms in the absence of target mutations and descrip-
tions of microorganisms exhibiting the unusual phenotype of nalidixic acid suscepti-
bility and ciprofloxacin resistance. Thus, the presence of more than one TMQR may
increase the final MIC for resistance breakpoints even in the absence of quinolone
target mutations (153, 227–231, 471). Furthermore, this finding has also been reported
in isolates in which only one TMQR was identified or introduced (15, 228, 231–233). For
instance, a recent swine isolate of Salmonella enterica serovar Rissen, without target
mutations and in which the only TMQR detected was QnrVC4, showed MICs of nalidixic
acid, norfloxacin, and ciprofloxacin of 32 g/ml, 1 g/ml, and 0.5 g/ml, respectively
(231). In this line, it has been shown that the levels of quinolone resistance produced
by QnrB and QnrS are directly related to their levels of expression. Thus, it was observed
that the final MICs of ciprofloxacin were increased when QnrB1, QnrS1, or derived
mutants with impaired functionality were cloned in expression vectors under IPTG
(isopropyl--D-thiogalactopyranoside) induction (234, 235). Furthermore, a study by
Garoff et al. (236) in which qnrB and qnrS (no allele was specified) were cloned alone
into E. coli (strain MG1655) and expressed under the control of different promoters
showed that the MIC of ciprofloxacin increased according to gene expression levels
until reaching plateaus of 0.375 and 1 g/ml for qnrB and qnrS, respectively, repre-
senting increases of 25 and 66.6 times the original MIC for MG1655 (0.015 g/ml) (Fig.
4). Although in this study, qnrB and qnrS were cloned as a single resistance determinant,
it is important to again highlight that the final MICs are the result of multiple
phenomena, therefore also being related to intrinsic bacterial factors such as cell wall
permeability or intrinsic efflux pump activity. In microorganisms such as Acinetobacter
spp. or P. aeruginosa, low membrane permeability and the extrusion of quinolones by
several powerful efflux pumps strongly affect the intrinsic MICs of quinolones. Thus,
intrinsic ciprofloxacin resistance ranges from 0.125 to 1 g/ml (⬎8 times that of E. coli
MG1655) to 0.25 to 4 g/ml (⬎16 times that of E. coli MG1655) in the cases of A.
baumannii and P. aeruginosa, respectively (37, 237–239). Thus, it could be predicted
that similar qnrB or qnrS expression levels in these microorganisms lead to the detec-
tion of higher final MICs.
Prior to the boom of TMQR, another atypical scenario was the presence of the
unusual phenotype of nalidixic acid susceptibility and ciprofloxacin (fluoroquinolone)
resistance or diminished susceptibility. Previously, this had been reported only in
specific microorganisms such as the above-mentioned S. maltophilia, in which this
phenotype is especially frequent, probably due to the unusual mechanisms involving
the development of quinolone resistance (214, 216). In addition, this phenotype is also
shown in a few E. coli clinical isolates, with decreased quinolone uptake associated with
the highly unusual GyrA mutation D82G (240, 241) and in in vitro-constructed E. coli
mutants carrying the GyrA substitution G81D (242). This has also been observed in
Campylobacter jejuni and Neisseria gonorrhoeae, for which no analysis of the mecha-
nisms of resistance has been reported (117, 243), or in C. jejuni presenting the GyrA
substitution T86A, thereby suggesting the presence of an unidentified factor (244).
FIG 4 Effect of expression levels of qnrS and qnrB on final MICs. In both panels, the mRNA levels are
relative to those of the control genes hcaT, idnT, and cysG and are expressed in log10 units (based on data
from reference 236). (A) qnrS; (B) qnrB.
p19051-IMP HI5 316.8 aac(6=)Ib-cr,l qnrB52, qnrS1 aacC2, aadA2, armA, strA, strB/(blaTEM-1B/blaCTX-M-3, blaSHV-12/blaIMP-4/—)/—/—/fosA3/mph(E), msr(E)/arr3/sul1/—/dfrA12/tmrBm 247
pUM505 I 123 crpP —/(—/—/—/—)/—/—/—/—/—/—/—/—/— 154
pHNSHP45-2 H12 251.5 oqxAB aac(3)-IV, aadA1, aadA2, aph(3=)-Ia/(—/blaCTX-M-14/—/—)/cmlA, floR/mcr-1/fosA3/—/—/sul1, sul2, sul3/—/dfrA12/— 278
pOLA52 X1 52 oqxAB —/(blaTEM-1/—/—/—)/—/—/—/—/—/—/—/—/— 200
pHPA FII 70 qepA1 rmtB/(blaTEM-1/blaCTX-M-12/—/—)/—/—/—/mph(A)/—/—/—/—/— 158
pIP1206 FI 168 qepA1 aadA4, rmtB/(blaTEM-1/—/—/—)/catA1/—/—/—/—/—/tet(A)/dfrA17/— 286
pMG252n 185.6 qnrA1 aadA2, aadB, aph(3)II/(blaCARB-2/—/—/blaFOX-5)/catB3, catB11, cmlA1, mdtL/—/—/mph(E), msr(E)/—/sul1/—/—/dfrA19 15, 251, 284, 317
SGI-1Vo 42.9 qnrA1 aac(6=)Ib, aadB/(—/blaVEB-6/—/—)/—/—/—/—/—/sul1/tet(A)⌬/dfrA1/— 277
pJIBE401p L/M ⬎150 qnrB2 aac(6=)Ib4/(—/—/blaIMP-4, blaOXA-73/—)/catB3c/—/—/mph(A)/—/sul1/—/—/— 279
pENVA H 253 qnrB4 aacC2, aadA/(blaTEM-1/blaCTX-M-15/—/blaDHA-1)/—/—/—/—/—/sul1/tet(A)/dfrA15/— 276
pECY6-7 ColE 2.7 qnrB19 —/(—/—/—/—)/—/—/—/—/—/—/—/—/— 466
pLRM24 80 qnrB19 aac(6=)Ib, aadA1/(blaTEM-1/—/blaKPC-3/—)/—/—/—/—/—/—/—/—/— 283
Chromq qnrB62 aac(6=)II/(—/—/blaVIM-2/—)/—/—/—/—/—/sul1/—/— 275
pHS10 120 qnrC —/(—/—/—/—)/—/—/—/—/—/—/—/—/— 340
p2007057 4.27 qnrD1 —/(—/—/—/—)/—/—/—/—/—/—/—/—/— 344
pKP41M M1 70 qnrE1 aac(6=)-Ib, aadA1/(blaTEM-1, blaOXA-9/blaCTX-M-8/—/—)/—/—/—/—/—/—/—/—/— 273
pGN26-KPC X6 46.3 qnrS1 —/(blaTEM-1⌬/—/blaKPC-2/—)/—/—/—/—/—/—/—/—/— 280
pK245 98 qnrS1 aacC2, strA, strB/(blaLAP-2/blaSHV-2/—/—)/cat2/—/—/—/—/—/tet(D)/dfrA14/— 272
(Continued on next page)
cmr.asm.org 15
Clinical Microbiology Reviews
TABLE 4 (Continued) Ruiz
(e.g., TMQR) may be only a modest increase in MICs, not reaching the established resistance breakpoint, or gene expression may be downregulated or impaired. Furthermore, in several cases, such as specific TMQR,
aminoglycoside-modifying enzymes, or transferable mechanisms of macrolide resistance, the resistance conferred by the reported mechanisms may not be extended to all antibiotic class family members. The number of described
genetic structures carrying TMQR alone or in association with other antibiotic resistance genes is enormous and growing on an almost daily basis. Only a series of representative genetic structures are described here. When a TMQR
is reported to be present within a plasmid but no exact plasmid name is provided, the name of the bacterial strain is used as the plasmid name, and the genetic structure name is highlighted in boldface type.
cIn several cases, the whole sequence or full content data of the reported genetic structure are not available, with the subsequent possible presence of unnoticed antibiotic resistance genes; for instance, although not
reported when fully sequenced (GenBank accession number MK638972), in the plasmid pMG252, which started the TMQR era, 2 other -lactamases with isoelectric points of 7.0 and 7.6 were detected, in addition to -
lactamases with isoelectric points of 5.6 and 7.2, and further identified as blaCARB-2 and blaFOX-5 and reported in the table (15).
daac(6=)Ib-cr is harbored within an ⬃4.8-kb integron {In1021 [aac(6=)Ib-cr-arr3-dfrA27-aadA16-IS15-sul1Δ-IS1]}.
OXA-1-catB3-arr3 qacE⌬1-sul1-ISCR1-qnrA1-ampR-qacE⌬1-sul1]}.
fBoth TMQR are harbored within an ⬃15-kb complex class 1 integron {In37 [aac(6=)Ib-cr-bla
gThe qnrA3 gene is located after a structure (qacE⌬1-sul1-ISCR1-qnrA3) suggesting the presence of an undescribed complex class 1 integron, while aac(6=)Ib-cr is placed upstream of qnrA3 just after the location of IS26 and
jThe aac(6=)Ib-cr and qnrB6 genes are harbored within a complex integron {In1229-like [aac(6=)Ib-cr-arr3-dfrA27-qacE⌬1-sul1-ISCR1-ORF (putative oxidoreductase)-qnrB6-pspF⌬-qacE⌬1-sul1]}. This integron is present twice in
the plasmid. An additional qnrB6 gene within a structure resembling that of the end of the complex integron is also present.
kIntegron identical to In37 but carrying a qnrB10 allele instead of qnrA1.
mTmrB confers resistance to tunicamycin, an antibiotic produced by members of the genus Streptomyces (e.g., Streptomyces lysosuperificus) which targets the cell wall synthesis of Gram-positive bacteria. Toxicity for
pThe TMQR is present within a complex class 1 integron environment {In585-like [bla
IMP-4-qacG2-aac(6=)Ib4-catB3-qacE⌬1-ISCR1-sapA-orf2-qnrB2-pspF-qacE⌬1-sul1]}.
qThe TMQR is present within a complex class 1 integron environment {In1184 [bla
VIM-2-aac(6=)II-gucD-qacE⌬1-sul1-ISCR1-pspF-qnrB62-qacE⌬1-sul1]}.
rThe qnrVC1 gene is harbored in a class 1 integron (In1214 [arr3-qnrVC1-bla
OXA-10-aadA1e-qacE⌬1-sul1]).
cmr.asm.org 16
Clinical Microbiology Reviews
Transferable Mechanisms of Quinolone Resistance Clinical Microbiology Reviews
Brahmi et al. and Moremi et al., did not find this association (291, 292), probably
suggesting the presence of local differences in the spread of microorganisms/plasmids
related to factors such as specific antimicrobial pressure and geographical factors.
TMQR are frequently encoded within plasmids belonging to different incompatibil-
ity groups and with heterogeneous sizes (Table 4), although surrounding structures are
often similar, suggesting the presence of a limited number of mobilizations of ancestral
chromosomal origin followed by a series of transpositions, recombinations, deletions,
insertions, and every other genetic material arrangement leading to the currently
observed variety. In this sense, the boom of next-generation sequencing approaches
has increased the availability of data related to the presence and distribution of TMQR.
Further molecular epidemiological data are presented in the specific sections devoted
to each TMQR.
Qnr
At the beginning of 1998, Martínez-Martínez and colleagues (15) described the
presence of a TMQR within the plasmid pMG252 in a K. pneumoniae strain isolated in
Alabama in 1994, which was named “qnr” (see “Nomenclature of the Transferable
Mechanisms of Quinolone Resistance,” above). The transfer of this plasmid led to
increases in the quinolone MICs of 8- to 64-fold irrespective of the initial MICs. The
effect was dissimilar among the different quinolones tested, being maximum with
nalidixic acid and minimum with clinafloxacin (15). Thus, when the plasmid was
transferred to E. coli J53, the nalidixic acid susceptibility changed from susceptible
(4 g/ml) to resistant (32 g/ml). Moreover, the authors showed that microorganisms
carrying the qnr determinant were more prone to developing full resistance to fluoro-
quinolones (15). Subsequently, different studies were designed to determine the true
prevalence and relevance of this mechanism of resistance, demonstrating its rare and
low prevalence. Thus, this mechanism was found in only 6 (1 E. coli, 4 K. pneumoniae,
and 1 Klebsiella species isolates) out of 420 Gram-negative microorganisms tested (338
clinical isolates from 19 countries and a series of laboratory strains carrying different
plasmids), with all 6 having been collected in 1994 in Alabama (13). In the subsequent
years, this qnr determinant as well as new qnr variants (Table 5) were increasingly
detected worldwide in different microorganisms, mainly Enterobacteriaceae (138, 293–
297) and a few isolates of Aeromonadaceae (297–300), Moraxellaceae (301) Pseudomon-
adaceae (206, 302), and Vibrionaceae (175, 303, 304), among others (Fig. 5).
Thereafter, some studies were designed using older bacterial collections in order to
detect the presence of these genes in isolates recovered prior to 1994. Thus, the older
transferable qnr gene detected belonged to the qnrB family, being identified in one K.
pneumoniae strain isolated in 1988 in Cordoba, Argentina (305). In the same study,
another qnrB gene carried by a Citrobacter freundii isolate from Brooklyn, NY, was
detected, which had also been isolated in 1988 (305). Nonetheless, in the absence of
specific analysis to determine its genetic environment, and since subsequent studies
have established the possible origin of qnrB genes in the chromosome of Citrobacter
spp. (306), it is uncertain whether this gene was present within a transferable structure
or was an intrinsic resident gene (see “QnrB,” below). In fact, the presence of qnrB60 and
a qnrB pseudogene has been described in the C. freundii collection strains ATCC 6879
and ATCC 8090, respectively (307), both of which were isolated in the late 1920s or early
1930s (308, 309).
It was observed that Qnr also confers slight protection against 2-pyridones,
quinazoline-2,4-diones (both of which are structurally closely related to quinolones),
and spiropyrimidinetriones but has no protective effect against other topoisomerase
type II-targeting molecules such as aminocoumarins (coumermycin A1, novobiocin, and
simocyclinone D8), gyramide A, microcin B17, pyrazolopyridones, or tricyclic pyrimi-
doindoles (310).
Although the presence of Gram-positive chromosomally encoded Qnr has been
described (311), all the currently known transferable Qnr families derive from Gram-
negative ancestors. Therefore, the presence of qnr-related genes has been described in
cOnly confirmed TMQR. When a TMQR was related to the presence of a specific plasmid but no gene-specific nomenclature is available, the name of the plasmid is
indicated.
dBased on GenBank and bibliographic searches. Regarding Qnr, only those included in the Lahey database (formerly at http://www.lahey.org/qnrStudies/) as of 31
December 2018 are shown. This database is no longer available. Other unnamed or erroneously assigned alleles may be found in GenBank (209).
eBased on a RefSeq search (https://www.ncbi.nlm.nih.gov/pathogens/isolates#/refgene/gene_family:XXX, where XXX is the name of the gene) (updated on 12 April
2019).
fPublication of the first allele of the family that has been considered a TMQR, irrespective of the time at which the ability to confer quinolone resistance was
demonstrated. Note that previous conference presentations may have been made and that these presentations may be included in reviews reported prior to the
reported data.
gY indicates that all or several alleles were detected within the integron environment.
iWhile considered at the Lahey website, QnrA8 is not included in RefSeq. Of note, the only description of this gene was in the S. algae chromosome; therefore,
qnrStudies/], and no data on the exact sequence are provided at either the Lahey website or GenBank).
kQnrS3 has not been included in RefSeq. Of note, the reported sequence lacks the initial amino acid (356).
lWhile QnrVC8 and QnrVC9 were considered at the Lahey website, they are not included in RefSeq. It is of note that in the only description of these genes, they were
located within Vibrio species chromosomes; therefore, transferability has not been demonstrated (363).
mThe proposed QepA8 protein has a 2-amino-acid insertion leading to a final size of 513 amino acids.
nStandard size of OqxA and OqxB, respectively. Note that transferable OqxA and OqxB presenting amino acid insertions have been detected (see Tables 11 and 12).
pNo data about the ability of one of these alleles to extrude quinolones have been reported.
qOnly the QacBIII allele has been associated with the ability to extrude quinolones.
rThe presence of more than 37 closely related alleles with identity levels of ⬎90% has been highlighted (414). Nonetheless, the effect on ciprofloxacin has been
established for only one allele, with the others remaining to be studied.
FIG 5 Main genera in which the presence of qnr genes has been described. This is a nonexhaustive list; the possible presence of nonreported qnr genes
in genera presented in the figure or the presence of qnr genes in genera not presented in the figure should be taken into account. a, genera in which
(Continued on next page)
FIG 6 Description of new qnr alleles (1998 to 2017). Only transferable genes/alleles with a standard name in the nomenclature as of 31 December 2018
according to the Lahey website (formerly at http://www.lahey.org/qnrStudies/) are shown. The reporting year has been considered following the next-priority
order. (A) Date of oldest publication by the describing authors. Note that in several cases, the paper by the original authors might have been published several
years after the original inclusion in GenBank, and therefore, data regarding these alleles may be present in other precedent articles. For example, QnrB6 was
included in GenBank in 2006, being considered when the QnrB nomenclature was normalized and reorganized (319), but the oldest article found by the
describing authors was published in 2009 (457). (B) In the absence of publication data by the describing authors, the oldest article published by any author
was considered. (C) Presence of the allele in meeting presentations by (i) describing authors and (ii) other authors. (D) When neither the published article nor
meeting presentation was found, reporting data have been annotated by the year of the GenBank record. Note that the presence of the allele in GenBank may
precede the time of article publication. In addition, several sequences might be added to the repository at a later time. Note that the absence of an identified
citing article/communication does not preclude the absence of related publications or communications. (E) In the absence of all above-described data, personal
communication was considered if recorded at the Lahey website (e.g., from qnrB84 to qnrB87). qnrE1 was proposed as a new gene in 2017 (176), although qnrB88
was submitted to GenBank in 2016 and also first reported (a meeting presentation) in 2016 (352).
Qnr Classification
In the first years after the description of qnrA1 in 1998, literature on qnr genes was
scarce, being mostly addressed to describe the presence and prevalence of qnrA1 in
different geographical areas (13, 253, 317) or to advance the knowledge of the mode
of action of qnr (318). Nonetheless, in the mid-2000s (Fig. 6), the presence of new qnr
genes/alleles in GenBank and published reports on qnr suddenly increased. This led to
the presence of different qnr genes with the same name and the subsequent increasing
chaos (319). Therefore, in 2008, a series of rules was implemented to unify the criteria
to define a new qnr allele or gene (319) (Table 6). Thus, the Qnr proteins were classified
within families (genes) and subdivided into alleles based on single or multiple amino
acid differences among them (319).
fTransferable Qnr genes are named “Qnr” followed by a letter (e.g., QnrA). Chromosomal Qnr should be named with the initials of the microorganism followed by
“Qnr” (e.g., accordingly, qnr from Photobacterium profundum should be named Ppqnr for the DNA and PpQnr for the protein). In addition, it was proposed that if a
chromosomal Qnr has at least 70% identity to one of the established transferable qnr families, it may be named according to transferable or chromosomal name
rules (e.g., SaQnrA3, where Sa represents Shewanella algae). Usually, the latter consideration is applied only for proposed original bacterial sources of established
transferable qnr families.
gThe qnr nomenclature rules published in 2008 (319) considered the presence of either a DNA or amino acid identity difference of ⱖ30% as a requisite to define a
new gene. While this has been considered a general rule, in 2017 qnrB88 was renamed qnrE1 because its original source (Enterobacter instead of Citrobacter) was
taken into account (176), classifying a new gene irrespective of having an amino acid identity of ⬃85% with established QnrB alleles, including QnrB1.
Silent mutations are not considered to determine the presence of new qnr alleles
(319). Nonetheless, the presence of silent mutations may be of epidemiological and
evolutive interest because they may play a role in the diversification of qnr, since further
codon alterations may result in new variants. In this sense, it is of note that in vitro
studies have shown that amino acid changes may alter the levels of resistance con-
ferred by different Qnr proteins (see “In vitro mutations,” below). Furthermore, the
effect of silent differences in DNA sequences in qnr genes on mRNA stability or on the
efficiency of translation to the final protein related to codon usage or other phenom-
ena, which may lead to different levels of Qnr proteins in the bacterial cytoplasm, with
a subsequent effect on final MICs, remains unexplored. In this sense, it has been
observed that increased levels of expression of qnrB and qnrS may be related to higher
MICs of ciprofloxacin (236) (Fig. 4).
Identity criteria (⬍70% identity) were established to determine the different Qnr
families. Accordingly, at present, 7 families of transferable qnr genes have been
described (qnrA, qnrB, qnrC, qnrD, qnrE, qnrS, and qnrVC) (see “QnrE,” below, for levels
of identity between QnrE and QnrB). Nonetheless, while a series of closely clustered qnr
alleles can be found and can even be easily classified within a common family, a series
of gradually divergent alleles, which act as a bridge between different clusters, may
obscure the Qnr family borders. These divergent alleles are the result of natural qnr
sequence divergences within the ancestral host, followed by more than one indepen-
dent mobilization phenomenon, the natural evolution of transferable qnr genes within
different hosts, and the description of transferable qnr alleles/genes with a different,
but closely phylogenetically related, original source (e.g., QnrB and QnrE). All these
forces are not exclusive, and indeed, all act in a simultaneous manner.
A repository website used to number and order new qnr families and alleles (the
Lahey database, formerly at https://www.lahey.org/qnrStudies/) was developed, includ-
ing more than 100 different qnr alleles (Table 5). Similar to what occurred on July
2015 with the repository of -lactamases (https://www.lahey.org/Studies/), in 2018,
this repository was transferred to the NCBI for Qnr nomenclature maintenance
and future gene/allele assignations, and the original Lahey database is no longer
online. Thus, -lactamase, qnr, as well as mcr gene/allele assignations are cur-
Qnr Families
Although the presence of Gram-positive chromosomally encoded Qnr proteins has
been described (311, 321), all the currently known transferable Qnr families derive from
Gram-negative ancestors (see from “QnrA” to “Chromosomal Qnr,” below).
As mentioned above (see “Nomenclature of the Transferable Mechanisms of Quin-
olone Resistance”), the names of transferable Qnr determinants have been assigned in
a mixed manner. Thus, some follow an alphabetical order according to the first
description, while others have been named based on the microorganisms from which
they were first recovered, irrespective of the ordinal moment and the true prevalence
in this microorganism. The next 7 subsections are strictly ordered alphabetically.
Therefore, the order in which the different families are presented does not preclude the
order described in time.
QnrA. As mentioned above, the qnrA gene, which encodes a 218-amino-acid
protein, was the first well-established TMQR (15). Further studies determined that this
qnr gene is located in an integron-like environment (318). The integron containing
qnrA1 was fully sequenced in 2007, confirming the presence of qnrA1 in a complex class
1 integron downstream from the first qacEΔ1-sul1, between 2 ISCR1 elements (284). The
same study also highlighted the presence of qnrA1 in other complex integrons (284).
The association between ISCR1 and qnrA1 has been largely reported, although in most
cases, only the first ISCR1 is present (253). Furthermore, other genetic structures lacking
the presence of ISCR1 have also been described (322). The presence of qnrA between
2 ISCR1 elements may facilitate spontaneous (or induced under quinolone pressure)
qnrA gene duplication, with the subsequent effect on the final MIC of quinolones (251).
Thus, when the in vitro-selected pMG252 plasmid-derived mutant pMG252A was
cloned into an E. coli J53 background, the qnrA1 expression level and the MIC of
ciprofloxacin rose 2.2- and 8-fold, respectively, higher than those related to the
presence of pMG252 (258). Recent reanalysis and sequencing of pMG252A showed that
these findings were due to the presence of 4 additional qnrA1 copies after the original
ISCR1-qnrA-qacE⌬1-sul1-ISCR1, following the scheme qnrA-qacE⌬1-sul1-ISCR1 (251). Of
note, this phenomenon might be extended to other TMQR placed between 2 insertion
sequences. For reading of literature published before 2006, note that ISCR1 is usually
referred to as orf513 or orf341. Additionally, the term “CR” for “common region” may
also be present (196, 323–325).
Although variants of qnrA1 containing silent mutations were detected in 2003 (253),
until 2004, no other qnrA allele (qnrA2) was described (GenBank accession number
AY675584). Nevertheless, to my knowledge, there has been no published report by
sequence authors, with the first report including QnrA2 data, together with those of
QnrA3, QnrA4, and QnrA5, being published in 2005 (172).
Currently, 8 qnrA alleles have been described in accordance with nomenclature
rules, mainly described within complex integron-like genetic environments (246, 284,
325–328), while a search of GenBank detected other new transferable QnrA alleles
together with 2 new chromosomal Shewanella algae qnr variants (see the next para-
graph). Of note, QnrA8 recorded previously in Lahey’s repository is not in the RefSeq
list, probably because no description within a plasmid or other transferable genetic
structure has been made.
Shewanella species has been proposed as the chromosomal ancestor of this Qnr
family. Thus, QnrA1 (329) and QnrA2 (GenBank accession number BAF95541) have been
detected in the chromosome of Shewanella putrefaciens, while QnrA2, QnrA3, Qnr4,
FIG 7 Example of a complex integron containing qnr determinants. In both panels, the first variable region is identical. Note that in addition to the qnr
determinants, other TMQR such as aac(6=)Ib-cr may be also present. (A) Complex integron containing a typical ISCR1 element (note that in older studies, ISCR1
is referred to as orf513 or orf341) after the classical 3=-end region of class 1 integrons (qacEΔ1-sulA) and just before the qnrB6 determinant environment (between
psp and sap clusters) present in the second variable region. (Adapted from reference 458 with permission.) (B) Complex integron in which ISCR1 has been
replaced by an IS26 element. Note that the environment of qnrB4 slightly differs from that in panel A (the sap cluster has been lost). (Adapted from reference
245 with permission from Elsevier.)
QnrA5, and QnrA8 have been identified in that of S. algae (172, 330, 331). QnrA7 has
also been detected in S. algae both in the chromosome (GenBank accession number
CP018456) and within a 33-kb plasmid (332). A GenBank search using QnrA1 (GenBank
accession number AAL60061) also detected other QnrA alleles within the chromosome
of S. algae, including the sequences under GenBank accession numbers WP_044735234
(333) and WP_045283443 (334).
QnrB. The first qnrB gene was described within the pMG298 plasmid in a South
Indian K. pneumoniae isolate (335), being the third transferable qnr family described. In
the same study, the second qnrB allele (qnrB2) was detected in isolates from the United
States when designing primers to determine the prevalence of qnrB in different
microorganisms (335). Since then, the number of new qnrB alleles has been continu-
ously rising, with a total of 87 unique sequences with an assigned allele numeration in
the Lahey database as of 31 December 2018 and, as remaining qnr genes, an uncertain
number of unnoticed alleles (209). Similar to what has been described for the qnrA
genes, qnrB may be located within a complex integron environment associated with
ISCR1 or other insertion sequences like IS26 (245, 275, 336) (Fig. 7). In addition, other
dissemination pathways, such as transposons like Tn2012 (formed by ISEcp1C and
qnrB19) or the so-called KQ element (for KPC and Qnr), have been described, in which
the acquisition of qnrB19 has been suggested to be related to ISEcp1-like mobilization
(283, 336, 337).
Although in 2005, two different sizes for QnrB1 (226 amino acids long) and QnrB2
(214 amino acids long) were described (335), in 2008, consensus was achieved regard-
ing the use of the initial codon present in QnrB2 as the initial QnrB family ATG because
of its commonness to all the QnrB proteins described (319). Thus, caution is needed
when analyzing QnrB in order to ensure the use of the correct size and amino acid
numeration.
In 2004, the presence of a qnrB gene was detected during marine metagenomic
studies, leading to the proposal that an unknown marine microorganism may be the
original source of QnrB (338, 339). Nonetheless, taking into account the high prevalence
and diversity of QnrB, together with the lack of surrounding mobile elements in
Citrobacter spp., members of the C. freundii complex are considered to be the original
source of QnrB (306).
Within the genomes of the C. freundii complex (as well as in most of the transferable
QnrB proteins detected), the genetic environment of qnrB is highly conserved, being
located between two gene clusters, the psp (phage shock protein) cluster downstream
and the sap (sensitivity to antimicrobial peptides) cluster upstream (306, 307). These
clusters are contiguously present in the chromosomes of other Enterobacteriaceae,
including other Citrobacter spp. such as Citrobacter koseri and Citrobacter rodentium, in
which no qnrB is present. In view of these findings, it was proposed that a common
ancestor of the C. freundii complex had acquired an exogenous qnrB-like gene between
the psp and sap clusters, which was thereafter maintained or partially deleted, leading
QnrD is one of the few TMQR that has been described among Gram-positive
microorganisms. Thus, in a study aimed at determining the presence of antibiotic-
resistant microorganisms in treated and untreated river water, qnrD was detected in
Bacillus spp. and Kurthia spp. (167). Unfortunately, no attempt to confirm these data by
DNA sequencing and to determine either the exact genetic environment or the exact
allelic variant of qnrD was performed, and no further data are available.
QnrE. In the middle of 2017, a K. pneumoniae strain isolated in 2007 in Argentina
exhibiting low-level quinolone resistance, wild-type GyrA, and negative results when
TMQR were sought was reported. Further analysis showed the presence of a 645-bp
ORF carried within an IncM1 transferable plasmid (pKp1130; GenBank accession num-
ber KY073238) (176). This ORF showed an average identity of 75% with members of the
QnrB family, and a literature search showed 100% identity with qnrB88, which was
isolated in Brazil from a K. pneumoniae plasmid (pKp145-11b; GenBank accession
number KX118608) (352). Thereafter, another K. pneumoniae isolate carrying this gene
within an IncM1 plasmid (pKP41M) as well as 3 S. enterica isolates belonging to
serotypes Enteritidis, Infantis, and Newport also carrying qnrE1 within an IncM1 plasmid
were again described in Brazil (273, 353). A further search of GenBank showed the
presence of qnrE1 within S. Typhimurium (e.g., GenBank accession number KYE08263),
C. freundii (e.g., GenBank accession number PUU65120), and other K. pneumoniae
isolates. Moreover, a variant with only 2 amino acid differences currently recorded as
QnrE2 in GenBank (GenBank accession number WP_078207746.1) is present within an
E. coli plasmid (pEC422_1; GenBank accession number CP018961) as well as (although
initially recorded as QnrS1) a whole-genome sequence of K. pneumoniae (GenBank
accession number UJVU01000046).
Although, according to the rules of qnr gene nomenclature, this gene should be
considered a member of the qnrB family (Table 6), QnrE has been classified as the first
member of a new family (qnrE). An in-depth analysis showed high identity with the
chromosomal qnr gene of Enterobacter spp., also suggesting that ISEcp1 is responsible
for gene mobilization (176) and therefore has a different ancestral origin than other
qnrB genes. Notwithstanding, this finding highlights the need to use the concept of
gene family with caution as well as to advance toward the introduction of the concept
of original bacterial chromosomal source to nomenclature rules. Indeed, there are
numerous genomic data for different Enterobacter spp., including an Enterobacter kobei
Qnr protein (GenBank accession number OTW32454) with 100% amino acid identity
with QnrE1. In addition, a series of closely related qnr genes (even with ⬎85% identity
in either DNA base pairs or amino acids) are also present in other bacterial genomes,
especially within the genus Serratia, such as Serratia plymuthica (e.g., GenBank acces-
sion number AGP44145) and other Serratia spp. (e.g., GenBank accession number
OKP25762), as well as in the genomes of other Enterobacteriaceae such as Lelliottia spp.
(e.g., GenBank accession number ASV55492) and Buttiauxella spp. (e.g., GenBank ac-
cession number KFC78735).
Curiously, at present, all transferable qnrE genes (those present outside a potential
indigenous bacterial source) for which the geographical source is available (18 out of
20 GenBank-recorded sequences) have been described in South America, within Citro-
bacter spp. from Argentina, Klebsiella spp. from Argentina and Brazil, Salmonella spp.
from Brazil, and E. coli from Ecuador. It is likely that upon routine screening of this gene,
the number of microorganisms carrying QnrE variants, alleles described, and geograph-
ical locations would be greatly increased.
QnrS. QnrS was first described in 2006 as being encoded within a conjugative
plasmid of 47 kb (pAH0376) of an S. flexneri 2b strain isolated in Japan in 2003 (171).
Studies performed in subsequent years showed a low, albeit increasing, prevalence of
QnrS and a wide geographical distribution. Thus, QnrS was described in microorgan-
isms such as S. enterica, E. coli, and Enterobacter cloacae, among others (328, 354). Since
then, up to 9 QnrS alleles have been described and numbered in a previous Internet
repository (https://www.lahey.org/qnrStudies), and at least 5 other alleles (QnrS10 to
QnrS15) are present in GenBank (209), being described in a great variety of microor-
ganisms and environments (133, 203, 209, 245, 297, 300, 355). Of note, QnrS3 is not
numbered in RefSeq, probably because the reported sequence lacks the initial amino
acid (356).
The mobilization and spread of qnrS alleles have been mediated by structures such
as insertion sequences such as IS2, IS26, and ISEc12 (a member of the IS21 family) (272,
354, 357, 358). Additionally, qnrS has also been described as occurring near, but not
within, Tn3-like structures carrying blaTEM-1 (171, 354). In this sense, Kehrenberg et al.
described the plasmid pINF5 from Salmonella enterica serovar Infantis which, after a
Tn3-like structure, carried a defective ISEc12 element followed by a qnrS2 gene, an
internal segment of the CS12 fimbrial gene cluster of E. coli, and a defective IS26
element (354).
The origin of QnrS has been proposed to be among aquatic microorganisms.
Thus, in 2007, the presence of chromosomal Qnr was observed in Vibrio splendidus,
with identity levels of 83.1 to 83.9% with QnrS1 and 87.1 to 87.6% with QnrS2 (the
2 QnrS proteins described at that time) (312). Thereafter, Vibrio species sequences
presenting higher identity levels have been detected. For instance, 97% and 95.5 to
96% amino acid identities with chromosomal Qnr from Vibrio mytili (GenBank
accession number KIN11186.1) and Vibrio parahaemolyticus (GenBank accession num-
bers WP_029802054.1 and WP_029823919.1), respectively, have been observed. Fur-
thermore, in a V. parahaemolyticus isolate from Malaysia, a Qnr protein (GenBank
accession number KKF68274.1) has been detected, located between transposases
having 100% amino acid identity with QnrS1. Despite being classified as “genomic,” the
size of the containing DNA sequence (2,178 bp long) allows the possible presence of
this qnr gene within a plasmid environment.
QnrVC. Although qnr genes were described within the genomes of Vibrionaceae as
early as 2005 (171, 174), the presence of a new transferable qnr family within an
integron environment in V. cholerae was not shown until 2008 (175). This transferable
Qnr variant was subsequently detected in other Vibrionaceae (303, 304, 359, 360) and
in other bacterial families such as Aeromonadaceae (361) and Moraxellaceae (362), even
within plasmids or in integron environments. Despite these evidences, the presence of
this transferable qnr family was not proposed and subsequently included in the Lahey
database until 2013 (210).
Nine different QnrVC alleles (QnrVC1 and QnrVC3 to QnrVC10), which have a length
of 218 amino acids, have been classified by the Lahey website. Of these, at present,
QnrVC8 and QnrVC9 are not included as numbered QnrVC variants in RefSeq, being in
fact present within the Vibrio species chromosome (363). Note that qnrVC2 is not
considered in the list because of the presence of 3 base insertions and 1 deletion (361)
leading to a frameshift and premature stop, erasing its functionality. Similar to QnrB,
another possible initial codon may be present 13 amino acids before the initial ATG
codon (e.g., GenBank accession number APA29731).
These alleles have been described worldwide in plasmid and integron environ-
ments, either in the first variable region of a classical class 1 integron or downstream
from ISCR1 within a complex class 1 integron (274, 364). In this sense, it is worth
mentioning that qnrVC may be included in a gene cassette with an attC site (175, 231,
361). In contrast to classical integron gene cassettes, qnrVC gene cassettes may possess
their own promoter, which allows them to surpass the decreased levels of expression
related to a delayed position within the overall integron or to the absence of the strong
promoter P2 (231).
It is of interest that QnrVC may be subdivided into two main groups, which are
composed of (i) QnrVC1, QnrVC3, QnrVC6, QnrVC10, and one P. aeruginosa sequence
erroneously classified as QnrVC7 (GenBank accession number AWT08553), in which the
more divergent allele is QnrVC3, differing by 4 and 5 amino acids with respect to
QnrVC1, QnrVC6, and QnrVC10 and QnrVCAWT08553, respectively, and therefore with
identity levels of 97 to 99.5%, and (ii) QnrVC4, QnrVC5, and QnrVC7, in which the
maximum difference is 3 amino acids between QnrVC5 and QnrVC7. The identity levels
of these alleles are around 98.5 to 99.5%. QnrVC1-like and QnrVC4-like proteins differ
by a minimum of 40 amino acids, with a maximum identity of 81.6%.
At present, QnrVC has been detected in additional genera within the Enterobacte-
riaceae (E. coli, Enterobacter spp. Citrobacter spp., Klebsiella spp., and Salmonella spp.)
and Pseudomonadaceae (P. aeruginosa and P. putida), among others (206, 231, 249, 302,
350, 364–366) (Fig. 5).
Chromosomal Qnr. The presence of chromosomal qnr genes was first observed in
2005 in Photobacterium profundum and Vibrio vulnificus (171). The same year, the
chromosomal qnr gene of a strain of V. parahaemolyticus was cloned in a pUC118-
derived plasmid and introduced into an E. coli isolate (174). The wild-type cloned gene
does not affect the MIC of quinolones, but a mutant cloned gene with a single amino
acid change, C115Y, accidentally introduced during PCR procedures showed 8- to
16-fold increases in the MICs of nalidixic acid, ciprofloxacin, and levofloxacin. After-
wards, the effect of this point mutation was confirmed by mutagenesis studies (174).
In subsequent years, a great number of chromosomally encoded Qnr-related pro-
teins have been detected, in both Gram-negative and Gram-positive microorganisms,
and their ability to confer resistance to quinolones has been confirmed in most cases
(215, 367). Thus, the 2008 Qnr nomenclature rules, in order to differentiate a plasmid-
encoded from a chromosomally encoded Qnr protein, proposed that the initials of the
microorganism be added before chromosomal Qnr (e.g., EfsQnr for chromosomal Qnr
of Enterococcus faecalis) (Table 6).
Among microorganisms possessing chromosomal Qnr, a high heterogeneity of
chromosome-encoded Qnr proteins of S. maltophilia was observed. Thus, 11 different
alleles were sequenced when SmQnr was detected (368). This finding was thereafter
confirmed (369, 370), and as mentioned above (see “Qnr Classification”), the develop-
ment of a website repository to bring order to the SmQnr nomenclature was proposed
(320). The number of SmQnr alleles has increased enormously in the last years, with
⬃150 different SmQnr alleles being detected in a GenBank search performed at the
time of this review.
Although indigenous chromosomal Qnr proteins have been detected in microor-
ganisms, such as in the above-mentioned E. faecalis and C. freundii (306, 311, 321), a
high number has been found in microorganisms from water environments (367). In fact,
as indicated when the different Qnr families are presented in this review, 4 out of 7 of
the currently established transferable Qnr families (QnrA, QnrC, QnrS, and QnrVC) are
derived from chromosomes of water-related microorganisms (367).
Qnr Structure
Qnr proteins are dimeric proteins belonging to the pentapeptide repeat protein
(PRP) family, which encompasses thousands of proteins present in more than 1,500
eukaryotic or prokaryotic organisms (371, 372).Thus, the PRP family includes MfpA-like
proteins, a chromosome-encoded Mycobacterium species protein group able to confer
slight protection against the action of quinolones (373, 374), and McbG from E. coli,
which is involved in resistance to microcin B17, a natural topoisomerase II inhibitor
(375). This protein family is characterized by a tandem repeat of 5 amino acids, in which
the residue located in a central position is indicated as “i.” Those on the right are
indicated by adding 1 or 2 (i⫹1, i⫹2), and those on the left are indicated by subtracting
1 or 2 (i⫺1, i⫺2), leading to the structure i⫺2, i⫺1, i, i⫹1, i⫹2 (372). The typical PRP motif
follows the amino acid scheme i⫺2 (S, T, A, or V), i⫺1 (D or N), i (L or F), i⫹1 (S, T, or R),
i⫹2 (G) (372).
Although data are inferred from the limited number of Qnr crystallography studies,
it has been observed that the Qnr dimer has a rodlike form in which each monomeric
structure folds into a right-handed quadrilateral -helix (376, 377). In this quadrilateral,
the PRP amino acid in position i interacts with the amino acid i⫺2, stabilizing the
structure; both residues i and i⫺2 are oriented toward the internal quadrilateral surface,
while the remaining residues are oriented toward the exterior, resulting in a usual
anionic surface (378). This quadrilateral structure has 9 square repeating units, called
coils, numbered from 0 to 9, and these coils are stacked on each other, leading to a
4-faced protein (372). Thus, each coil has 4 faces numbered from 1 to 4, and 5 amino
acids forming a PRP motif are presented on each face (372, 376). Most of the coils
present a type II turn, which clusters toward the C-terminal region, while a few (7 in
QnrB1) have a type IV turn, being located on faces 2 and 4 toward the N-terminal region
(376). Of these coils, the first (coil 0) is capped in some Qnr proteins, such as QnrB1, due
to the presence of the noncanonical PRP residues E8 and E18 at the i⫺2 position (376).
The last (coil 9) is also capped in all the Qnr proteins analyzed (376, 379) because of the
presence in the protein C terminus of the dimerization module formed by the structure
strand ()/helix (␣)/strand () (376, 377, 380). In this structure, the orientation of each
subunit is flexible thanks to the small, ⬃730-Å2/subunit, and hydrophobic contact area
(376).
In transferable as well as in several chromosomally encoded Qnr proteins, the
quadrilateral structure is broken by 2 outward-projecting loops, named loop A and loop
B, present between the second and third faces of coil 2 and between the fourth face
of coil 4 and the first face of coil 5, respectively. These loops account for 8 amino acids
(loop A) (from amino acid 46 to amino acid 53) and 12 amino acids (loop B) (from amino
acid 102 to amino acid 113) (371, 376, 377) (Fig. 8). These loops are essential for the
protective action of Qnr. Thus, in QnrB1 and AhQnr (the Qnr-like protein encoded in the
chromosome of Aeromonas hydrophila), it has been shown that a deletion of loop A
produces a strong decrease in the protective action of Qnr, while a deletion of loop B
(or of both loops) leads to a full loss of Qnr protection (376, 377). In this line, point
amino acid changes within loops may also lead to deleterious effects on protective
activity (see “In vitro mutations,” below) (Table 7). Nonetheless, it has been observed
that EfsQnr (chromosomal Qnr of E. faecalis) has no loop, retaining its protective action
(381). This finding has been related to a series of peculiarities in the N-terminal region,
in which the first 26 amino acids are not part of the quadrilateral -helix (381). Thus, the
first 18 amino acids of EfsQnr are extended over the -helix, with which they interact,
while amino acids 19 to 26 transverse the diagonal of the -helix (381). Of note, the
deletion of the first 17 amino acids strongly affects the protective action of EfsQnr (381).
FIG 8 Structure of Qnr. (A) Sequence of QnrB1. The sequence is presented under 4 columns, one for each of the four faces of the
right-handed quadrilateral -helix. At the top are the faces (each one marked with a different color; note that the colors are maintained
in the 3 panels) as well as the naming convention for each PRP residue. The positions of loop A and loop B are marked (*, loop A; **, loop
B). The sequences of both loops are at the bottom of the panel. Highlighted in salmon is the N-terminal ␣-helix. (B) Structures of 4 Qnr
proteins, QnrB1, MfpA (PRP from Mycobacterium spp.), AlbG (PRP produced by Xanthomonas albilineans), and EfsQnr (chromosomal Qnr of
Enterococcus faecalis). MfpA is able to confer slight resistance to quinolones, while AlbG results in resistance to albicin (a natural
topoisomerase inhibitor). (C) QnrB1 dimeric rodlike structure. The amino acid positions of loop A and loop B are indicated. A diamond shows
the molecular 2-fold symmetry. Type II turn-containing faces are shown as spheres, and type IV-containing faces are shown as strands.
(Reproduced from reference 376 with permission of the publisher.)
TABLE 7 Effect of specific amino acid substitutions and deletions at loops A and B on
Qnr protection levelsa
Change(s) in indicated Qnr causing a decrease in protection
Position(s) within TR QnrA1 QnrB1 QnrC1 QnrS1
Loop A ⌬A41–G56
⌬I49–E55
⌬S51–G56
Y46A
ΔY46–Q51
S50A
ΔG56/D ⌬G53A/D/E ⌬G56D/E
Loop B N103A
M104A
⌬M104–S113
I105A V108A
ΔT106 S109A
ΔT106–T107
ΔT107
ΔR108
⌬T106–R108
ΔT107-R108
M112A
W110A Y113A
F114D F111A/D F114A
C115Y C112A/Y C115A/Y
R58H ⴙ R87H ⴙ C112Yb ⴙ N178Y
S116D S113A/D S116P S116A
QnrB1
ΔM1–F15
ΔM1–I20
M205–G214
ΔI210–G214
ΔV212–G214
QnrC1
ΔI11–S20
ΔN77–G96
ΔC137–K156
aIn all cases, these deletions lead to a full loss of Qnr activity and a
subsequent lack of an effect on the final quinolone MICs. Note that
different studies have determined the Qnr effect on different
quinolones. Deletions within loop A or B are reported in Table 7.
Deletions of single amino acids are reported in Table 9 (based on
data from references 234, 235, 376, and 391–394).
quinolones interact with topoisomerase and subsequently allows Qnr to interact with
and disrupt the complex between quinolones and topoisomerase (377), and the latter
recovers its catalytic activity.
At around the same time, working with a crystallographic QnrB1 model, Vetting et
al. (376) proposed a model in which Qnr acts in a posterior step, when the DNA-poison-
topoisomerase is cleaved, destabilizing the cleavage complex and restoring DNA
replication.
Subsequent studies analyzing the effect of point mutations of either loop A or loop
B found no correlation between the Qnr-topoisomerase interaction and final protection
levels, proposing that the loops are not involved in direct interactions with topoisom-
erase but rather are involved in proper Qnr positioning in gyrase, blocking the access
of quinolones. Thus, when the Qnr-gyrase complex interacts with DNA, Qnr is removed,
and the enzyme becomes catalytic (390).
In vitro mutations. As mentioned above, the first evidence of the effect of point
mutations on quinolone protection levels conferred by Qnr determinants was observed
in 2005 due to a PCR accident which produced an amino acid change, C115Y, during
cloning of a V. parahaemolyticus chromosomal Qnr determinant (174). Further studies
analyzing the effect of the same amino acid change in QnrA1 and QnrS1 showed a
different scenario, in which despite both mutated Qnr proteins exhibiting a certain
degree of protection, this was lower than that for parental Qnr (391). Along the same
line, when this mutation was generated in the equivalent position of QnrB1 (C112), a full
loss of Qnr activity was observed (392) (Table 8).
Similar differences in final quinolone MICs were observed when different S. malto-
philia chromosomal Qnr alleles were cloned (368). Thus, SmQnr5 does not affect the
MICs of norfloxacin, ciprofloxacin, sparfloxacin, or gatifloxacin but rather has an almost
imperceptible effect (ⱕ2-fold) on the MICs of nalidixic acid, levofloxacin, and moxi-
floxacin. Meanwhile, except in the case of nalidixic acid, the effect of SmQnr6 leads to
⬎5-fold increases, reaching increases of up to ⬎62-fold for the MICs of sparfloxacin
(from ⬍0.002 to 0.125 g/ml) (368).
Rodríguez-Martínez et al. (392) analyzed the effects of different amino acid codon
changes by random and directed mutagenesis. Although most of the amino acid
variations detected led to decreased or null Qnr protection, the results showed that in
QnrS1, the amino acid change D185Y increases the MICs of ciprofloxacin and moxifloxa-
cin in E. coli 4-fold (both from 0.125 to 0.5 g/ml), and increases of 2-fold were observed
when norfloxacin, ofloxacin, and levofloxacin were tested (392). In the same study, it
was also observed that the effect of the same substitution led to different final results
when introduced into different Qnr families. Thus, the D185Y amino acid change had no
effect on the final MICs when introduced into QnrA, while it negatively impacted the
Qnr protective role when introduced into QnrB (392). Furthermore, while the deletion
of Gly56 in QnrA results in a reduction of protective levels and the substitution G56D
produces a full lack of QnrA activity, the same modifications lead to a different scenario
in QnrS, in which the deletion of G56 results in a loss of Qnr activity, and the substitution
G56D was able to confer slight protection. Meanwhile, in QnrB, both modifications
inactivated Qnr (392). It is of note that G56 (QnrA and QnrS) or the equivalent amino
acid G53 (QnrB) is located in loop A (Table 7).
In fact, a series of studies attempted to determine the impact of substitutions on the
Qnr loops or on specific positions within pentapeptides. Along these lines, as men-
tioned above, the loss of loop A, loop B, or both loops has a negative effect on QnrB
activity. In addition, it has been proposed that amino acid substitutions for polar amino
acids in position i, or for amino acids with bulky side chains in position i⫹2 or i⫺2,
destabilize the right-handed quadrilateral -helix structure, having a negative impact-
ing on Qnr activity (393, 394) (Tables 8 and 9).
The effect of amino acid changes on different levels of quinolone protection has also
been observed among different wild variants of Qnr belonging to the same family.
Thus, QnrVC7 confers lower levels of protection than QnrVC5 or QnrVC6 (394). This
finding has been related to the wild-type presence of A152 in QnrVC5 and QnrVC6.
QUINOLONE MODIFICATION
The first evidence of quinolone biodegradation was provided in the mid-1990s
when Martens and colleagues demonstrated the ability of some wood-rotting fungi to
degrade enrofloxacin (395). Further studies focused on the ability of Gloeophyllum
striatum to degrade enrofloxacin have proposed up to 4 possible degradation path-
ways, including oxidative decarboxylation, defluorination, hydroxylation at C-8, or
oxidation of the piperazinyl moiety (396), all showing different hydroxyl radical quin-
olone attack points (396). This model was thereafter expanded to other basidiomycetes,
including Cyathus stercoreus, a coprophilous fungus, and quinolones such as ciprofloxa-
cin (397). In addition, it showed the ability of other fungi (i.e., Xylaria longipes) to
transform danofloxacin into danofloxacin N-oxide, a metabolite with substantially less
antibacterial activity (398).
Further analyses of the ability of soil microorganisms to degrade quinolones suggest
the potential of different fungi (e.g., Candida spp.) and also of Mycobacterium spp. and
Pseudomonas spp. to fully degrade the danofloxacin piperazine ring (399). On the other
hand, the same study also described the ability of several microorganisms, including
Pseudomonas spp. and Mycobacterium spp., to metabolize danofloxacin to N-desmethyl
danofloxacin (399), a metabolite which retains antibacterial activity.
It is obvious that the intrinsic basal quinolone MIC is silently influenced in bacteria
that are able to inactivate or modify quinolones by any of these routes. Nonetheless, to
date, none of these mechanisms has been detected in a transferable genetic element.
AAC(6=)Ib-cr
In 2003, during a study focused on analyzing a series of plasmids carrying qnrA, a
few plasmids conferring unusually high levels of ciprofloxacin resistance (⬃4-fold
higher than usual) were detected (253). The authors suggested that differences in qnrA
expression levels might explain these differences but highlighted that the modulatory
effect on final qnr expression of unnoticed plasmid factors or the presence of unknown
TMQR might also be a reason (253). Further analyses resulted in the description of the
first bacterial enzyme able to selectively modify several fluoroquinolones, such as
ciprofloxacin and levofloxacin, increasing bacterial resistance levels (153, 170). Cur-
rently, this enzyme has been largely described in different microorganisms from various
TABLE 9 Effect of single amino acid substitutions at PRP units on Qnr protection levelsa
Change(s) in the indicated Qnr causing the indicated change in protection
QnrS1 QnrVC7
Position within TR QnrA1 (Neg) QnrB1 (Neg) QnrC1 (Neg) Neg Pos Neg Pos
i F13S
i F25A
i⫹2 T27I
i F30A
i⫺2 A36L/D
i L38P L35A L38R
i F40A
i⫺2 C43A
i F45A
i⫺2 S62D/G/L
i F66A
i⫺2 C72Y C69A/Y C72Y C72Y
i F76A
1⫺2 A82I/D/G/L/T
i⫺2 C92Y C89Y C92Y
i⫹2 G96D G93D G96D
i⫺2 A97Y
i F96A
i⫺2 A102D/G/L/T
i F101A
i⫺2 A114C/V A117C/V
i I116A
i⫺1 N120A
i L121A
i⫹2 Y123A
i⫺1 N125A
i⫺2 Q132D Q132L
i⫹1 E132A
i L136A
i⫺2 N139A
i⫹2 G148A
i⫺2 T152D/G/L T152A
i⫺1 S153P
i⫺2 S154A
i⫺1 D155A
i L159D L156A/D L159D
i⫹1 S157A
i⫺2 G159A
i F161A
i W166A
i⫺2 C172D/G C172L
i L176A
i L181A
i⫹1 D182Y D185Y
i⫺2 L184A
i⫹1 D188V
i⫹2 G193A
dm G209A
N27I ⴙ A91V ⴙ K195R
Q132L ⫹ T152A
T152A ⫹ C172L
F114Y ⫹ G196S
G7R ⫹ G98D
V6A ⫹ D95Y
aAmino acid substitutions are listed from top to bottom in the direction from the N to C termini. When more than one amino acid change is reported simultaneously,
they are placed at the bottom. Changes in the same row are analogous to each other. The amino acids highlighted in boldface type are those leading to a full loss
of Qnr activity and a subsequent lack of an effect on the final quinolone MICs. Note that cloning in different vectors may slightly affect the final full or partial loss of
activity. The amino acid change is reported only when there is a positive or negative effect of at least 4-fold on the MIC of quinolones described. Note that different
studies have determined the Qnr effects on different quinolones. No data obtained by IPTG induction have been considered because they represent the effect of
higher Qnr expression levels but not the intrinsic Qnr activity. TR, tandem repeat; dm, dimerization module; Neg, protective levels conferred by Qnr decrease; Pos,
protective levels conferred by Qnr increase (based on data from references 234, 235, 376, and 391–394).
environments, probably being the most common and frequent TMQR. In addition to its
presence in microorganisms from human samples, a recent report has shown a high
prevalence of AAC(6=)Ib-cr in animal and environmental samples (400).
The aac(6=)Ib-cr gene is frequently detected as a cassette of class 1 integrons or
associated with mobile elements such as IS26 (197, 245, 401, 402). Among these,
In37-like integrons of around 3.6 kb are largely detected in K. pneumoniae and have
disseminated to other bacterial species, including other Enterobacteriaceae and Aero-
monadaceae. In this integron, aac(6)-Ib-cr is placed in the first position, thereafter being
followed by three other gene cassettes: blaOXA-1, catB3, and arr3 (403–405). The
different In37 variants may in addition possess other antimicrobial resistance genes
after the first variable region, becoming complex integrons. Thus, the presence of qnrA1
or qnrB alleles (for instance, qnrB4 or qnrB10), which may also be linked to a blaDHA-1
gene, has also been reported in In37-like structures (245, 249, 253).
Differences have been observed in the final MICs conferred by AAC(6=)Ib-cr. Thus, in
addition to possible differences related to additional amino acid substitutions (see
below), upon analyzing 2 Klebsiella aerogenes (formerly Enterobacter aerogenes) isolates,
Ruiz et al. (402) showed differences in final ciprofloxacin MICs associated with differ-
ences in AAC(6=)Ib-cr expression levels. It was proposed that these differences were
related to the presence of a 12-bp deletion between IS26 and the initial ATG codon,
which displaces the ⫺10 box in one of the K. aerogenes isolates, leading to an almost
complete absence of expression of aac(6=)Ib-cr (402). It should be mentioned that the
AAC(6=)Ib-like enzymes have a great diversity in size (170, 406, 407); therefore, this text
follows the numeration provided by Robicsek et al. (170), but it should be noted that
other numerations may be considered in different reports (407, 408).
The originally described AAC(6=)Ib-cr protein [see “AAC(6=)Ib-cr subtypes,” below]
was reported as a protein of 172 amino acids in length belonging to the AAC(6=)Ib
group (253), but the initial ATG codon was corrected early on and was reported as a
protein of 199 amino acids in length. AAC(6=)Ib-cr is characterized by 2 amino-acid-
specific substitutions, W102R and D179Y, which are located in two external enzyme loops
(153, 407, 409). Furthermore, at the time of its description, it also presented its own
N-terminal region with 12 unique amino acids. Currently, this original enzyme is present
in GenBank as AAC(6=)Ib-cr5 (GenBank accession number WP_001749987). Subsequent
sequencing has shown the presence of several AAC(6=)Ib-cr variants with high heter-
ogeneity in size, mainly related to the enlarged N-terminal region [see “AAC(6=)Ib-cr
subtypes,” below]. The above-mentioned amino acid modifications confer to the
enzyme the ability to acetylate quinolones possessing an unsubstituted piperazinyl
group, such as ciprofloxacin and norfloxacin, but not other quinolones such as levo-
floxacin and moxifloxacin (153). In addition to the above-mentioned substrates, a later
study showed AAC(6=)Ib-cr (at least the original allelic variant described by Robicsek et
al.) activity over tosufloxacin (410). Mutagenesis analyses focused on both single amino
acid changes showed that when these are absent, the enzyme loses its ability to confer
quinolone resistance, while in the absence of W102R, D179Y also confers slight protec-
tion against quinolones but lower than that of wild-type AAC(6=)Ib-cr (153).
Similar to AAC(6=)Ib, acetyl coenzyme A acts as an acetyl donor (410), while a series
of differences regarding specific AAC(6=)Ib-cr substrates have been reported. Of these,
the most evident is a modest decrease [⬃10-fold with respect wild-type AAC(6=)Ib] in
the ability to acetylate kanamycin and the inability to acetylate neomycin B (410). In
fact, despite being characterized for its ability to acetylate some fluoroquinolones,
AAC(6=)Ib-cr acetylates kanamycin more efficiently (catalytic efficiency of kanamycin
acetylation ⬃50-fold higher than that of fluoroquinolones) (410). In addition, it is
interesting that the specific optimum substrate pHs for AAC(6=)Ib-cr also differ, being
6.1 to acetylate aminoglycosides and 7.7 to act on fluoroquinolones (410).
When AAC(6=)Ib-cr acts on aminoglycosides, the interactions are based on the
presence of numerous hydrogen-bonding interactions, while it has been suggested
that the interactions with quinolones are related to stacking interactions. In this sense,
the model of interaction between AAC(6=)Ib-cr and quinolones proposed by Vetting et
al. (410) considers that the amino nitrogen of the piperazinyl group located 2.7 Å from
Asp115 interacts with acetyl coenzyme A, playing a role as a proton acceptor during
catalysis. In this model, the quinolones establish stacking interactions with the 6/7
and ␣1=-␣2 loops of AAC(6=)Ib-cr (410). Within the ␣1=-␣2 loop, Trp49 directly interacts
with quinolones, and the Gly50-Ala54 region interacts with the pyridinone ring of the
quinolones through van der Waals forces. Tyr179 located on the 6/7 loop can interact
with the quinolone ring due to the establishment of attractive, noncovalent interac-
tions (-stacking interactions) of the p-orbitals of Tyr179 and quinolone rings. In this
model, Trp49 plays a relevant role in the stabilization of the structure. Meanwhile, Arg102
establishes interactions with Tyr179 leading to the stabilization of the optimal interac-
tion position of Tyr179 (410).
Maurice et al. (407) proposed an alternative interaction model. While this model
considered a similar role in stacking interactions for Tyr179, it proposed that Arg102
directly interacts with the keto or carboxy groups of fluoroquinolones through the
development of a hydrogen bond (407).
AAC(6=)Ib-cr subtypes. As mentioned above, 2 amino acid substitutions are suffi-
cient to confer a new ability to AAC(6=)Ib-cr. This is of special relevance because it
highlights the fact that small changes may modify the enzyme substrates. In this sense,
different AAC(6=)Ib-cr variants have been shown to confer quinolone resistance, or at
least this role has been strongly suggested (406, 409). Nonetheless, a series of studies
have not addressed this question and have mainly described the presence of
AAC(6=)Ib-cr by means of approaches such as specific PCR followed by restriction
fragment length polymorphism (RFLP) analysis (138, 411), while other studies report full
sequences presenting the 2 above-mentioned specific amino acids, as well as other
amino acid differences, even in the absence of specific studies to determine enzyme
activity. While these are valid approaches, they have the same limitations as any other
antibiotic resistance gene PCR detection method or new allele descriptions in the
absence of activity determinations. Indeed, a series of enzymes have been reported to
be able to confer quinolone resistance in the absence of definitive data.
On the other hand, different AAC(6=)Ib variants may possess the unnoticed ability to
confer slight resistance to specific quinolones. In this sense, many AAC(6=)Ib-cr se-
quences currently present in GenBank possess W102R and/or D179Y, differing mainly
in the N-terminal region or presenting a few amino acid differences. Therefore,
systematic cloning and evaluation of these enzymes could lead to a series of
relevant modifications in the current knowledge about this mechanism of quino-
lone resistance. In this sense, Kim et al. (406) detected a variant of AAC(6=)Ib-cr able
to acetylate gemifloxacin and zabofloxacin. This AAC(6=)Ib-cr variant was charac-
terized to present Y179 plus L117 (406), with both amino acids being present in the
enzyme described by Robicsek et al. (170) but lacking the amino acid change W102R
(406). Table 10 shows a series of AAC(6=)Ib-cr variants which have shown the ability
to acetylate quinolones.
cFollowing the numeration provided by Robicsek et al. (170), note that the different sizes of acetylases may lead to different numeric positions in any variant. For
instance, the reported amino acid substitution “N20T” under GenBank accession number WP_015059932 is located in the equivalent position 46. When longer, amino
acid positions prior to M1 under GenBank accession number WP_001749987 have not been analyzed.
dQuinolones which might be acetylated.
eNo GenBank accession number is provided in the article by Kim et al. (406), and after a search in GenBank using the sequence under accession number
WP_001749987 and introducing a glycine in position 179, no concordant enzyme was found among the first 100 results.
catalytic APH enzyme motifs, presents Gly7 and Ile26, two conserved residues in APH
enzymes which are involved in catalysis and in ATP binding, respectively. Of note, the
concomitant presence of G7 and Ile26 was detected only in 63 of the ⬎1,000 closely
related (identity levels of ⱖ90%) amino acid sequences present in GenBank (see below)
(414). CrpP seems to be able to confer a slight increase in the MIC of ciprofloxacin (from
0.008 to 0.06 g/ml) when cloned into pUC_20 and introduced into an E. coli J53
environment but had no effect on the remaining quinolones tested (levofloxacin,
moxifloxacin, nalidixic acid, and norfloxacin). Interestingly, upon comparing the effects
of CrpP when cloned into pUC_20 and when the native pUM505 plasmid was trans-
ferred to another P. aeruginosa isolate, the effect of the native plasmid was greater and
extended to norfloxacin and moxifloxacin, suggesting the presence of another un-
known quinolone resistance mechanism (154). Further analysis using the above-
mentioned quinolones as substrates showed that CrpP is able to phosphorylate nor-
floxacin but with an even lower efficiency than that of ciprofloxacin (with a Vmax/Km
ratio approximately 5-fold lower than that for ciprofloxacin) (154). Due to the novelty
of the CrpP description (to my knowledge, at the time of writing of this review, it has
been reported in only four articles), there is practically no knowledge of the extension of
this mechanism. Nonetheless, more than a thousand CrpP-like proteins grouped into more
than 37 different alleles, possessing one or a few amino acid changes, are present in
GenBank (414). Interestingly, all these sequences except for one uncharacterized 102-
amino-acid-long protein of A. baumannii (GenBank accession number SST11961) belong to
members of the Pseudomonadaceae (414). Most of these sequences were reported within
the P. aeruginosa genomic islands PAPI-1 and PAGI-5 (414), which are able to disseminate
through conjugation (415). These genomic islands have evolved from the ancestral conju-
gative plasmid pKLC102, which has been proposed to have an environmental origin (416).
These finding might suggest that the origin of CrpP is from plant-related bacterial com-
munities, perhaps belonging to the Pseudomonadaceae (414).
At the beginning of 2019, Chávez-Jacobo et al. (417) explored the presence of
crpP-like genes in 77 ESBL-carrying Enterobacteriaceae and in a historical collection of
66 E. coli J53-2 isolates containing plasmids transferred from ESBL-carrying clinical
isolates which were collected between 1988 and 2012. The authors detected 9 samples
(4 clinical isolates and 5 plasmids from the historical collection) carrying a series of
related crpP genes. Of these, the original sources were an E. coli isolate in 5 cases and
a K. pneumoniae isolate in the remaining 4 cases (417). Further analysis of 5 of these
plasmids (2 from E. coli and 3 from K. pneumoniae) showed that the first 2 encoded
products had protein identity levels ranging from 10.1% to 43.7% with original
CrpP, while the identity levels between the 3 CrpP-like proteins from K. pneumoniae
were higher, ranging between 57.1% and 65.7% (417). An analysis of the specific
effects of 3 of these CrpP-like proteins on the ciprofloxacin MICs showed increases
equivalent to those of the original CrpP protein (417). Nonetheless, neither data on
the presence of quinolone target selection nor those on the nalidixic acid MIC were
provided, allowing the possible selection of spontaneous mutants. In this sense, an
in silico analysis of these crpP-like sequences showed the presence of base inser-
tions/deletions that result in frameshifts (414). Pending further confirmation, cur-
rent data suggest the existence of a family of genes with the ability to slightly affect
the MICs of specific quinolones.
It should be taken into account that the authors proposed that a carboxyl group
present at position 3 of the ciprofloxacin molecule binds with ATP to be thereafter
phosphorylated and degraded (154). This radical is common to all quinolones (except
metioxate) (Fig. 3); therefore, it should be considered that CrpP may present activity
against untested quinolones, and as indicated above, the presence of CrpP modifica-
tions may lead to alterations of either expansion or constraint in the substrate pattern
as well as the final activity levels. In this sense, further antibiotic spectrum determina-
tion of detected CrpP variants will be of relevance.
OqxAB
In 2003, Sørensen et al. (155) were the first to describe the presence of a plasmid
(pOLA52)-borne mechanism of olaquindox resistance in E. coli isolated from swine.
Further studies to determine the exact mechanism of olaquindox resistance encoded
within pOLA52 showed its efflux pump nature, detecting 2 ORFs, encoding a 391- and
a 1,050-amino-acid-long protein, which were named OqxA and OqxB (here, these 2
specific alleles are referred to as OqxA1 and OqxB1). OqxA and OqxB present a certain
degree of identity, especially at the amino acid level, with inner membrane (OqxA) and
periplasmic (OqxB) components of previously described RND efflux pumps, such as
MexEF of Xanthomonas axonopodis, MexXY of P. aeruginosa, and AcrAB of E. coli (156).
The RND efflux pumps need a third component to be active, that is, an outer membrane
protein (OMP). Hansen et al. (156) showed that in an E. coli genetic background, TolC
plays this role, and therefore, TolC-like proteins presumably play this role in other
Enterobacteriaceae. Nonetheless, the role of OqxAB as a quinolone resistance mecha-
nism was not established until 2007, when its ability to extrude different quinolones,
such as nalidixic acid, ciprofloxacin, and norfloxacin, together with a series of unrelated
antibacterial agents and toxins was observed (157). Similar to what has been described
for other RND efflux pumps, such as AcrAB-like efflux pumps in Enterobacteriaceae or
MexAB-like efflux pumps in Pseudomonadaceae (239, 418, 419), it has been suggested
that Phe-Arg--naphtylamide may inhibit OqxAB (420).
Further studies aimed at characterizing pOLA52 showed a plasmid size of 51,602 bp,
classified as belonging to the IncX1 incompatibility group (200). Norman et al. (200) also
showed that together with a putative regulator gene (later proposing the name OqxR),
OqxAB proteins were flanked by IS26, thus being a composite transposon, demonstrat-
ing the presence of 100% (oqxA) and 99% (oqxB) identities with the components of a
K. pneumoniae putative efflux pump, which was thereafter considered the OqxAB
original bacterial source (199–201). Posterior analyses of the role of OqxR have shown
that it may be involved in the downregulation of OqxAB expression levels. Thus, upon
analyzing the expression levels of K. pneumoniae chromosomal oqxA and oqxB genes,
it was shown that the presence of the specific OqxR amino acid changes reported
below correlates with higher levels of expression of these genes, which revert when a
plasmid carrying wild-type OqxR is used to complement the bacteria (421, 422):
● F6S
● V102G
● Q11L, D95E, and V113I
● N38T, D95E, V113I, and H159L
● Δ73–77
● Δ88 –94
where Δ indicates a deletion. An association between these mutations and the final
levels of OqxAB expression has been proposed (421, 422). Note that when there is
more than one amino acid change, the presence of simple polymorphisms needs to
be considered. To my knowledge, there is no study on the role of OqxR in
transferable oqxAB expression levels. Nonetheless, transferable oqxAB regulation
mediated by chromosomal regulatory genes such as ramA and rarA has been shown
in S. Typhimurium (201, 420). This finding correlates with the final MICs of the
antibacterial agents tested. Therefore, the specific chromosomal genetic back-
ground plays a crucial role in the final levels of expression of oqxAB and, subse-
quently, in its contribution to the final antimicrobial resistance levels. This finding
can likely be extrapolated to other TMQR.
Although the presence of differences in the sequence of K. pneumoniae chromo-
some-encoded OqxAB has been described (tens of different chromosomal OqxA and
OqxB alleles are recorded in GenBank), most of the studies on transferable OqxAB
address only the presence/absence of OqxAB. In 2012, the presence of new transferable
allelic variants of OqxA and OqxB were described, differing in 1 or 2 amino acids from
the original OqxA and OqxB (thereafter named OqxA1 and OqxB1, respectively) (177).
Furthermore, an in silico analysis of the presence of new transferable OqxA/OqxB
variants in GenBank showed the presence of at least 14 new OqxA and 28 OqxB
previously unnoticed variants, which had clearly evolved from K. pneumoniae chromo-
somal OqxAB, as well as the dissemination of these variants through a series of
microorganisms (Tables 11 and 12). While most of these alterations are probably
polymorphisms that do not affect the final OqxAB activities, the possible involvement
of activity levels and/or the expansion/contraction of substrate profiles cannot be ruled
out. In any case, the effect of these modifications on the final activity of OqxAB remains
to be elucidated. Interestingly, K. pneumoniae is not the only member of the Entero-
bacteriaceae carrying an OqxAB-like efflux pump. Closely related efflux pumps have
been described as being indigenous to other microorganisms such as other Klebsiella
spp. or Enterobacter spp. (201). In this regard, the above-mentioned analysis also
showed the presence in GenBank of an E. coli genome (strain TUM18641; GenBank
accession number NZ_BGTY01000006) carrying OqxA (GenBank accession number
WP_087857967) and OqxB (GenBank accession number WP_113374178) with 30 and 29
amino acid differences from OqxA1 and OqxB1, respectively, but closely related to
OqxAB of K. aerogenes; thus, OqxA presents a maximum identity of 100% with that of
K. aerogenes strain CRK0055 (GenBank accession number RNT35078), and OqxB, with
only 2 amino acid differences, presents an identity of 99.8% with OqxB of K. aerogenes
strains AR_0431 and GN04835 (GenBank accession number WP_047066491). Therefore,
the unnoticed dissemination of another variant of OqxAB with an undefined effect on
quinolones (or other antibiotic agents) has been suggested. This finding, especially if
this new OqxAB-like protein expands to other E. coli isolates or other microorganisms,
opens the door to suggesting an alternative name for this new variant, as occurred with
QnrE.
Internal partial genomic integrations or recombinations of transferable genetic
structures leading to the full or partial loss of oqxA or oqxB may explain the studies in
which only one of these genes is detected in nonindigenous microorganisms (341, 423).
cDate of first published report. If no published report has been found, the data for the older GenBank record are reported.
dThe minimum degree of identity considered was 99%. A series of OqxAB efflux pumps with lower levels of identity are present in GenBank, including a transferable oqxAB gene probably derived from an ancestral
environment data. The presence of transferable OqxA in Klebsiella spp. is not reported here.
fFirst published report.
cmr.asm.org 40
Clinical Microbiology Reviews
TABLE 12 Amino acid changes described in transferable OqxBa
GenBank accession no.b Gram-negative
% genus or
Yrc DNA Amino acid identityd Amino acid difference generae Referencef
2007 EU370913g WP_000347934 100.0 P102 L138 G148 V217 M313 Q325 A432 L284 H434 G463 A493 G540 G544 G547 A551 V612 V645 A650 L672 A699 Q778 D749 Y783 E788 Q799 V839 T1050 ins Enterobacter, 156
Escherichia,
Salmonella,
Shigella
2012 JF912901 AEM45928 99.9 I Escherichia 177
2017 KX518744 APO16513 99.9 I Escherichia 468
2018 NZ_NRYU01000105 WP_113449358 99.9 L Escherichia
2018 NZ_PJHN01000068 WP_112910686 99.9 Q Escherichia 469
2018 NZ_PJGF01000097 WP_112889165 99.9 K Escherichia 469
2017 MOIC01000152 OJO61665 99.9 E Escherichia 470
2016 FLWH01000008 SBY67683 99.9 N Escherichia
2018 NZ_NRXP01000002 WP_113402128 99.9 S Escherichia
cDate of the first published report. If no published report has been found, the data for the older GenBank record are reported.
dThe minimum degree of identity considered was 99%. A series of OqxAB efflux pumps with lower levels of identity are present in GenBank, including a transferable oqxAB gene probably derived from an ancestral K.
environment data. The presence of transferable OqxB in Klebsiella spp. is not reported here.
fFirst published report.
cmr.asm.org 41
Clinical Microbiology Reviews
Ruiz Clinical Microbiology Reviews
In addition, the presence of transferable OqxAB not derived from the K. pneumoniae
genome may also contribute to explaining this phenomenon.
Initial studies were developed using samples of veterinary origin and thereafter in
animal-exposed human populations (e.g., farmworkers), suggesting a relevant role of
livestock management in the selection and spread of OqxAB-carrying isolates (155,
230). Nonetheless, despite the relevance of the farm environment, transferable OqxAB
proteins have currently also been described in samples from healthy and sick people as
well as from food samples, wild animals, and environmental samples (202, 424–426).
Furthermore, the range of plasmids in which oqxAB has been reported has expanded,
including plasmids classified within the IncF or IncHI2 group, among others (354, 425,
427).
In 2018, the presence of OqxAB in Gram-positive microorganisms was first reported.
A study performed in China showed the presence of this efflux pump in E. faecalis in a
genetic structure flanked by IS26 and linked to an incomplete aph(3=)-IIa gene (169).
Interestingly, these samples were from a pig farm.
QepA
To date, 4 QepA variants have been described in the literature. The first qepA gene
was described at almost the same time by 2 independent French and Japanese groups
in 2007 (158, 159). A new QepA variant, QepA2, differing in 2 amino acid codons, was
described in 2008 (178). Finally, QepA3 and QepA4 were described in 2015 and 2017,
respectively (428–430). Nonetheless, an analysis of GenBank records showed that at
least 6 other new complete qepA genes have been fully sequenced as well as the
presence of additional partial QepA sequences exhibiting their own specific amino acid
codon changes (179). All 6 uncharacterized QepA proteins were detected in members
of the Enterobacteriaceae, thereby being acquired from an external source, irrespective
of their exact genetic environment (chromosome or plasmid) (179). Similar to Qnr (and,
in fact, with a long series of genes), a centralized QepA repository is needed to bring
order to the nomenclature.
Similar to other TMQR, the mobilization of qepA has been related to the presence of
different insertion sequences, such as IS26 and ISCR3C (158, 175, 430). The qepA4 gene
has been detected in an atypical class 1 integron preceded by a defective dfrB4 gene
and followed by an ISCR3 element (428).
QepA is a proton-dependent efflux pump which varies in size, presenting 14
transmembrane segments (14-TMS) which belong to the major facilitator super-
family (MFS) (159). An extrusion product pattern has been established in QepA1 and
QepA2, including more-hydrophilic quinolones such as ciprofloxacin and norfloxa-
cin but with a marginal or null ability to extrude hydrophobic quinolones such as
ofloxacin levofloxacin, moxifloxacin, and nalidixic acid (159, 178, 431). Furthermore,
these variants are unable to pump out nonquinolone agents such as erythromycin,
chloramphenicol, rifampin, and tetracycline (159, 178). No data on pumping sub-
strates of the remaining QepA variants are available. In this regard, it should be
mentioned that despite having identities of ⱖ90% among the 10 QepA variants,
these slight differences include insertions and deletions (179). Thus, while the 4
reported QepA variants have 511 amino acids, a deletion of 5 amino acids
(Δ432SAALP), just after the 13th transmembrane section, and an insertion of 2 amino
acids (Ins357LG) within the 11th transmembrane transept have been detected in
QepA GenBank records (179). These insertions and deletions may affect the final
QepA structure and may therefore have a major impact on either the expansion or
the restriction of efflux pump substrate profiles as well as on substrate affinities,
which may lead to different efficiencies of pumping out, with a direct impact on the
final effect on antimicrobial resistance levels.
The first phylogenetic analysis proposed that the origin of QepA was within the
Actinomycetales order (158). Furthermore, a possible ancestor within the actinomycetes
also agrees with the G⫹C content of QepA (⬃72%) (158). Nonetheless, the addition of
new genomic data in GenBank has shown that the higher levels of identity with the
currently described or putative Actinomycetales efflux pumps are ⬃40%, while the
identities of QepA with a series of 512 putative efflux pumps of the Comamonadaceae
family (i.e., Pseudorhodoferax spp.) reach values of ⬃80%. Despite the scarce data in this
respect, the G⫹C content of the Pseudorhodoferax genus has been reported to be ⬃68
to 70%, thereby agreeing with that of QepA. These findings suggested the possibility
of an alternative origin within the Comamonadaceae family (179). In agreement with
this alternative origin of QepA, in a study by Yamane et al., the chromosomal efflux
pump with higher levels of identity with QepA1 in fact belonged to a member of the
Comamonadaceae (i.e., Polaromonas spp.) (158).
CONCLUSION
Within a little over 20 years, TMQR have evolved from being a scientific, almost
undetectable rarity to a worldwide epidemic. This finding has been made possible by
FIG 9 Selection of antibiotic resistance. (A) In the preantibiotic era, the presence of antibiotic-resistant
microorganisms was rare. The introduction of antibiotics led to killing of antibiotic-susceptible bacterial
populations; the selection of the minority antibiotic-resistant populations, which were present in the
preantibiotic era; the dispersion of plasmid-encoded mechanisms of resistance; as well as the generation
of new resistant strains (by selecting chromosomal mutations). This phenomenon led to the current high
levels of antibiotic resistance. (B) Antibiotic resistance may be stable, with a minimal or null effect on bacterial
fitness, thus being easily fixed in the bacterial population. The antibiotic-resistant microorganisms tend to be
present even in the absence of antibiotic pressure. Antibiotic resistance may also be unstable, affecting
bacterial fitness; the absence of antibiotic pressure tends to dilute in the general bacterial population.
Nonetheless, antibiotic resistance may act in an indirect manner by other antibacterial agents favoring the
fixing of genetic structures carrying multiple mechanisms of antibiotic resistance. Compensatory mutations
are mutations which have no direct effect on antibiotic resistance but lead to the recovery of lost fitness.
When selected, the microorganisms act as those with stable resistance.
human attitudes. Thus, quinolone use, similar to that of all antibacterial agents, has
become one of absolute overuse and misuse; that is, the use of quinolones has
expanded beyond the treatment of bacterial infectious diseases to a series of unnec-
essary roles, such as livestock growth promotion. This, in addition to the high levels of
use in veterinary settings as a prophylactic agent, the uncontrolled access to antibac-
terial agents in a long series of countries, the high rate of misprescription, self-
medication, erroneous posology, and early treatment discontinuation, has led to the
current scenario of quinolone resistance that, as with all other antibacterial agents, has
squandered the treasure that antimicrobials are.
The direct, continuous, and growing selection pressure exerted by antibiotics in
human and microbiota commensal members and pathogenic microorganisms is inces-
sant, favoring the survival of microorganisms which carry antibiotic resistance deter-
minants such as Qnr or other TMQR. These microorganisms that possess adequate
antibiotic resistance genes are the winners of the race involving the survival of bacteria
in the antibiotic era (Fig. 9).
It is undeniable that TMQR will remain with us. At present, not even the strictest
antibiotic control policies will return the presence of TMQR determinants to the level of
the prequinolone era, but these measures are necessary to control and limit their
continuous spread.
ACKNOWLEDGMENTS
This research received no specific grant from any funding agency in the public,
commercial, or not-for-profit sectors.
I thank Donna Pringle for language correction.
REFERENCES
1. Burman LG. 1977. Apparent absence of transferable resistance to nali- 23. Slade DA. 1963. A clinical trial of nalidixic acid (Negram, WIN. 18,320) in
dixic acid in pathogenic Gram-negative bacteria. J Antimicrob Che- thirty-three patients with urinary infections treated in general practice. Br
mother 3:509 –516. https://doi.org/10.1093/jac/3.5.509. J Urol 35:125–128. https://doi.org/10.1111/j.1464-410X.1963.tb02604.x.
2. Jonsson M. 1973. Antibiotic resistance and R factors in Gram-negative 24. Cahal DA. 1965. Nalidixic acid—request for prompt reporting of sus-
bacteria isolated in a hospital for infectious diseases. III. The effect of pected adverse reactions. Br Med J i:130.
antibacterial treatment on the incidence of R factor-mediated antibiotic 25. Tillotson GS. 1996. Quinolones: structure-activity relationships and fu-
resistance. Scand J Infect Dis 5:41– 47. https://doi.org/10.3109/inf.1973 ture predictions. J Med Microbiol 44:320 –324. https://doi.org/10.1099/
.5.issue-1.08. 00222615-44-5-320.
3. Lebek G. 1967. Medizinische aspekte der infektiösen antibiotika-resistenz 26. Emmerson AM, Jones AM. 2003. The quinolones: decades of develop-
gram-negativer darmbakterien. Pathol Microbiol 30:1015–1036. https://doi ment and use. J Antimicrob Chemother 51(Suppl S1):13–20. https://doi
.org/10.1159/000161739. .org/10.1093/jac/dkg208.
4. Babcock GF, Berryhill DL, Marsh DH. 1973. R-factors of Escherichia coli 27. Heyman SN, Ginosar Y, Shapiro M, Kluger Y, Marx N, Maayan S. 1997.
from dressed beef and humans. Appl Microbiol 25:21–23. Diarrheal epidemics among Rwandan refugees in 1994: management
5. Munshi MH, Sack DA, Haider K, Ahmed ZU, Rahaman MM, Morshed MG. and outcome in a field hospital. J Clin Gastroenterol 25:595– 601.
1987. Plasmid-mediated resistance to nalidixic acid in Shigella dysente- https://doi.org/10.1097/00004836-199712000-00009.
riae type 1. Lancet ii:419 – 421. https://doi.org/10.1016/s0140-6736 28. Mandomando IM, Macete EV, Ruiz J, Sanz S, Abacassamo F, Vallès X,
(87)90957-3. Sacarlal J, Navia MM, Vila J, Alonso PL, Gascon J. 2007. Etiology of
6. Panhotra BR, Desai B, Sharma PL. 1985. Nalidixic-acid-resistant Shigella diarrhea in children younger than 5 years of age admitted in a rural
dysenteriae I. Lancet i:763. https://doi.org/10.1016/s0140-6736 hospital of southern Mozambique. Am J Trop Med Hyg 76:522–527.
(85)91310-8. https://doi.org/10.4269/ajtmh.2007.76.522.
7. Clerch B, Rivera E, Llagostera M. 1996. Identification of a pKM101 region 29. Rogerie F, Ott D, Vandepitte J, Verbist L, Lemmens P, Habiyaremye I.
which confers a slow growth rate and interferes with susceptibility to 1986. Comparison of norfloxacin and nalidixic acid for treatment of
quinolone in Escherichia coli AB1157. J Bacteriol 178:5568 –5572. dysentery caused by Shigella dysenteriae type 1 in adults. Antimicrob
https://doi.org/10.1128/jb.178.19.5568-5572.1996. Agents Chemother 29:883– 886. https://doi.org/10.1128/aac.29.5.883.
8. Sanders CC, Sanders WE, Jr, Goering RV, Werner V. 1984. Selection of 30. Salam MA, Bennish ML. 1988. Therapy for shigellosis. I. Randomized,
multiple antibiotic resistance by quinolones, beta-lactams, and amin- double-blind trial of nalidixic acid in childhood shigellosis. J Pediatr
oglycosides with special reference to cross-resistance between unre- 113:901–907. https://doi.org/10.1016/S0022-3476(88)80029-5.
lated drug classes. Antimicrob Agents Chemother 26:797– 801. https:// 31. Turner FJ, Ringel SM, Martin JF, Storino PJ, Daly JM, Schwartz BS. 1967.
doi.org/10.1128/aac.26.6.797. Oxolinic acid, a new synthetic antimicrobial agent. I. In vitro and in vivo
9. Smith JT. 1986. Frequency and expression of mutational resistance to activity. Antimicrob Agents Chemother 7:475– 479.
the 4-quinolone antibacterials. Scand J Infect Dis Suppl 49:115–123. 32. Shimizu M, Nakamura S, Takase Y. 1970. Piromidic acid, a new antibac-
10. Courvalin P. 1990. Plasmid-mediated 4-quinolone resistance: a real or terial agent: antibacterial properties. Antimicrob Agents Chemother
apparent absence? Antimicrob Agents Chemother 34:681– 684. https:// 10:117–122.
doi.org/10.1128/aac.34.5.681. 33. Wick WE, Preston DA, White WA, Gordee RS. 1973. Compound 64716,
11. Crumplin GC. 1987. Plasmid-mediated resistance to nalidixic acid and a new synthetic antibacterial agent. Antimicrob Agents Chemother
new 4-quinolones. Lancet ii:854 – 855. https://doi.org/10.1016/s0140 4:415– 420. https://doi.org/10.1128/aac.4.4.415.
-6736(87)91039-7. 34. de Lajudie P, Roquet F, Reynier M, Adamowicz P. 1974. Etude d’un
12. Ashraf MM, Ahmed ZU, Sack DA. 1991. Unusual association of a plasmid nouvel antibactérien de synthesé: l’acyde éthyl-8 oxo-5 pipérazinyl-2
with nalidixic acid resistance in an epidemic strain of Shigella dysente- dihydro-5,8 pyrido [2,3-d]pyrimidine-6 carboxylique (ácide pipemi-
riae type 1 from Asia. Can J Microbiol 37:59 – 63. https://doi.org/10 dique). C R Acad Sci Hebd Seances Acad Sci D 279:1931–1934.
.1139/m91-009. 35. Ito A, Hirai K, Inoue M, Koga H, Suzue S, Irikura T, Mitsuhashi S. 1980. In
13. Jacoby GA, Chow N, Waites KB. 2003. Prevalence of plasmid-mediated vitro antibacterial activity of AM-715, a new nalidixic acid analog.
quinolone resistance. Antimicrob Agents Chemother 47:559 –562. Antimicrob Agents Chemother 17:103–108. https://doi.org/10.1128/aac
https://doi.org/10.1128/AAC.47.2.559-562.2003. .17.2.103.
14. Lesher GY, Froelich EJ, Gruett MD, Bailey JH, Brundage RP. 1962. 36. Stilwell G, Holmes K, Turck M. 1975. In vitro evaluation of a new
1,8-Naphthyridine derivatives: a new class of chemotherapy agents. J quinolone antibacterial. Antimicrob Agents Chemother 7:483– 485.
Med Chem 5:1063–1068. https://doi.org/10.1021/jm01240a021. https://doi.org/10.1128/aac.7.4.483.
15. Martínez-Martínez L, Pascual A, Jacoby GA. 1998. Quinolone resistance 37. Ruiz J. 2003. Mechanisms of resistance to quinolones: target alterations,
from a transferable plasmid. Lancet 351:797–799. https://doi.org/10 decreased accumulation and DNA gyrase protection. J Antimicrob
.1016/S0140-6736(97)07322-4. Chemother 51:1109 –1117. https://doi.org/10.1093/jac/dkg222.
16. Price JR. 1949. Alkaloids of the Australian Rutaceae: Melicope fareana. 38. Fung-Tomc JC, Minassian B, Kolek B, Huczko E, Aleksunes L, Stickle T,
IV. Some reactions of I-methyl-y-quinolone-3-carboxylic acid, a degra- Washo T, Gradelski E, Valera L, Bonner DP. 2000. Antibacterial spectrum of
dation product of the alkaloids. Aust J Sci Res 2:272–281. https://doi a novel des-fluoro(6) quinolone, BMS-284756. Antimicrob Agents Che-
.org/10.1071/CH9490272. mother 44:3351–3356. https://doi.org/10.1128/AAC.44.12.3351-3356.2000.
17. Barton N, Crowther AF, Hepworth W, Richardson ND, Driver GW. 1960. 39. Gropper S, Albareda N, Chelius K, Kruger D, Mitha I, Vahed Y, Gani M,
New quinolones and therapeutic compositions containing them. British García-Alonso F, Ozenoxacin in Impetigo Trial Investigators Group.
patent 830832. 2014. Ozenoxacin 1% cream in the treatment of impetigo: a multi-
18. Bisacchi GS. 2015. Origins of the quinolone class of antibacterials: an center, randomized, placebo- and retapamulin-controlled clinical trial.
expanded “discovery story.” J Med Chem 58:4874 – 4882. https://doi Future Microbiol 9:1013–1023. https://doi.org/10.2217/fmb.14.78.
.org/10.1021/jm501881c. 40. Poole RM. 2014. Nemonoxacin: first global approval. Drugs 74:
19. Hepworth W. 1959. Manufacture of 1-methyl-6-nitro-4-quinolone-3- 1445–1453. https://doi.org/10.1007/s40265-014-0270-0.
carboxylic acid. British patent 820295. 41. Roychoudhury S, Ledoussal B. 2002. Non-fluorinated quinolones
20. Carroll G. 1963. Neggram (nalidixic acid): a new antimicrobial chemo- (NFQs): new antibacterials with unique properties against quinolone-
therapeutic agent. J Urol 90:476 – 478. https://doi.org/10.1016/s0022 resistant gram-positive pathogens. Curr Drug Targets Infect Disord
-5347(17)64441-0. 2:51– 65. https://doi.org/10.2174/1568005024605891.
21. Lishman IV, Swinney J. 1963. Studies of a new antibacterial agent, 42. Wiles JA, Bradbury BJ, Pucci MJ. 2010. New quinolone antibiotics: a
nalidixic acid. Br J Urol 35:116 –121. https://doi.org/10.1111/j.1464 survey of the literature from 2005 to 2010. Expert Opin Ther Pat
-410X.1963.tb02602.x. 20:1295–1319. https://doi.org/10.1517/13543776.2010.505922.
22. Jameson RM, Swinney J. 1963. A clinical trial of the treatment of 43. Slayton ET, Hay AS, Babcock CK, Long TE. 2016. New antibiotics in
gram-negative urinary infections with nalidixic acid. Br J Urol 35: clinical trials for Clostridium difficile. Expert Rev Anti Infect Ther 14:
122–124. https://doi.org/10.1111/j.1464-410X.1963.tb02603.x. 789 – 800. https://doi.org/10.1080/14787210.2016.1211931.
44. Klopman G, Wang S, Jacobs MR, Bajaksouzian S, Edmonds K, Ellner JJ. 65. Song TR, Rao ZS, Qiu Y, Liu JP, Huang ZL, Wang XD, Lin T. 2016.
1993. Anti-Mycobacterium avium activity of quinolones: in vitro activi- Fluoroquinolone prophylaxis in preventing BK polyomavirus infection
ties. Antimicrob Agents Chemother 37:1799 –1806. https://doi.org/10 after renal transplant: a systematic review and meta-analysis. Kaohsi-
.1128/aac.37.9.1799. ung J Med Sci 32:152–159. https://doi.org/10.1016/j.kjms.2016.01.004.
45. Mandell L, Tillotson G. 2002. Safety of fluoroquinolones: an update. Can 66. Beteck RM, Smit FJ, Haynes RK, N’Da DD. 2014. Recent progress in the
J Infect Dis 13:54 – 61. https://doi.org/10.1155/2002/864789. development of anti-malarial quinolones. Malar J 13:339. https://doi
46. Alksne L. 2003. Balofloxacin Choongwae. Curr Opin Investig Drugs .org/10.1186/1475-2875-13-339.
4:224 –229. 67. Dalhoff A. 2015. Antiviral, antifungal, and antiparasitic activities of
47. Panda BK, Pawar A, Patel C, Marne SR, Singh P. 2012. Balofloxacin (Q35), fluoroquinolones optimized for treatment of bacterial infections: a
new fluoroquinolone as an anti-infective: a systematic review. Am J puzzling paradox or a logical consequence of their mode of action? Eur
Pharmtech Res 2:287–303. J Clin Microbiol Infect Dis 34:661– 668. https://doi.org/10.1007/s10096
48. King DE, Malone R, Lilley SH. 2000. New classification and update on -014-2296-3.
the quinolone antibiotics. Am Fam Physician 61:2741–2748. 68. Gaillard T, Madamet M, Tsombeng FF, Dormoi J, Pradines B. 2016.
49. Naber K, Adam D. 1998. Classification of fluoroquinolones. Int J Anti- Antibiotics in malaria therapy: which antibiotics except tetracyclines
microb Agents 10:255–257. https://doi.org/10.1016/S0924-8579 and macrolides may be used against malaria? Malar J 15:556. https://
(98)00059-4. doi.org/10.1186/s12936-016-1613-y.
50. Flamm RK, Vojtko C, Chu DT, Li Q, Beyer J, Hensey D, Ramer N, Clement 69. Montazeri M, Sharif M, Sarvi S, Mehrzadi S, Ahmadpour E, Daryani A.
JJ, Tanaka SK. 1995. In vitro evaluation of ABT-719, a novel DNA gyrase 2017. A systematic review of in vitro and in vivo activities of anti-
inhibitor. Antimicrob Agents Chemother 39:964 –970. https://doi.org/ Toxoplasma drugs and compounds (2006-2016). Front Microbiol 8:25.
10.1128/aac.39.4.964. https://doi.org/10.3389/fmicb.2017.00025.
51. Alonso D, Muñoz J, Ruiz J, Carmona F, Nadal A, Gascón J. 2007. Salmonella 70. Romero IC, Saravia NG, Walker J. 2005. Selective action of fluoroquino-
ovarian abscess following travel diarrhoea episode. Arch Gynecol Obstet lones against intracellular amastigotes of Leishmania (Viannia) pana-
276:551–553. https://doi.org/10.1007/s00404-007-0380-y. mensis in vitro. J Parasitol 91:1474 –1479. https://doi.org/10.1645/GE
52. Horcajada JP, Vila J, Moreno-Martínez A, Ruiz J, Martínez JA, Sánchez M, -3489.1.
Soriano E, Mensa J. 2002. Molecular epidemiology and evolution of 71. Leyva-Ramos S, Hernández-López H. 2017. Fluoroquinolonas: perspec-
resistance to quinolones in Escherichia coli after prolonged administra- tivas no antibacterianas. Rev Esp Quimioter 30:1– 8.
tion of ciprofloxacin in patients with prostatitis. J Antimicrob Che- 72. Stergiopoulou T, Meletiadis J, Sein T, Papaioannidou P, Tsiouris I, Roilides
mother 49:55–59. https://doi.org/10.1093/jac/49.1.55. E, Walsh TJ. 2009. Comparative pharmacodynamic interaction analysis
53. Kohlhoff SA, Hammerschlag MR. 2015. Treatment of chlamydial between ciprofloxacin, moxifloxacin and levofloxacin and antifungal
infections: 2014 update. Expert Opin Pharmacother 16:205–212. agents against Candida albicans and Aspergillus fumigatus. J Antimicrob
https://doi.org/10.1517/14656566.2015.999041. Chemother 63:343–348. https://doi.org/10.1093/jac/dkn473.
54. Mogle BT, Steele JM, Thomas SJ, Bohan KH, Kufel WD. 2018. Clinical 73. Jeffers-Francis LK, Burger-Calderon R, Webster-Cyriaque J. 2015. Effect
review of delafloxacin: a novel anionic fluoroquinolone. J Antimicrob of leflunomide, cidofovir and ciprofloxacin on replication of BKPyV in a
Chemother 73:1439 –1451. https://doi.org/10.1093/jac/dkx543. salivary gland in vitro culture system. Antiviral Res 118:46 –55. https://
55. Ruiz J, Marco F, Oliveira I, Vila J, Gascón J. 2007. Trends in antimicrobial doi.org/10.1016/j.antiviral.2015.02.002.
resistance in Campylobacter spp. causing traveler’s diarrhea. APMIS 74. Khan IA, Siddiqui S, Rehmani S, Kazmi SU, Ali SH. 2012. Fluoroquino-
115:218 –224. https://doi.org/10.1111/j.1600-0463.2007.apm_567.x. lones inhibit HCV by targeting its helicase. Antivir Ther 17:467– 476.
56. Wang J, Xu H, Wang D, Li M. 2017. Comparison of pathogen eradication https://doi.org/10.3851/IMP1937.
rate and safety of anti-bacterial agents for bronchitis: a network meta- 75. Yamaya M, Nishimura H, Hatachi Y, Yasuda H, Deng X, Sasaki T, Mizuta K,
analysis. J Cell Biochem 118:3171–3183. https://doi.org/10.1002/jcb Kubo H, Nagatomi R. 2012. Levofloxacin inhibits rhinovirus infection in
.25951. primary cultures of human tracheal epithelial cells. Antimicrob Agents
57. Copeland V, McLaughlin M, Trifilio S. 2018. Ciprofloxacin vs levofloxacin Chemother 56:4052–4061. https://doi.org/10.1128/AAC.00259-12.
for prophylaxis during hematopoietic stem-cell transplantation. Clin 76. Dryden C, Wilkinson J, Young D, Brooker RJ, Scottish Paediatric Cystic
Transplant 32:e13145. https://doi.org/10.1111/ctr.13145. Fibrosis Managed Clinical Network. 2018. The impact of 12 months
58. Freifeld AG, Bow EJ, Sepkowitz KA, Boeckh MJ, Ito JI, Mullen CA, Raad treatment with ivacaftor on Scottish paediatric patients with cystic
II, Rolston KV, Young JA, Wingard JR. 2011. Clinical practice guideline fibrosis with the G551D mutation: a review. Arch Dis Child 103:68 –70.
for the use of antimicrobial agents in neutropenic patients with cancer: https://doi.org/10.1136/archdischild-2015-310420.
2010 update by the Infectious Diseases Society of America. Clin Infect 77. Psichogiou M, Poulakou G, Basoulis D, Paraskevis D, Markogiannakis A,
Dis 52:e56 – e93. https://doi.org/10.1093/cid/cir073. Daikos GL. 2017. Recent advances in antiretroviral agents: potent inte-
59. Haripriya A, Chang DF, Ravindran RD. 2017. Endophthalmitis reduction grase inhibitors. Curr Pharm Des 23:2552–2567. https://doi.org/10
with intracameral moxifloxacin prophylaxis: analysis of 600 000 surgeries. .2174/1381612823666170329142059.
Ophthalmology 124:768 –775. https://doi.org/10.1016/j.ophtha.2017.01 78. Sato M, Motomura T, Aramaki H, Matsuda T, Yamashita M, Ito Y,
.026. Kawakami H, Matsuzaki Y, Watanabe W, Yamataka K, Ikeda S, Kodama
60. Gustinetti G, Mikulska M. 2016. Bloodstream infections in neutropenic E, Matsuoka M, Shinkai H. 2006. Novel HIV-1 integrase inhibitors de-
cancer patients: a practical update. Virulence 7:280 –297. https://doi rived from quinolone antibiotics. J Med Chem 49:1506 –1508. https://
.org/10.1080/21505594.2016.1156821. doi.org/10.1021/jm0600139.
61. Humar A, Gill J, Johnston O, Fergusson D, House AA, Lebel L, Cockfield 79. Kan JY, Hsu YL, Chen YH, Chen TC, Wang JY, Kuo PL. 2013. Gemifloxa-
S, Kim SJ, Zaltzman J, Cantarovich M, Karpinski M, Ramsay T, Knoll GA. cin, a fluoroquinolone antimicrobial drug, inhibits migration and inva-
2013. Quinolone prophylaxis for the prevention of BK virus infection in sion of human colon cancer cells. Biomed Res Int 2013:159786. https://
kidney transplantation: study protocol for a randomized controlled doi.org/10.1155/2013/159786.
trial. Trials 14:185. https://doi.org/10.1186/1745-6215-14-185. 80. Sayar H, Bashardoust P. 2017. Therapies for acute myeloid leukemia:
62. Thevenot T, Degand T, Grelat N, Elkrief L, Christol C, Moreau R, Henrion vosaroxin. Onco Targets Ther 10:3957–3963. https://doi.org/10.2147/
J, Cadranel JF, Sheppard F, Bureau C, di Martino V, Pauwels A, National OTT.S121477.
Association of General Hospital Hepatogastroenterologists. 2013. A 81. Acar JF, Goldstein FW. 1997. Trends in bacterial resistance to fluoro-
French national survey on the use of antibiotic prophylaxis in cirrhotic quinolones. Clin Infect Dis 24(Suppl 1):S67–S73. https://doi.org/10
patients. Liver Int 33:389 –397. https://doi.org/10.1111/liv.12093. .1093/clinids/24.Supplement_1.S67.
63. Knoll G, Humar A, Fergusson D, Johnston O, House AA, Kim SJ, Ramsay 82. Castanon JIR. 2007. History of the use of antibiotic as growth promoters
T, Chassé M, Pang X, Zaltzman J, Cockfield S, Cantarovich M, Karpinski in European poultry feeds. Poult Sci 86:2466 –2471. https://doi.org/10
M, Lebel L, Gill JS. 2014. Levofloxacin for BK virus prophylaxis following .3382/ps.2007-00249.
kidney transplantation: a randomized clinical trial. JAMA 312: 83. Anonymous. 2005. Ban on antibiotics as growth promoters in animal
2106 –2114. https://doi.org/10.1001/jama.2014.14721. feed enters into effect, European Commission—IP/05/1687. European
64. Mikulska M, Cordonnier C. 2018. Fluoroquinolone prophylaxis during Commission, Brussels, Belgium. http://europa.eu/rapid/press-release_IP
neutropenia: what can we expect nowadays? Clin Microbiol Infect -05-1687_en.htm.
24:678 – 679. https://doi.org/10.1016/j.cmi.2018.02.031. 84. Om C, McLaws ML. 2016. Antibiotics: practice and opinions of Cambo-
dian commercial farmers, animal feed retailers and veterinarians. Anti- sitivity reactions to fluoroquinolones: analysis of the factors involved.
microb Resist Infect Control 5:42. https://doi.org/10.1186/s13756-016 Clin Exp Allergy 43:560 –567. https://doi.org/10.1111/cea.12099.
-0147-y. 107. Sachs B, Riegel S, Seebeck J, Beier R, Schichler D, Barger A, Merk HF,
85. Trouchon T, Lefebvre S. 2016. A review of enrofloxacin for veterinary use. Erdmann S. 2006. Fluoroquinolone-associated anaphylaxis in spon-
Open J Vet Med 6:40–58. https://doi.org/10.4236/ojvm.2016.62006. taneous adverse drug reaction reports in Germany: differences in
86. De Sarro A, De Sarro G. 2001. Adverse reactions to fluoroquinolones. An reporting rates between individual fluoroquinolones and occur-
overview on mechanistic aspects. Curr Med Chem 8:371–384. https:// rence after first-ever use. Drug Saf 229:1087–1100. https://doi.org/
doi.org/10.2174/0929867013373435. 10.2165/00002018-200629110-00008.
87. Owens RC, Jr, Ambrose PG. 2005. Antimicrobial safety: focus on fluo- 108. Peters B, Pinching AJ. 1989. Fatal anaphylaxis associated with cipro-
roquinolones. Clin Infect Dis 41(Suppl 2):S144 –S157. https://doi.org/10 floxacin in a patient with AIDS related complex. BMJ 298:605. https://
.1086/428055. doi.org/10.1136/bmj.298.6673.605-b.
88. Grady RW. 2005. Systemic quinolone antibiotics in children: a review of 109. Jespersen HG, Stenderup A. 1965. Treatment of urinary tract infections
the use and safety. Expert Opin Drug Saf 4:623– 630. https://doi.org/10 in the aged with nalidixic acid. Gerontol Clin 7:303–310. https://doi.org/
.1517/14740338.4.4.623. 10.1159/000244913.
89. Dawe RS, Ibbotson SH, Sanderson JB, Thomson EM, Ferguson J. 2003. 110. Barry AL, Jones RN, Thornsberry C, Ayers LW, Gerlach EH, Sommers HM.
A randomized controlled trial (volunteer study) of sitafloxacin, enoxa- 1984. Antibacterial activities of ciprofloxacin, norfloxacin, oxolinic acid,
cin, levofloxacin and sparfloxacin phototoxicity. Br J Dermatol 149: cinoxacin, and nalidixic acid. Antimicrob Agents Chemother 25:
1232–1241. https://doi.org/10.1111/j.1365-2133.2003.05582.x. 633– 637. https://doi.org/10.1128/aac.25.5.633.
90. Rubinstein E. 2001. History of quinolones and their side effects. Che- 111. Hoge CW, Gambel JM, Srijan A, Pitarangsi C, Echeverria P. 1998. Trends
motherapy 47(Suppl 3):3– 8. https://doi.org/10.1159/000057838. in antibiotic resistance among diarrheal pathogens isolated in Thailand
91. Thamlikitkul V, Tiengrim S. 2014. In vitro susceptibility test of sitafloxa- over 15 years. Clin Infect Dis 26:341–345. https://doi.org/10.1086/
cin against resistant gram-negative bacilli isolated from Thai patients 516303.
by disk diffusion method. J Med Assoc Thai 97(Suppl 3):S7–S12. 112. Rudrik JT, Cavalieri SJ, Britt EM. 1984. In vitro activities of enoxacin and
92. Lipsky BA, Baker CA. 1999. Fluoroquinolone toxicity profiles: a review 17 other antimicrobial agents against multiply resistant, gram-negative
focusing on newer agents. Clin Infect Dis 28:352–364. https://doi.org/ bacteria. Antimicrob Agents Chemother 26:97–100. https://doi.org/10
10.1086/515104. .1128/aac.26.1.97.
93. Kaguelidou F, Turner MA, Choonara I, Jacqz-Aigrain E. 2011. Ciprofloxa- 113. Dalhoff A. 2012. Global fluoroquinolone resistance epidemiology and
cin use in neonates: a systematic review of the literature. Pediatr Infect implications for clinical use. Interdiscip Perspect Infect Dis 2012:976273.
Dis J 30:e29 – e37. https://doi.org/10.1097/INF.0b013e3181fe353d. https://doi.org/10.1155/2012/976273.
94. Okoye NV, Oyawole MR, Uzochukwu PU, Oyetunde OO. 2013. Review of 114. Jones RN. 1996. Impact of changing pathogens and antimicrobial
ciprofloxacin use in children. Nig Q J Hosp Med 23:43– 47. susceptibility patterns in the treatment of serious infections in hospi-
95. Patel K, Goldman JL. 2016. Safety concerns surrounding quinolone use talized patients. Am J Med 100(6A):3S–12S. https://doi.org/10.1016/
in children. J Clin Pharmacol 56:1060 –1075. https://doi.org/10.1002/ s0002-9343(96)00102-7.
jcph.715. 115. Navia MM, Ruiz J, Ribera A, Jiménez de Anta MT, Vila J. 1999. Analysis
96. Principi N, Esposito S. 2015. Appropriate use of fluoroquinolones in of the mechanisms of quinolone resistance in clinical isolates of Citro-
children. Int J Antimicrob Agents 45:341–346. https://doi.org/10.1016/ bacter freundii. J Antimicrob Chemother 44:743–748. https://doi.org/10
j.ijantimicag.2015.01.004. .1093/jac/44.6.743.
97. Ouchi K, Sunakawa K. 2014. Effect of new oral antimicrobial agents in 116. Rautelin H, Renkonen OV, Kosunen TU. 1991. Emergence of fluoroquin-
outpatient treatment of pneumonia in children. Jpn J Antibiot 67: olone resistance in Campylobacter jejuni and Campylobacter coli in
157–166. subjects from Finland. Antimicrob Agents Chemother 35:2065–2069.
98. Tanaka E, Hara N, Wajima T, Ochiai S, Seyama S, Shirai A, Shibata M, https://doi.org/10.1128/aac.35.10.2065.
Shiro H, Natsume Y, Noguchi N. 2019. Emergence of Haemophilus 117. Reina J, Ros MJ, Muñoz-López A. 1996. Aislamiento de una cepa de
influenzae with low susceptibility to quinolones and persistence in Campylobacter jejuni con resistencia a las fluoroquinolonas y sensibili-
tosufloxacin treatment. J Glob Antimicrob Resist 18:104 –108. https:// dad al ácido nalídixico. Enferm Infecc Microbiol Clin 14:124 –125.
doi.org/10.1016/j.jgar.2019.01.017. 118. Ruiz J, Castro D, Goñi P, Santamaria JA, Borrego JJ, Vila J. 1997. Analysis
99. Ziv A, Masarwa R, Perlman A, Ziv D, Matok I. 2018. Pregnancy outcomes of the mechanism of quinolone-resistance in nalidixic acid-resistant
following exposure to quinolone antibiotics—a systematic-review and clinical isolates of Salmonella serotype Typhimurium. J Med Microbiol
meta-analysis. Pharm Res 35:109. https://doi.org/10.1007/s11095-018 46:623– 628. https://doi.org/10.1099/00222615-46-7-623.
-2383-8. 119. Ruiz J, Marco F, Goñi P, Gallardo F, Mensa J, Trilla A, Jiménez de Anta
100. Frankel WC, Trautner BW, Spiegelman A, Grigoryan L, LeMaire SA. 2019. T, Vila J. 1995. High frequency of mutations at codon 83 of the gyrA
Patients at risk for aortic rupture often exposed to fluoroquinolones gene of quinolone-resistant clinical isolates of Escherichia coli. J Anti-
during hospitalization. Antimicrob Agents Chemother 63:e01712-18. microb Chemother 36:737–738. https://doi.org/10.1093/jac/36.4.737.
https://doi.org/10.1128/AAC.01712-18. 120. Endtz HP, Ruijs GJ, van Klingeren B, Jansen WH, van der Reyden T,
101. Pasternak B, Inghammar M, Svanström H. 2018. Fluoroquinolone use Mouton RP. 1991. Quinolone resistance in Campylobacter isolated from
and risk of aortic aneurysm and dissection: nationwide cohort study. man and poultry following the introduction of fluoroquinolones in
BMJ 360:k678. https://doi.org/10.1136/bmj.k678. veterinary medicine. J Antimicrob Chemother 27:199 –208. https://doi
102. LeMaire SA, Zhang L, Luo W, Ren P, Azares AR, Wang Y, Zhang C, Coselli .org/10.1093/jac/27.2.199.
JS, Shen YH. 2018. Effect of ciprofloxacin on susceptibility to aortic 121. Mirelis B, Miro E, Navarro F, Ogalla CA, Bonal J, Prats G. 1993. Increased
dissection and rupture in mice. JAMA Surg 153:e181804. https://doi resistance to quinolone in Catalonia, Spain. Diagn Microbiol Infect Dis
.org/10.1001/jamasurg.2018.1804. 16:137–139. https://doi.org/10.1016/0732-8893(93)90009-V.
103. Manfredi M, Severino M, Testi S, Macchia D, Ermini G, Pichler WJ, Campi 122. Ruiz J, Goñi P, Marco F, Gallardo F, Mirelis B, Jiménez de Anta T, Vila J.
P. 2004. Detection of specific IgE to quinolones. J Allergy Clin Immunol 1998. Increased resistance to quinolones in Campylobacter jejuni: a
113:155–160. https://doi.org/10.1016/j.jaci.2003.09.035. genetic analysis of gyrA gene mutations in quinolone-resistant clinical
104. Doña I, Blanca-López N, Torres MJ, García-Campos J, García-Núñez I, isolates. Microbiol Immunol 42:223–226. https://doi.org/10.1111/j.1348
Gómez F, Salas M, Rondón C, Canto MG, Blanca M. 2012. Drug hyper- -0421.1998.tb02274.x.
sensitivity reactions: response patterns, drug involved, and temporal 123. Deguchi T, Saito I, Tanaka M, Sato K, Deguchi K, Yasuda M, Nakano M,
variations in a large series of patients. J Investig Allergol Clin Immunol Nishino Y, Kanematsu E, Ozeki S, Kawada Y. 1997. Fluoroquinolone
22:363–371. https://doi.org/10.18176/jiaci.0078. treatment failure in gonorrhea. Emergence of a Neisseria gonorrhoeae
105. Aranda A, Mayorga C, Ariza A, Doña I, Rosado A, Blanca-Lopez N, strain with enhanced resistance to fluoroquinolones. Sex Transm Dis
Andreu I, Torres MJ. 2011. In vitro evaluation of IgE-mediated hyper- 24:247–250. https://doi.org/10.1097/00007435-199705000-00002.
sensitivity reactions to quinolones. Allergy 66:247–254. https://doi.org/ 124. Le Lostec Z, Fegueux S, Jouve P, Cheron M, Mornet P, Boisivon A. 1997.
10.1111/j.1398-9995.2010.02460.x. Reduced susceptibility to quinolones in Salmonella typhi acquired in
106. Blanca-López N, Ariza A, Doña I, Mayorga C, Montañez MI, Garcia- Europe: a clinical failure of treatment. Clin Microbiol Infect 3:576 –577.
Campos J, Gomez F, Rondón C, Blanca M, Torres MJ. 2013. Hypersen- https://doi.org/10.1111/j.1469-0691.1997.tb00313.x.
125. Vasallo FJ, Martín-Rabadán P, Alcalá L, García-Lechuz JM, Rodríguez- pressure: the experience of a very remote community in the Amazon
Créixems M, Bouza E. 1998. Failure of ciprofloxacin therapy for invasive forest. PLoS Negl Trop Dis 6:e1790. https://doi.org/10.1371/journal
nontyphoidal salmonellosis. Clin Infect Dis 126:535–536. https://doi .pntd.0001790.
.org/10.1086/517087. 143. Pons MJ, Vubil D, Guiral E, Jaintilal D, Fraile O, Soto SM, Sigauque B,
126. Vila J, Olmos L, Ballesteros J, Vázquez JA, Giménez MJ, Marco F, Aguilar Nhampossa T, Aide P, Alonso PL, Vila J, Mandomando I, Ruiz J. 2015.
L. 1997. Development of in-vivo resistance after quinolone treatment of Characterisation of extended spectrum -lactamases among Klebsiella
gonococcal urethritis. J Antimicrob Chemother 39:841. https://doi.org/ pneumoniae isolates causing bacteraemia and urinary tract infection in
10.1093/jac/39.6.841a. Mozambique. J Glob Antimicrob Resist 3:19 –25. https://doi.org/10
127. Vila J, Ruiz J, Sanchez F, Navarro F, Mirelis B, Jiménez de Anta MT, Prats .1016/j.jgar.2015.01.004.
G. 1999. Increase in quinolone resistance in a Haemophilus influenzae 144. Ahmed A, Bashir T. 2015. Trends in quinolone resistance among com-
strain isolated from a patient with recurrent respiratory infections mon urinary tract isolates over three years. J Coll Physicians Surg Pak
treated with ofloxacin. Antimicrob Agents Chemother 43:161–162. 25:543–545.
https://doi.org/10.1128/AAC.43.1.161. 145. Lahra MM, Australian Gonococcal Surveillance Programme. 2015. Aus-
128. Llor C, Bjerrum L. 2014. Antimicrobial resistance: risk associated with tralian Gonococcal Surveillance Programme annual report, 2014. Com-
antibiotic overuse and initiatives to reduce the problem. Ther Adv Drug mun Dis Intell Q Rep 39:E347–E354.
Saf 5:229 –241. https://doi.org/10.1177/2042098614554919. 146. Lluque A, Riveros M, Prada A, Ochoa TJ, Ruiz J. 2017. Campylobacter
129. Ruiz J, Pons MJ. 2013. Prevention of travellers’ diarrhoea: where and spp. from a Peruvian pediatric cohort. Scientifica (Cairo) 2017:7848926.
who? Lancet Infect Dis 13:911–912. https://doi.org/10.1016/S1473 https://doi.org/10.1155/2017/7848926.
-3099(13)70243-3. 147. Palma N, Pons MJ, Gomes C, Mateu J, Riveros M, García W, Jacobs J,
130. US Centers for Diseases Control and Prevention. 2013. Antibiotic García C, Ochoa TJ, Ruiz J. 2017. Resistance to quinolones, cephalospo-
resistance threats in the United States, 2013. US Department of rins and macrolides in Escherichia coli causing bacteremia in Peruvian
Health and Human Services, Centers for Disease Control and Pre- children. J Glob Antimicrob Resist 11:28 –33. https://doi.org/10.1016/j
vention, Atlanta, GA. .jgar.2017.06.011.
131. Gavaldà L, Soriano AM, Cámara J, Gasull R, Arch O, Ferrer M, Shaw E, 148. Sáenz Y, Ruiz J, Zarazaga M, Teixidó M, Torres C, Vila J. 2004. Effect of
Granada RM, Dominguez MA, Pujol M. 2016. Control of endemic ex- the efflux pump inhibitor Phe-Arg--naphthylamide on the MIC values
tensively drug-resistant Acinetobacter baumannii with a cohorting pol- of the quinolones, tetracycline and chloramphenicol, in Escherichia coli
icy and cleaning procedures based on the 1 room, 1 wipe approach. isolates of different origin. J Antimicrob Chemother 53:544 –545.
Am J Infect Control 44:520 –524. https://doi.org/10.1016/j.ajic.2015.11 https://doi.org/10.1093/jac/dkh117.
.036. 149. Ince D, Hooper DC. 2003. Quinolone resistance due to reduced target
132. Saam M, Huttner B, Harbarth S. 2017. Evaluation of antibiotic awareness enzyme expression. J Bacteriol 185:6883–6892. https://doi.org/10.1128/JB
campaigns. World Health Organization, Geneva, Switzerland. .185.23.6883-6892.2003.
133. de Toro M, Rojo-Bezares B, Vinué L, Undabeitia E, Torres C, Sáenz Y.
150. Abdi-Ali A, Mohammadi-Mehr M, Agha Alaei Y. 2006. Bactericidal activity
2010. In vivo selection of aac(6=)-Ib-cr and mutations in the gyrA gene
of various antibiotics against biofilm-producing Pseudomonas aeruginosa.
in a clinical qnrS1-positive Salmonella enterica serovar Typhimurium
Int J Antimicrob Agents 27:196–200. https://doi.org/10.1016/j.ijantimicag
DT104B strain recovered after fluoroquinolone treatment. J Antimicrob
.2005.10.007.
Chemother 65:1945–1949. https://doi.org/10.1093/jac/dkq262.
151. Blango MG, Mulvey MA. 2010. Persistence of uropathogenic Escherichia
134. Gao L, Lyu Y, Li Y. 2017. Trends in drug resistance of Acinetobacter
coli in the face of multiple antibiotics. Antimicrob Agents Chemother
baumannii over a 10-year period: nationwide data from the China
54:1855–1863. https://doi.org/10.1128/AAC.00014-10.
surveillance of antimicrobial resistance program. Chin Med J 130:
152. Crumplin GC, Smith JT. 1975. Nalidixic acid: an antibacterial paradox.
659 – 664. https://doi.org/10.4103/0366-6999.201601.
Antimicrob Agents Chemother 8:251–261. https://doi.org/10.1128/aac
135. Horna G, Quezada K, Ramos S, Mosqueda N, Rubio M, Guerra H, Ruiz J.
.8.3.251.
2019. Specific type IV pili groups in clinical isolates of Pseudomonas
153. Robicsek A, Strahilevitz J, Jacoby GA, Macielag M, Abbanat D, Park CH,
aeruginosa. Int Microbiol 22:131–141. https://doi.org/10.1007/s10123
Bush K, Hooper DC. 2006. Fluoroquinolone-modifying enzyme: a new
-018-00035-3.
136. Mendez Arancibia E, Pitart C, Ruiz J, Marco F, Gascón J, Vila J. 2009. adaptation of a common aminoglycoside acetyltransferase. Nat Med
Evolution of antimicrobial resistance in enteroaggregative Esche- 12:83– 88. https://doi.org/10.1038/nm1347.
richia coli and enterotoxigenic Escherichia coli causing traveller’s 154. Chávez-Jacobo VM, Hernández-Ramírez KC, Romo-Rodríguez P, Pérez-
diarrhoea. J Antimicrob Chemother 64:343–347. https://doi.org/10 Gallardo RV, Campos-García J, Gutiérrez-Corona JF, García-Merinos JP,
.1093/jac/dkp178. Meza-Carmen V, Silva-Sánchez J, Ramírez-Díaz MI. 2018. CrpP is a novel
137. Pons MJ, Gomes C, Martínez-Puchol S, Ruiz L, Mensa L, Vila J, Gascón J, ciprofloxacin-modifying enzyme encoded by the Pseudomonas aerugi-
Ruiz J. 2013. Antimicrobial resistance in Shigella spp. causing traveller’s nosa pUM505 plasmid. Antimicrob Agents Chemother 62:e02629-17.
diarrhoea (1995-2010): a retrospective analysis. Travel Med Infect Dis https://doi.org/10.1128/AAC.02629-17.
5:315–319. https://doi.org/10.1016/j.tmaid.2013.06.010. 155. Sørensen AH, Hansen LH, Johannesen E, Sørensen SJ. 2003. Conjugative
138. Pons MJ, Mosquito S, Gomes C, del Valle LJ, Ochoa TJ, Ruiz J. 2014. plasmid conferring resistance to olaquindox. Antimicrob Agents Che-
Analysis of quinolone-resistance in commensal and diarrheagenic Esch- mother 47:798–799. https://doi.org/10.1128/AAC.47.2.798-799.2003.
erichia coli isolates from infants in Lima, Peru. Trans R Soc Trop Med 156. Hansen LH, Johannesen E, Burmølle M, Sørensen AH, Sørensen SJ. 2004.
Hyg 108:22–28. https://doi.org/10.1093/trstmh/trt106. Plasmid-encoded multidrug efflux pump conferring resistance to
139. Salazar de Vegas EZ, Nieves B, Ruiz M, Ruíz J, Vila J, Araque M, Velázco olaquindox in Escherichia coli. Antimicrob Agents Chemother 48:
E. 2007. Molecular epidemiology and characterization of resistance 3332–3337. https://doi.org/10.1128/AAC.48.9.3332-3337.2004.
mechanisms to various antimicrobial agents in Acinetobacter bauman- 157. Hansen LH, Jensen LB, Sørensen HI, Sørensen SJ. 2007. Substrate
nii isolated in Mérida, Venezuela. Med Sci Monit 13:BR89 –BR94. specificity of the OqxAB multidrug resistance pump in Escherichia coli
140. WHO Western Pacific Gonococcal Antimicrobial Surveillance Pro- and selected enteric bacteria. J Antimicrob Chemother 60:145–147.
gramme. 2006. Surveillance of antibiotic resistance in Neisseria gonor- https://doi.org/10.1093/jac/dkm167.
rhoeae in the WHO Western Pacific Region, 2005. Commun Dis Intell Q 158. Yamane K, Wachino J, Suzuki S, Kimura K, Shibata N, Kato H, Shibayama
Rep 30:430 – 433. K, Konda T, Arakawa Y. 2007. New plasmid-mediated fluoroquinolone
141. Guiral E, Pons MJ, Vubil D, Marí M, Sigauque B, Soto SM, Alonso PL, Ruiz efflux pump, QepA, found in an Escherichia coli clinical isolate. Antimi-
J, Vila J, Mandomando I. 2018. Epidemiology and molecular character- crob Agents Chemother 51:3354 –3360. https://doi.org/10.1128/AAC
ization of multidrug-resistant Escherichia coli isolates harboring blaCTX-M .00339-07.
group 1 extended-spectrum -lactamases causing bacteremia and uri- 159. Périchon B, Courvalin P, Galimand M. 2007. Transferable resistance to
nary tract infection in Manhiça, Mozambique. Infect Drug Resist 11: aminoglycosides by methylation of G1405 in 16S rRNA and to hydro-
927–936. https://doi.org/10.2147/IDR.S153601. philic fluoroquinolones by QepA-mediated efflux in Escherichia coli.
142. Pallecchi L, Bartoloni A, Riccobono E, Fernandez C, Mantella A, Magnelli Antimicrob Agents Chemother 51:2464 –2469. https://doi.org/10.1128/
D, Mannini D, Strohmeyer M, Bartalesi F, Rodriguez H, Gotuzzo E, AAC.00143-07.
Rossolini GM. 2012. Quinolone resistance in absence of selective 160. Ruiz J, Pons MJ, Gomes C. 2012. Transferable mechanisms of quinolone
resistance. Int J Antimicrob Agents 40:196 –203. https://doi.org/10 179. Ruiz J. 2018. In silico analysis of transferable QepA variants and related
.1016/j.ijantimicag.2012.02.011. chromosomal efflux pumps. Rev Esp Quimioter 31:537–541.
161. Aviv G, Rahav G, Gal-Mor O. 2016. Horizontal transfer of the Salmonella 180. Lyon BR, Skurray R. 1987. Antimicrobial resistance of Staphylococcus
enterica serovar Infantis resistance and virulence plasmid pESI to the aureus: genetic basis. Microbiol Rev 51:88 –134.
gut microbiota of warm-blooded hosts. mBio 7:e01395-16. https://doi 181. Andersen JL, He GX, Kakarla P, KC R, Kumar S, Lakra WS, Mukherjee MM,
.org/10.1128/mBio.01395-16. Ranaweera I, Shrestha U, Tran T, Varela MF. 2015. Multidrug efflux
162. Courvalin P. 1994. Transfer of antibiotic resistance genes between pumps from Enterobacteriaceae, Vibrio cholerae and Staphylococcus
gram-positive and gram-negative bacteria. Antimicrob Agents Che- aureus bacterial food pathogens. Int J Environ Res Public Health 12:
mother 38:1447–1451. https://doi.org/10.1128/aac.38.7.1447. 1487–1547. https://doi.org/10.3390/ijerph120201487.
163. Alonso A, Sanchez P, Martínez JL. 2000. Stenotrophomonas maltophilia 182. Ramírez MS, Tolmasky ME. 2010. Aminoglycoside modifying enzymes.
D457R contains a cluster of genes from gram-positive bacteria involved Drug Resist Updat 13:151–171. https://doi.org/10.1016/j.drup.2010.08
in antibiotic and heavy metal resistance. Antimicrob Agents Chemother .003.
44:1778 –1782. https://doi.org/10.1128/AAC.44.7.1778-1782.2000. 183. Foster TJ, Howe TG, Richmond KM. 1975. Translocation of the tetracy-
164. Ribera A, Ruiz J, Vila J. 2003. Presence of the Tet M determinant in a clinical cline resistance determinant from R100-1 to the Escherichia coli K-12
isolate of Acinetobacter baumannii. Antimicrob Agents Chemother 47: chromosome. J Bacteriol 124:1153–1158.
2310–2312. https://doi.org/10.1128/AAC.47.7.2310-2312.2003. 184. Rychlik I, Gregorova D, Hradecka H. 2006. Distribution and function of
165. Gomes C, Martínez-Puchol S, Palma N, Horna G, Ruiz-Roldán L, Pons MJ, plasmids in Salmonella enterica. Vet Microbiol 112:1–10. https://doi.org/
Ruiz J. 2017. Macrolide resistance mechanisms in Enterobacteriaceae: 10.1016/j.vetmic.2005.10.030.
focus in azithromycin. Crit Rev Microbiol 43:1–30. https://doi.org/10 185. Coelho A, González-López JJ, Miró E, Alonso-Tarrés C, Mirelis B, Larrosa
.3109/1040841X.2015.1136261. MN, Bartolomé RM, Andreu A, Navarro F, Johnson JR, Prats G. 2010.
166. Nakaminami H, Noguchi N, Sasatsu M. 2010. Fluoroquinolone efflux by Characterisation of the CTX-M-15-encoding gene in Klebsiella pneu-
the plasmid-mediated multidrug efflux pump QacB variant QacBIII in moniae strains from the Barcelona metropolitan area: plasmid diversity
Staphylococcus aureus. Antimicrob Agents Chemother 54:4107– 4111. and chromosomal integration. Int J Antimicrob Agents 36:73–78.
https://doi.org/10.1128/AAC.01065-09. https://doi.org/10.1016/j.ijantimicag.2010.03.005.
167. Osińska A, Harnisz M, Korzeniewska E. 2016. Prevalence of plasmid- 186. Fabre L, Delauné A, Espié E, Nygard K, Pardos M, Polomack L, Guesnier
mediated multidrug resistance determinants in fluoroquinolone-resistant F, Galimand M, Lassen J, Weill FX. 2009. Chromosomal integration of
bacteria isolated from sewage and surface water. Environ Sci Pollut Res Int the extended-spectrum -lactamase gene blaCTX-M-15 in Salmonella
23:10818–10831. https://doi.org/10.1007/s11356-016-6221-4. enterica serotype Concord isolates from internationally adopted chil-
168. Laport MS, Pontes PVM, Dos Santos DS, Santos-Gandelman JDF, Muricy dren. Antimicrob Agents Chemother 53:1808 –1816. https://doi.org/10
G, Bauwens M, Giambiagi-deMarval M, George I. 2016. Antibiotic resis- .1128/AAC.00451-08.
tance genes detected in the marine sponge Petromica citrina from 187. Ferreira JC, Penha Filho RAC, Andrade LN, Berchieri A, Jr, Darini ALC.
Brazilian coast. Braz J Microbiol 47:617– 620. https://doi.org/10.1016/j
2014. Detection of chromosomal blaCTX-M-2 in diverse Escherichia coli
.bjm.2016.04.016.
isolates from healthy broiler chickens. Clin Microbiol Infect 20:
169. Yuan L, Zhai YJ, Wu H, Sun HR, He ZP, Wang YB, Pan YS, Kuang NN, Hu
O623–O626. https://doi.org/10.1111/1469-0691.12531.
GZ. 2018. Identification and prevalence of RND family multidrug efflux
188. Hamamoto K, Ueda S, Toyosato T, Yamamoto Y, Hirai I. 2016. High
pump oqxAB genes in Enterococci isolates from swine manure in China.
prevalence of chromosomal blaCTX-M-14 in Escherichia coli isolates pos-
J Med Microbiol 67:733–739. https://doi.org/10.1099/jmm.0.000736.
sessing blaCTX-M-14. Antimicrob Agents Chemother 60:2582–2584.
170. Robicsek A, Jacoby GA, Hooper DC. 2006. The worldwide emergence of
https://doi.org/10.1128/AAC.00108-16.
plasmid-mediated quinolone resistance. Lancet Infect Dis 6:629 – 640.
189. Harada S, Ishii Y, Saga T, Kouyama Y, Tateda K, Yamaguchi K. 2012.
https://doi.org/10.1016/S1473-3099(06)70599-0.
Chromosomal integration and location on IncT plasmids of the blaCTX-
171. Hata M, Suzuki M, Matsumoto M, Takahashi M, Sato K, Ibe S, Sakae K.
M-2 gene in Proteus mirabilis clinical isolates. Antimicrob Agents Che-
2005. Cloning of a novel gene for quinolone resistance from a trans-
mother 56:1093–1096. https://doi.org/10.1128/AAC.00258-11.
ferable plasmid in Shigella flexneri 2b. Antimicrob Agents Chemother
190. Hirai I, Fukui N, Taguchi M, Yamauchi K, Nakamura T, Okano S,
49:801– 803. https://doi.org/10.1128/AAC.49.2.801-803.2005.
172. Poirel L, Rodríguez-Martínez JM, Mammeri H, Liard A, Nordmann P. Yamamoto Y. 2013. Detection of chromosomal blaCTX-M-15 in Escherichia
2005. Origin of plasmid-mediated quinolone resistance determinant coli O25b-B2-ST131 isolates from the Kinki region of Japan. Int J
QnrA. Antimicrob Agents Chemother 49:3523–3525. https://doi.org/10 Antimicrob Agents 42:500 –506. https://doi.org/10.1016/j.ijantimicag
.1128/AAC.49.8.3523-3525.2005. .2013.08.005.
173. Robicsek A, Sahm DF, Strahilevitz J, Jacoby GA, Hooper DC. 2005. 191. Huang W, Wang G, Sebra R, Zhuge J, Yin C, Aguero-Rosenfeld ME,
Broader distribution of plasmid-mediated quinolone resistance in the Schuetz AN, Dimitrova N, Fallon JT. 2017. Emergence and evolution of
United States. Antimicrob Agents Chemother 49:3001–3003. https://doi multidrug-resistant Klebsiella pneumoniae with both blaKPC and blaCTX-M
.org/10.1128/AAC.49.7.3001-3003.2005. integrated in the chromosome. Antimicrob Agents Chemother 61:
174. Saga T, Kaku M, Onodera Y, Yamachika S, Sato K, Takase H. 2005. Vibrio e00076-17. https://doi.org/10.1128/AAC.00076-17.
parahaemolyticus chromosomal qnr homologue VPA0095: demonstration 192. Mahrouki S, Belhadj O, Chihi H, Mohamed BM, Celenza G, Amicosante
by transformation with a mutated gene of its potential to reduce quino- G, Perilli M. 2012. Chromosomal blaCTX-M-15 associated with ISEcp1 in
lone susceptibility in Escherichia coli. Antimicrob Agents Chemother 49: Proteus mirabilis and Morganella morganii isolated at the Military Hos-
2144–2145. https://doi.org/10.1128/AAC.49.5.2144-2145.2005. pital of Tunis, Tunisia. J Med Microbiol 61(Part 9):1286 –1289. https://
175. Fonseca EL, Dos Santos Freitas F, Vieira VV, Vicente ACP. 2008. New qnr doi.org/10.1099/jmm.0.039487-0.
gene cassettes associated with superintegron repeats in Vibrio cholerae 193. Castanheira M, Deshpande LM, DiPersio JR, Kang J, Weinstein MP, Jones
01. Emerg Infect Dis 14:1129 –1131. https://doi.org/10.3201/eid1407 RN. 2009. First descriptions of blaKPC in Raoultella spp. (R. planticola and
.080132. R. ornithinolytica): report from the SENTRY Antimicrobial Surveillance
176. Albornoz E, Tijet N, De Belder D, Gomez S, Martino F, Corso A, Melano Program. J Clin Microbiol 47:4129 – 4130. https://doi.org/10.1128/JCM
RG, Petroni A. 2017. qnrE1, a member of a new family of plasmid- .01502-09.
located quinolone resistance genes, originated from the chromosome 194. Villegas MV, Lolans K, Correa A, Kattan JN, Lopez JA, Quinn JP, Colom-
of Enterobacter species. Antimicrob Agents Chemother 61:e02555-16. bian Nosocomial Resistance Study Group. 2007. First identification of
https://doi.org/10.1128/AAC.02555-16. Pseudomonas aeruginosa isolates producing a KPC-type carbapenem-
177. Yuan J, Xu X, Guo Q, Zhao X, Ye X, Guo Y, Wang M. 2012. Prevalence of hydrolyzing  -lactamase. Antimicrob Agents Chemother 51:
the oqxAB gene complex in Klebsiella pneumoniae and Escherichia coli 1553–1555. https://doi.org/10.1128/AAC.01405-06.
clinical isolates. J Antimicrob Chemother 67:1655–1659. https://doi 195. Martínez T, Vázquez GJ, Aquino EE, Martínez I, Robledo IE. 2014. ISEcp1-
.org/10.1093/jac/dks086. mediated transposition of blaKPC into the chromosome of a clinical isolate
178. Cattoir V, Poirel L, Nordmann P. 2008. Plasmid-mediated quinolone of Acinetobacter baumannii from Puerto Rico. J Med Microbiol 63(Part
resistance pump QepA2 in an Escherichia coli isolate from France. 12):1644–1648. https://doi.org/10.1099/jmm.0.080721-0.
Antimicrob Agents Chemother 52:3801–3804. https://doi.org/10.1128/ 196. Toleman MA, Bennett PM, Walsh TR. 2006. Common regions e.g. orf513
AAC.00638-08. and antibiotic resistance: IS91-like elements evolving complex class 1
integrons. J Antimicrob Chemother 58:1– 6. https://doi.org/10.1093/jac/ 214. Ribera A, Doménech-Sanchez A, Ruiz J, Benedi VJ, Jimenez de Anta MT,
dkl204. Vila J. 2002. Mutations in gyrA and parC QRDRs are not relevant for
197. Ruiz E, Sáenz Y, Zarazaga M, Rocha-Gracia R, Martínez-Martínez L, Arlet quinolone resistance in epidemiological unrelated Stenotrophomonas
G, Torres C. 2012. qnr, aac(6=)-Ib-cr and qepA genes in Escherichia coli maltophilia clinical isolates. Microb Drug Resist 8:245–252. https://doi
and Klebsiella spp.: genetic environments and plasmid and chromo- .org/10.1089/10766290260469499.
somal location. J Antimicrob Chemother 67:886 – 897. https://doi.org/ 215. Sánchez MB, Martínez JL. 2010. SmQnr contributes to intrinsic resis-
10.1093/jac/dkr548. tance to quinolones in Stenotrophomonas maltophilia. Antimicrob
198. Musumeci R, Rausa M, Giovannoni R, Cialdella A, Bramati S, Sibra B, Agents Chemother 54:580 –581. https://doi.org/10.1128/AAC.00496-09.
Giltri G, Viganò F, Cocuzza CE. 2012. Prevalence of plasmid-mediated 216. Valdezate S, Vindel A, Saéz-Nieto JA, Baquero F, Cantón R. 2005.
quinolone resistance genes in uropathogenic Escherichia coli isolated in Preservation of topoisomerase genetic sequences during in vivo and in
a teaching hospital of northern Italy. Microb Drug Resist 18:33– 41. vitro development of high-level resistance to ciprofloxacin in isogenic
https://doi.org/10.1089/mdr.2010.0146. Stenotrophomonas maltophilia strains. J Antimicrob Chemother 56:
199. Kim HB, Wang M, Park CH, Kim EC, Jacoby GA, Hooper DC. 2009. oqxAB 220 –223. https://doi.org/10.1093/jac/dki182.
encoding a multidrug efflux pump in human clinical isolates of Enter- 217. Cabrera R, Ruiz J, Marco F, Oliveira I, Arroyo M, Aladueña A, Usera MA,
obacteriaceae. Antimicrob Agents Chemother 53:3582–3584. https:// Jiménez De Anta MT, Gascón J, Vila J. 2004. Mechanism of resistance to
doi.org/10.1128/AAC.01574-08. several antimicrobial agents in Salmonella clinical isolates causing trav-
200. Norman A, Hansen LH, She Q, Sørensen SJ. 2008. Nucleotide sequence eler’s diarrhea. Antimicrob Agents Chemother 48:3934 –3939. https://
of pOLA52: a conjugative IncX1 plasmid from Escherichia coli which doi.org/10.1128/AAC.48.10.3934-3939.2004.
enables biofilm formation and multidrug efflux. Plasmid 60:59 –74. 218. Capilla S, Ruiz J, Goñi P, Castillo J, Rubio MC, Jiménez de Anta MT,
https://doi.org/10.1016/j.plasmid.2008.03.003. Gómez-Lus R, Vila J. 2004. Characterization of the molecular mecha-
201. Wong MHY, Chan EWC, Chen S. 2015. Evolution and dissemination of nisms of quinolone-resistance in Yersinia enterocolitica O:3 clinical iso-
OqxAB-like efflux pumps, an emerging quinolone resistance determi- lates. J Antimicrob Chemother 53:1068 –1071. https://doi.org/10.1093/
nant among members of Enterobacteriaceae. Antimicrob Agents Che- jac/dkh225.
mother 59:3290 –3297. https://doi.org/10.1128/AAC.00310-15. 219. Ruiz J, Gómez J, Navia MM, Ribera A, Sierra JM, Marco F, Mensa J, Vila
202. Wong MHY, Chen S. 2013. First detection of oqxAB in Salmonella spp. J. 2002. High prevalence of nalidixic acid resistant, ciprofloxacin sus-
isolated from food. Antimicrob Agents Chemother 57:658 – 660. https:// ceptible phenotype among clinical isolates of Escherichia coli and other
doi.org/10.1128/AAC.01144-12. Enterobacteriaceae. Diagn Microbiol Infect Dis 42:257–261. https://doi
203. Cavaco LM, Hansen DS, Friis-Møller A, Aarestrup FM, Hasman H, .org/10.1016/S0732-8893(01)00357-1.
Frimodt-Møller N. 2007. First detection of plasmid-mediated quinolone 220. Wain J, Hoa NT, Chinh NT, Vinh H, Everett MJ, Diep TS, Day NP,
resistance (qnrA and qnrS) in Escherichia coli strains isolated from Solomon T, White NJ, Piddock LJ, Parry CM. 1997. Quinolone-
humans in Scandinavia. J Antimicrob Chemother 59:804 – 805. https:// resistant Salmonella typhi in Viet Nam: molecular basis of resistance
doi.org/10.1093/jac/dkl554. and clinical response to treatment. Clin Infect Dis 25:1404 –1410.
204. Tomova A, Ivanova L, Buschmann AH, Godfrey HP, Cabello FC. 2018. https://doi.org/10.1086/516128.
Plasmid-mediated quinolone resistance (PMQR) genes and class 1 in- 221. CLSI. 2015. Performance standards for antimicrobial susceptibility
tegrons in quinolone-resistant marine bacteria and clinical isolates of testing; twenty-fifth informational supplement. Document M100-S25,
Escherichia coli from an aquacultural area. Microb Ecol 75:104 –112. 25th ed. CLSI, Wayne, PA.
https://doi.org/10.1007/s00248-017-1016-9. 222. CLSI. 2016. Performance standards for antimicrobial susceptibility test-
205. Veldman K, Kant A, Dierikx C, van Essen-Zandbergen A, Wit B, ing. Document M100S, 26th ed. CLSI, Wayne, PA.
Mevius D. 2014. Enterobacteriaceae resistant to third-generation 223. Hakanen A, Kotilainen P, Jalava J, Siitonen A, Huovinen P. 1999.
cephalosporins and quinolones in fresh culinary herbs imported Detection of decreased fluoroquinolone susceptibility in salmonel-
from Southeast Asia. Int J Food Microbiol 177:72–77. https://doi.org/ las and validation of nalidixic acid screening test. J Clin Microbiol
10.1016/j.ijfoodmicro.2014.02.014. 37:3572–3577.
206. Chen D, Yang L, Peters BM, Liu J, Li L, Li B, Xu Z, Shirtliff ME. 2019. 224. Angelakis E, Biswas S, Taylor C, Raoult D, Rolain JM. 2008. Heteroge-
Complete sequence of a novel multidrug-resistant Pseudomonas putida neity of susceptibility to fluoroquinolones in Bartonella isolates from
strain carrying two copies of qnrVC6. Microb Drug Resist 25:1–7. https:// Australia reveals a natural mutation in gyrA. J Antimicrob Chemother
doi.org/10.1089/mdr.2018.0104. 61:1252–1255. https://doi.org/10.1093/jac/dkn094.
207. Pérez-Moreno MO, Picó-Plana E, de Toro M, Grande-Armas J, Quiles- 225. del Valle LJ, Flores L, Vargas M, García-de-la-Guarda R, Quispe RL,
Fortuny V, Pons MJ, Gomes C, Sáenz Y, Torres C, Ruiz J. 2013. Ibañez ZB, Alvarado D, Ramírez P, Ruiz J. 2010. Bartonella bacilliformis,
-Lactamases, transferable quinolone resistance determinants, and endemic pathogen of the Andean region, is intrinsically resistant to
class 1 integron-mediated antimicrobial resistance in human clinical quinolones. Int J Infect Dis 14:e506 – e510. https://doi.org/10.1016/j.ijid
Salmonella enterica isolates of non-Typhimurium serotypes. Int J Med .2009.07.025.
Microbiol 303:25–31. https://doi.org/10.1016/j.ijmm.2012.11.003. 226. Han XY, Andrade RA. 2005. Brevundimonas diminuta infections and its
208. Zhao Z, Lan F, Liu M, Chen W, Huang L, Lin Q, Li B. 2017. Evaluation of resistance to fluoroquinolones. J Antimicrob Chemother 55:853– 859.
automated systems for aminoglycosides and fluoroquinolones suscep- https://doi.org/10.1093/jac/dki139.
tibility testing for carbapenem-resistant Enterobacteriaceae. Antimicrob 227. Sato T, Yokota S, Uchida I, Okubo T, Ishihara K, Fujii N, Tamura Y. 2011.
Resist Infect Control 6:77. https://doi.org/10.1186/s13756-017-0235-7. A fluoroquinolone-resistant Escherichia coli clinical isolate without
209. Ruiz J. 2018. Analysis of the presence of erroneous Qnr sequences in quinolone resistance-determining region mutations found in Japan.
GenBank. J Antimicrob Chemother 73:1213–1216. https://doi.org/10 Antimicrob Agents Chemother 55:3964 –3965. https://doi.org/10.1128/
.1093/jac/dky025. AAC.00532-11.
210. Pons MJ, Gomes C, Ruiz J. 2013. QnrVC a new transferable Qnr-like 228. Vien LTM, Baker S, Thao LTP, Tu LTP, Thuy CT, Nga TTT, Hoang NVM,
family. Enferm Infecc Microbiol Clin 31:191–192. https://doi.org/10 Campbell JI, Yen LM, Hieu NT, Chau NVV, Farrar J, Schultsz C. 2009. High
.1016/j.eimc.2012.09.008. prevalence of plasmid-mediated quinolone resistance determinants in
211. Leone P, Méresse S. 2011. Kinesin regulation by Salmonella. Virulence commensal members of the Enterobacteriaceae in Ho Chi Minh City,
2:63– 66. https://doi.org/10.4161/viru.2.1.14603. Vietnam. J Med Microbiol 58:1585–1592. https://doi.org/10.1099/jmm
212. Zhao L, Zhang J, Zheng B, Wei Z, Shen P, Li S, Li L, Xiao Y. 2015. .0.010033-0.
Molecular epidemiology and genetic diversity of fluoroquinolone- 229. Vinué L, Hooper DC, Jacoby GA. 2018. Chromosomal mutations that
resistant Escherichia coli isolates from patients with community-onset accompany qnr in clinical isolates of Escherichia coli. Int J Antimicrob
infections in 30 Chinese county hospitals. J Clin Microbiol 53:766 –770. Agents 51:479 – 483. https://doi.org/10.1016/j.ijantimicag.2018.01.012.
https://doi.org/10.1128/JCM.02594-14. 230. Zhao J, Chen Z, Chen S, Deng Y, Liu Y, Tian W, Huang X, Wu C, Sun Y,
213. García-León G, Ruiz de Alegría Puig C, García de la Fuente C, Martínez- Sun Y, Zeng Z, Liu JH. 2010. Prevalence and dissemination of oqxAB in
Martínez L, Martínez JL, Sánchez MB. 2015. High-level quinolone resis- Escherichia coli isolates from animals, farmworkers, and the environ-
tance is associated with the overexpression of smeVWX in Stenotroph- ment. Antimicrob Agents Chemother 54:4219 – 4224. https://doi.org/10
omonas maltophilia clinical isolates. Clin Microbiol Infect 21:464 – 467. .1128/AAC.00139-10.
https://doi.org/10.1016/j.cmi.2015.01.007. 231. Luk-In S, Pulsrikarn C, Bangtrakulnonth A, Chatsuwan T, Kulwichit W.
2017. Occurrence of a novel class 1 integron harboring qnrVC4 in regions, including aac(6=)-Ib-cr, and a novel allele, qnrB10, associated
Salmonella Rissen. Diagn Microbiol Infect Dis 88:282–286. https://doi with ISCR1 in clinical enterobacterial isolates from Argentina. Antimi-
.org/10.1016/j.diagmicrobio.2017.03.016. crob Agents Chemother 51:4466 – 4470. https://doi.org/10.1128/AAC
232. Galani I, Souli M, Mitchell N, Chryssouli Z, Giamarellou H. 2010. Pres- .00726-07.
ence of plasmid-mediated quinolone resistance in Klebsiella pneu- 249. Tamariz J, Llanos C, Seas C, Montenegro P, Lagos J, Fernandes MR,
moniae and Escherichia coli isolates possessing blaVIM-1 in Greece. Int J Cerdeira L, Lincopan N. 2018. Draft genome sequence of the first New
Antimicrob Agents 36:252–254. https://doi.org/10.1016/j.ijantimicag Delhi metallo--lactamase (NDM-1)-producing Escherichia coli strain
.2010.05.004. isolated in Peru. Genome Announc 6(13):e00199-18. https://doi.org/10
233. Karczmarczyk M, Martins M, McCusker M, Mattar S, Amaral L, Leonard .1128/genomeA.00199-18.
N, Aarestrup FM, Fanning S. 2010. Characterization of antimicrobial 250. Villa L, Poirel L, Nordmann P, Carta C, Carattoli A. 2012. Complete
resistance in Salmonella enterica food and animal isolates from sequencing of an IncH plasmid carrying the blaNDM-1, blaCTX-M-15 and
Colombia: identification of a qnrB19-mediated quinolone resistance qnrB1 genes. J Antimicrob Chemother 67:1645–1650. https://doi.org/
marker in two novel serovars. FEMS Microbiol Lett 313:10 –19. https:// 10.1093/jac/dks114.
doi.org/10.1111/j.1574-6968.2010.02119.x. 251. Vinué L, Sater MRA, Herriott I, Huntley MH, Jacoby GA, Hooper DC. 3
234. Jacoby GA, Corcoran MA, Mills DM, Griffin CM, Hooper DC. 2013. Muta- June 2019. Multiple copies of qnrA1 on an Inc A/C2 plasmid explain
tional analysis of quinolone resistance protein QnrB1. Antimicrob Agents enhanced quinolone resistance in an Escherichia coli mutant. Antimi-
Chemother 57:5733–5736. https://doi.org/10.1128/AAC.01533-13. crob Agents Chemother https://doi.org/10.1128/AAC.00718-19.
235. Tavío MM, Jacoby GA, Hooper DC. 2014. QnrS1 structure-activity rela- 252. Wang J, Li X, Li J, Hurley D, Bai X, Yu Z, Cao Y, Wall E, Fanning S, Bai L.
tionships. J Antimicrob Chemother 69:2102–2109. https://doi.org/10 2017. Complete genetic analysis of a Salmonella enterica serovar Indi-
.1093/jac/dku102. ana isolate accompanying four plasmids carrying mcr-1, ESBL and other
236. Garoff L, Yadav K, Hughes D. 2018. Increased expression of Qnr is sufficient resistance genes in China. Vet Microbiol 210:142–146. https://doi.org/
to confer clinical resistance to ciprofloxacin in Escherichia coli. J Antimicrob 10.1016/j.vetmic.2017.08.024.
Chemother 73:348–352. https://doi.org/10.1093/jac/dkx375. 253. Wang M, Tran JH, Jacoby GA, Zhang Y, Wang F, Hooper DC. 2003.
237. Akasaka T, Tanaka M, Yamaguchi A, Sato K. 2001. Type II topoisomerase Plasmid-mediated quinolone resistance in clinical isolates of Escherichia
mutations in fluoroquinolone-resistant clinical strains of Pseudomonas coli from Shanghai, China. Antimicrob Agents Chemother 47:
aeruginosa isolated in 1998 and 1999: role of target enzyme in mech- 2242–2248. https://doi.org/10.1128/AAC.47.7.2242-2248.2003.
anism of fluoroquinolone resistance. Antimicrob Agents Chemother 254. Wong MHY, Chan EWC, Xie L, Li R, Chen S. 2016. IncHI2 plasmids are the
45:2263–2268. https://doi.org/10.1128/AAC.45.8.2263-2268.2001. key vectors responsible for oqxAB transmission among Salmonella
238. Bruchmann S, Dötsch A, Nouri B, Chaberny IF, Häussler S. 2013. Quan- species. Antimicrob Agents Chemother 60:6911– 6915. https://doi.org/
titative contributions of target alteration and decreased drug accumu- 10.1128/AAC.01555-16.
lation to Pseudomonas aeruginosa fluoroquinolone resistance. Antimi- 255. Li X, Zhang Y, Zhou X, Hu X, Zhou Y, Liu D, Maxwell A, Mi K. 2019. The
crob Agents Chemother 57:1361–1368. https://doi.org/10.1128/AAC plasmid-borne quinolone resistance protein QnrB, a novel DnaA-
.01581-12. binding protein, increases the bacterial mutation rate by triggering
239. Li XZ, Plésiat P, Nikaido H. 2015. The challenge of efflux-mediated DNA replication stress. Mol Microbiol 111:1529 –1543. https://doi.org/
antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev 10.1111/mmi.14235.
28:337– 418. https://doi.org/10.1128/CMR.00117-14. 256. Cesaro A, Bettoni RR, Lascols C, Mérens A, Soussy CJ, Cambau E. 2008.
240. Cambau E, Bordon F, Collatz E, Gutmann L. 1993. Novel gyrA point Low selection of topoisomerase mutants from strains of Escherichia coli
mutation in a strain of Escherichia coli resistant to fluoroquinolones but harbouring plasmid-borne qnr genes. J Antimicrob Chemother 61:
not to nalidixic acid. Antimicrob Agents Chemother 37:1247–1252. 1007–1015. https://doi.org/10.1093/jac/dkn077.
https://doi.org/10.1128/AAC.37.6.1247. 257. Goto K, Kawamura K, Arakawa Y. 2015. Contribution of QnrA, a plasmid-
241. Moniot-Ville N, Guibert J, Moreau N, Acar JF, Collatz E, Gutmann L. mediated quinolone resistance peptide, to survival of Escherichia coli
1991. Mechanisms of quinolone resistance in a clinical isolate of Esch- exposed to a lethal ciprofloxacin concentration. Jpn J Infect Dis 68:
erichia coli highly resistant to fluoroquinolones but susceptible to 196 –202. https://doi.org/10.7883/yoken.JJID.2014.153.
nalidixic acid. Antimicrob Agents Chemother 35:519 –523. https://doi 258. Vinué L, Corcoran MA, Hooper DC, Jacoby GA. 2016. Mutations that
.org/10.1128/AAC.35.3.519. enhance the ciprofloxacin resistance of Escherichia coli with qnrA1.
242. Truong QC, Nguyen Van JC, Shlaes D, Gutmann L, Moreau NJ. 1997. A Antimicrob Agents Chemother 60:1537–1545. https://doi.org/10.1128/
novel, double mutation in DNA gyrase A of Escherichia coli conferring AAC.02167-15.
resistance to quinolone antibiotics. Antimicrob Agents Chemother 41: 259. Hirai K, Aoyama H, Irikura T, Iyobe S, Mitsuhashi S. 1986. Difference in
85–90. https://doi.org/10.1128/AAC.41.1.85. susceptibility to quinolones of outer membrane mutants of Salmonella
243. Ragunathan PL, Ison C, Livermore DM. 2005. Nalidixic acid-susceptible, typhimurium and Escherichia coli. Antimicrob Agents Chemother 29:
ciprofloxacin-resistant Neisseria gonorrhoeae strain in the UK. J Antimi- 535–538. https://doi.org/10.1128/AAC.29.3.535.
crob Chemother 56:437. https://doi.org/10.1093/jac/dki201. 260. Ebbensgaard A, Mordhorst H, Aarestrup FM, Hansen EB. 2018. The role
244. Bachoual R, Ouabdesselam S, Mory F, Lascols C, Soussy CJ, Tankovic of outer membrane proteins and lipopolysaccharides for the sensitivity
J. 2001. Single or double mutational alterations of gyrA associated of Escherichia coli to antimicrobial peptides. Front Microbiol 9:2153.
with fluoroquinolone resistance in Campylobacter jejuni and Cam- https://doi.org/10.3389/fmicb.2018.02153.
pylobacter coli. Microb Drug Resist 7:257–261. https://doi.org/10 261. Linkevicius M, Sandegren L, Andersson DI. 2013. Mechanisms and
.1089/10766290152652800. fitness costs of tigecycline resistance in Escherichia coli. J Antimicrob
245. Pérez-Moreno MO, Estepa V, Sáenz Y, Cortell-Ortolá M, Fort-Gallifa I, Chemother 68:2809 –2819. https://doi.org/10.1093/jac/dkt263.
Ruiz J, Torres C. 2012. Intrahospitalary dissemination of Klebsiella pneu- 262. Lee CC, Lui G, Ip M, Ling TK, Lee N. 2012. Frequent detection of
moniae carrying blaDHA-1 and qnrB4 genes within a novel complex class plasmid-mediated quinolone resistance (qnr) genes in multidrug-
1 integron. Diagn Microbiol Infect Dis 73:210 –211. https://doi.org/10 resistant Enterobacteriaceae blood isolates, Hong Kong. Eur J Clin Mi-
.1016/j.diagmicrobio.2012.02.011. crobiol Infect Dis 31:3183–3189. https://doi.org/10.1007/s10096-012
246. Lascols C, Podglajen I, Verdet C, Gautier V, Gutmann L, Soussy CJ, -1683-x.
Collatz E, Cambau E. 2008. A plasmid-borne Shewanella algae gene, 263. Liao CH, Hsueh PR, Jacoby GA, Hooper DC. 2013. Risk factors and
qnrA3, and its possible transfer in vivo between Kluyvera ascorbata and clinical characteristics of patients with qnr-positive Klebsiella pneu-
Klebsiella pneumoniae. J Bacteriol 190:5217–5223. https://doi.org/10 moniae bacteraemia. J Antimicrob Chemother 68:2907–2914. https://
.1128/JB.00243-08. doi.org/10.1093/jac/dkt295.
247. Liang Q, Jiang X, Hu L, Yin Z, Gao B, Zhao Y, Yang W, Yang H, Tong Y, 264. Domínguez-Herrera J, Velasco C, Docobo-Pérez F, Rodríguez-Martínez
Li W, Jiang L, Zhou D. 2019. Sequencing and genomic diversity analysis JM, López-Rojas R, Briales A, Pichardo C, Díaz-de-Alba P, Rodríguez-
of IncHI5 plasmids. Front Microbiol 9:3318. https://doi.org/10.3389/ Baño J, Pascual A, Pachón J. 2013. Impact of qnrA1, qnrB1 and qnrS1 on
fmicb.2018.03318. the efficacy of ciprofloxacin and levofloxacin in an experimental pneu-
248. Quiroga MP, Andres P, Petroni A, Soler Bistué AJC, Guerriero L, Vargas monia model caused by Escherichia coli with or without the GyrA
LJ, Zorreguieta A, Tokumoto M, Quiroga C, Tolmasky ME, Galas M, mutation Ser83Leu. J Antimicrob Chemother 68:1609 –1615. https://doi
Centrón D. 2007. Complex class 1 integrons with diverse variable .org/10.1093/jac/dkt063.
265. Jakobsen L, Cattoir V, Jensen KS, Hammerum AM, Nordmann P, carbapenemase-producing Enterobacteriaceae (EuSCAPE). Front Micro-
Frimodt-Møller N. 2012. Impact of low-level fluoroquinolone resis- biol 9:2834. https://doi.org/10.3389/fmicb.2018.02834.
tance genes qnrA1, qnrB19 and qnrS1 on ciprofloxacin treatment of 282. Mendes RE, Bell JM, Turnidge JD, Yang Q, Yu Y, Sun Z, Jones RN. 2008.
isogenic Escherichia coli strains in a murine urinary tract infection Carbapenem-resistant isolates of Klebsiella pneumoniae in China and
model. J Antimicrob Chemother 67:2438 –2444. https://doi.org/10 detection of a conjugative plasmid (blaKPC-2 plus qnrB4) and a blaIMP-4
.1093/jac/dks224. gene. Antimicrob Agents Chemother 52:798 –799. https://doi.org/10
266. Ruiz J, Pons MJ. 2018. In an inhospitable ICU, not even antibiotic cycling .1128/AAC.01185-07.
or mixing are the solutions. Ann Infect 2:2. https://doi.org/10.21037/ 283. Rice LB, Carias LL, Hutton RA, Rudin SD, Endimiani A, Bonomo RA. 2008.
aoi.2018.07.01. The KQ element, a complex genetic region conferring transferable
267. Card R, Zhang J, Das P, Cook C, Woodford N, Anjum MF. 2013. Evalu- resistance to carbapenems, aminoglycosides, and fluoroquinolones in
ation of an expanded microarray for detecting antibiotic resistance Klebsiella pneumoniae. Antimicrob Agents Chemother 52:3427–3429.
genes in a broad range of Gram-negative bacterial pathogens. Antimi- https://doi.org/10.1128/AAC.00493-08.
crob Agents Chemother 57:458 – 465. https://doi.org/10.1128/AAC 284. Rodríguez-Martínez JM, Velasco C, García I, Cano ME, Martínez-Martínez
.01223-12. L, Pascual A. 2007. Characterisation of integrons containing the
268. Rossen JWA, Friedrich AW, Moran-Gilad J, ESCMID Study Group for plasmid-mediated quinolone resistance gene qnrA1 in Klebsiella pneu-
Genomic and Molecular Diagnostics. 2018. Practical issues in imple- moniae. Int J Antimicrob Agents 29:705–709. https://doi.org/10.1016/j
menting whole-genome-sequencing in routine diagnostic microbiol- .ijantimicag.2007.02.003.
ogy. Clin Microbiol Infect 24:355–360. https://doi.org/10.1016/j.cmi 285. Ye L, Li R, Lin D, Zhou Y, Fu A, Ding Q, Chan EW, Yao W, Chen S. 2016.
.2017.11.001. Characterization of an IncA/C multidrug resistance plasmid in Vibrio
269. Schubert S, Kostrzewa M. 2017. MALDI-TOF MS in the microbiology alginolyticus. Antimicrob Agents Chemother 60:3232–3235. https://doi
laboratory: current trends. Curr Issues Mol Biol 23:17–20. https://doi .org/10.1128/AAC.00300-16.
.org/10.21775/cimb.023.017. 286. Périchon B, Bogaerts P, Lambert T, Frangeul L, Courvalin P, Galimand M.
270. Cattoir V, Poirel L, Rotimi V, Soussy CJ, Nordmann P. 2007. Multiplex 2008. Sequence of conjugative plasmid pIP1206 mediating resistance
PCR for detection of plasmid-mediated quinolone resistance qnr genes to aminoglycosides by 16S rRNA methylation and to hydrophilic fluo-
in ESBL-producing enterobacterial isolates. J Antimicrob Chemother roquinolones by efflux. Antimicrob Agents Chemother 52:2581–2592.
60:394 –397. https://doi.org/10.1093/jac/dkm204. https://doi.org/10.1128/AAC.01540-07.
271. Ciesielczuk H, Hornsey M, Choi V, Woodford N, Wareham DW. 2013. 287. Jeong HS, Bae IK, Shin JH, Jung HJ, Kim SH, Lee JY, Oh SH, Kim HR,
Development and evaluation of a multiplex PCR for eight plasmid- Chang CL, Kho WG, Lee JN. 2011. Prevalence of plasmid-mediated
mediated quinolone-resistance determinants. J Med Microbiol 62(Part quinolone resistance and its association with extended-spectrum beta-
12):1823–1827. https://doi.org/10.1099/jmm.0.064428-0. lactamase and AmpC beta-lactamase in Enterobacteriaceae. Korean J
272. Chen YT, Shu HY, Li LH, Liao TL, Wu KM, Shiau YR, Yan JJ, Su IJ, Tsai SF, Lab Med 31:257–264. https://doi.org/10.3343/kjlm.2011.31.4.257.
Lauderdale TL. 2006. Complete nucleotide sequence of pK245, a 98- 288. Tuon FF, Rocha JL, Toledo P, Arend LN, Dias CH, Leite TM, Penteado-
kilobase plasmid conferring quinolone resistance and extended- Filho SR, Pilonetto M, Zavascki AP. 2012. Risk factors for KPC-producing
spectrum--lactamase activity in a clinical Klebsiella pneumoniae iso- Klebsiella pneumoniae bacteremia. Braz J Infect Dis 16:416 – 419. https://
late. Antimicrob Agents Chemother 50:3861–3866. https://doi.org/10 doi.org/10.1016/j.bjid.2012.08.006.
.1128/AAC.00456-06. 289. Pitout JD, Wei Y, Church DL, Gregson DB. 2008. Surveillance for
273. Cunha MPV, Davies YM, Cerdeira L, Dropa M, Lincopan N, Knöbl T. 2017. plasmid-mediated quinolone resistance determinants in Enterobacteri-
Complete DNA sequence of an IncM1 plasmid bearing the novel qnrE1 aceae within the Calgary Health Region, Canada: the emergence of
plasmid-mediated quinolone resistance variant and blaCTX-M-8 from aac(6=)-Ib-cr. J Antimicrob Chemother 61:999 –1002. https://doi.org/10
Klebsiella pneumoniae sequence type 147. Antimicrob Agents Che- .1093/jac/dkn068.
mother 61:e00592-17. https://doi.org/10.1128/AAC.00592-17. 290. Piekarska K, Zacharczuk K, Wołkowicz T, Wolaniuk N, Rzeczkowska M,
274. Flach CF, Johnning A, Nilsson I, Smalla K, Kristiansson E, Larsson DG. Gierczyński R. 2019. Emergence of Enterobacteriaceae co-producing
2015. Isolation of novel IncA/C and IncN fluoroquinolone resistance CTX-M-15, ArmA and PMQR in Poland. Adv Clin Exp Med 28:249 –254.
plasmids from an antibiotic-polluted lake. J Antimicrob Chemother https://doi.org/10.17219/acem/94165.
70:2709 –2717. https://doi.org/10.1093/jac/dkv167. 291. Brahmi S, Touati A, Dunyach-Remy C, Sotto A, Pantel A, Lavigne JP.
275. Lee JJ, Kim MN, Park KS, Lee JH, Karim AM, Park M, Kim JH, Lee SH. 2016. 2018. High prevalence of extended-spectrum -lactamase-producing
Complex class 1 integron carrying qnrB62 and blaVIM-2 in a Citrobacter Enterobacteriaceae in wild fish from the Mediterranean sea in Algeria.
freundii clinical isolate. Antimicrob Agents Chemother 60:6937– 6940. Microb Drug Resist 24:290 –298. https://doi.org/10.1089/mdr.2017
https://doi.org/10.1128/AAC.00614-16. .0149.
276. Schlüter A, Nordmann P, Bonnin RA, Millemann Y, Eikmeyer FG, Wib- 292. Moremi N, Manda EV, Falgenhauer L, Ghosh H, Imirzalioglu C, Matee M,
berg D, Pühler A, Poirel L. 2014. IncH-type plasmid harboring blaCTX-M- Chakraborty T, Mshana SE. 2016. Predominance of CTX-M-15 among
15, blaDHA-1, and qnrB4 genes recovered from animal isolates. Antimi- ESBL producers from environment and fish gut from the shores of Lake
crob Agents Chemother 58:3768 –3773. https://doi.org/10.1128/AAC Victoria in Mwanza, Tanzania. Front Microbiol 7:1862. https://doi.org/
.02695-14. 10.3389/fmicb.2016.01862.
277. Siebor E, Neuwirth C. 2011. The new variant of Salmonella genomic 293. Argente M, Miró E, Martí C, Vilamala A, Alonso-Tarrés C, Ballester F,
island 1 (SGI1-V) from a Proteus mirabilis French clinical isolate harbours Calderón A, Gallés C, Gasós A, Mirelis B, Morta M, Olsina M, Sauca G,
blaVEB-6 and qnrA1 in the multiple antibiotic resistance region. J Anti- Sierra M, Rivera A, Navarro F. 2019. Molecular characterization of
microb Chemother 66:2513–2520. https://doi.org/10.1093/jac/dkr335. OXA-48 carbapenemase-producing Klebsiella pneumoniae strains after
278. Zhi C, Lv L, Yu L-F, Doi Y, Liu J-H. 2016. Dissemination of the mcr-1 a carbapenem resistance increase in Catalonia. Enferm Infecc Microbiol
colistin resistance gene. Lancet Infect Dis 16:292–293. https://doi.org/ Clin 37:82– 88. https://doi.org/10.1016/j.eimc.2018.02.003.
10.1016/S1473-3099(16)00063-3. 294. Riveros M, Riccobbono E, Durand D, Mosquito S, Ruiz J, Gotuzzo E,
279. Espedido BA, Partridge SR, Iredell JR. 2008. blaIMP-4 in different Rossolini GM, Ochoa TJ, Pallecchi L. 2012. Plasmid-mediated quinolone
genetic contexts in Enterobacteriaceae isolates from Australia. Anti- resistance genes in enteroaggregative Escherichia coli from infants in
microb Agents Chemother 52:2984 –2987. https://doi.org/10.1128/ Lima, Perú. Int J Antimicrob Agents 39:540 –542. https://doi.org/10
AAC.01634-07. .1016/j.ijantimicag.2012.02.008.
280. Li B, Feng J, Zhan Z, Yin Z, Jiang Q, Wei P, Chen X, Gao B, Hou J, Mao 295. Rodríguez-Martínez JM, Poirel L, Pascual A, Nordmann P. 2006. Plasmid-
P, Wu W, Chen W, Tong Y, Wang J, Li B, Zhou D. 2018. Dissemination mediated quinolone resistance in Australia. Microb Drug Resist 12:
of KPC-2-encoding IncX6 plasmids among multiple Enterobacteriaceae 99 –102. https://doi.org/10.1089/mdr.2006.12.99.
species in a single Chinese hospital. Front Microbiol 9:478. https://doi 296. Veldman K, Cavaco LM, Mevius D, Battisti A, Franco A, Botteldoorn N,
.org/10.3389/fmicb.2018.00478. Bruneau M, Perrin-Guyomard A, Cerny T, De Frutos Escobar C, Guerra B,
281. Manageiro V, Romão R, Moura IB, Sampaio DA, Vieira L, Ferreira E, Schroeter A, Gutierrez M, Hopkins K, Myllyniemi AL, Sunde M, Wasyl D,
Network EuSCAPE-Portugal, Caniça M. 2018. Molecular epidemiology Aarestrup FM. 2011. International collaborative study on the occur-
and risk factors of carbapenemase-producing Enterobacteriaceae iso- rence of plasmid-mediated quinolone resistance in Salmonella enterica
lates in Portuguese hospitals: results from European survey on and Escherichia coli isolated from animals, humans, food and the
environment in 13 European countries. J Antimicrob Chemother 66: genes. J Antimicrob Chemother 69:1265–1274. https://doi.org/10.1093/
1278 –1286. https://doi.org/10.1093/jac/dkr084. jac/dkt528.
297. Wen Y, Pu X, Zheng W, Hu G. 2016. High prevalence of plasmid- 316. Marti E, Variatza E, Balcázar JL. 2014. Bacteriophages as a reservoir of
mediated quinolone resistance and IncQ plasmids carrying qnrS2 gene extended-spectrum -lactamase and fluoroquinolone resistance genes
in bacteria from rivers near hospitals and aquaculture in China. PLoS in the environment. Clin Microbiol Infect 20:O456 –O459. https://doi
One 11:e0159418. https://doi.org/10.1371/journal.pone.0159418. .org/10.1111/1469-0691.12446.
298. Cattoir V, Poirel L, Aubert C, Soussy CJ, Nordmann P. 2008. Unexpected 317. Rodríguez-Martínez JM, Pascual A, García I, Martínez-Martínez L. 2003.
occurrence of plasmid-mediated quinolone resistance determinants in Detection of the plasmid-mediated quinolone resistance determinant
environmental Aeromonas spp. Emerg Infect Dis 14:231–237. https:// qnr among clinical isolates of Klebsiella pneumoniae producing AmpC-
doi.org/10.3201/eid1402.070677. type -lactamase. J Antimicrob Chemother 52:703–706. https://doi.org/
299. Chenia HY. 2016. Prevalence and characterization of plasmid-mediated 10.1093/jac/dkg388.
quinolone resistance genes in Aeromonas spp. isolated from South 318. Tran JH, Jacoby GA. 2002. Mechanism of plasmid-mediated quinolone
African freshwater fish. Int J Food Microbiol 231:26 –32. https://doi.org/ resistance. Proc Natl Acad Sci U S A 99:5638 –5642. https://doi.org/10
10.1016/j.ijfoodmicro.2016.04.030. .1073/pnas.082092899.
300. Sánchez-Céspedes J, Blasco MD, Martí S, Alba V, Alcalde E, Esteve C, Vila 319. Jacoby G, Cattoir V, Hooper D, Martínez-Martínez L, Nordmann P, Pascual
J. 2008. Plasmid-mediated QnrS2 determinant from a clinical Aeromo- A, Poirel L, Wang M. 2008. qnr gene nomenclature. Antimicrob Agents
nas veronii isolate. Antimicrob Agents Chemother 52:2990 –2991. Chemother 52:2297–2299. https://doi.org/10.1128/AAC.00147-08.
https://doi.org/10.1128/AAC.00287-08. 320. Wareham DW, Gordon NC, Shimizu K. 2011. Two new variants of and
301. Islam MA, Islam M, Hasan R, Hossain MI, Nabi A, Rahman M, Goessens creation of a repository for Stenotrophomonas maltophilia quinolone
WHF, Endtz HP, Boehm AB, Faruque SM. 2017. Environmental spread of protection protein (Smqnr) genes. Int J Antimicrob Agents 37:89 –90.
New Delhi metallo--lactamase-1-producing multidrug-resistant bac- https://doi.org/10.1016/j.ijantimicag.2010.10.002.
teria in Dhaka, Bangladesh. Appl Environ Microbiol 83:e00793-17. 321. Arsène S, Leclercq R. 2007. Role of a qnr-like gene in the intrinsic resistance
https://doi.org/10.1128/AEM.00793-17. of Enterococcus faecalis to fluoroquinolones. Antimicrob Agents Che-
302. Belotti PT, Thabet L, Laffargue A, André C, Coulange-Mayonnove L, Arpin mother 51:3254–3258. https://doi.org/10.1128/AAC.00274-07.
C, Messadi A, M’Zali F, Quentin C, Dubois V. 2015. Description of an original 322. Park YJ, Yu JK, Lee S, Oh EJ, Woo GJ. 2007. Prevalence and diversity of
integron encompassing blaVIM-2, qnrVC1 and genes encoding bacterial qnr alleles in AmpC-producing Enterobacter cloacae, Enterobacter aero-
group II intron proteins in Pseudomonas aeruginosa. J Antimicrob Che- genes, Citrobacter freundii and Serratia marcescens: a multicentre study
mother 70:2237–2240. https://doi.org/10.1093/jac/dkv103. from Korea. J Antimicrob Chemother 60:868 – 871. https://doi.org/10
303. Kumar P, Thomas S. 2011. Presence of qnrVC3 gene cassette in SXT and .1093/jac/dkm266.
class 1 integrons of Vibrio cholerae. Int J Antimicrob Agents 37: 323. Fluit AC, Schmitz FJ. 2004. Resistance integrons and super-integrons.
280 –281. https://doi.org/10.1016/j.ijantimicag.2010.12.006. Clin Microbiol Infect 10:272–288. https://doi.org/10.1111/j.1198-743X
.2004.00858.x.
304. Singh R, Rajpara N, Tak J, Patel A, Mohanty P, Vinothkumar K, Chowd-
324. Toleman MA, Bennett PM, Walsh TR. 2006. ISCR elements: novel gene-
hury G, Ramamurthy T, Ghosh A, Bhardwaj AK. 2012. Clinical isolates of
capturing systems of the 21st century? Microbiol Mol Biol Rev 70:
Vibrio fluvialis from Kolkata, India, obtained during 2006: plasmids, qnr
296 –316. https://doi.org/10.1128/MMBR.00048-05.
and mutation in gyrase A as mechanisms of multidrug resistance. J
325. Cambau E, Lascols C, Sougakoff W, Bébéar C, Bonnet R, Cavallo JD,
Med Microbiol 61:369 –374. https://doi.org/10.1099/jmm.0.037226-0.
Gutmann L, Ploy MC, Jarlier V, Soussy CJ, Robert J. 2006. Occurrence of
305. Jacoby GA, Gacharna N, Black TA, Miller GH, Hooper DC. 2009. Tem-
qnrA-positive clinical isolates in French teaching hospitals during 2002-
poral appearance of plasmid-mediated quinolone resistance genes.
2005. Clin Microbiol Infect 12:1013–1020. https://doi.org/10.1111/j
Antimicrob Agents Chemother 53:1665–1666. https://doi.org/10.1128/
.1469-0691.2006.01529.x.
AAC.01447-08.
326. Mahrouki S, Perilli M, Bourouis A, Chihi H, Ferjani M, Ben Moussa M,
306. Jacoby GA, Griffin CM, Hooper DC. 2011. Citrobacter spp. as a source of
Amicosante G, Belhadj O. 2013. Prevalence of quinolone resistance
qnrB alleles. Antimicrob Agents Chemother 55:4979 – 4984. https://doi
determinant qnrA6 among broad- and extended-spectrum beta-
.org/10.1128/AAC.05187-11.
lactam-resistant Proteus mirabilis and Morganella morganii clinical iso-
307. Saga T, Sabtcheva S, Mitsutake K, Ishii Y, Tateda K, Yamaguchi K, Kaku lates with sul1-type class 1 integron association in a Tunisian hospital.
M. 2013. Characterization of qnrB-like genes in Citrobacter species of Scand J Infect Dis 45:600 – 605. https://doi.org/10.3109/00365548.2013
the American Type Culture Collection. Antimicrob Agents Chemother .795657.
57:2863–2866. https://doi.org/10.1128/AAC.02396-12. 327. Mammeri H, Van De Loo M, Poirel L, Martinez-Martinez L, Nordmann P.
308. Sarles WB, Hammer BW. 1933. Species of Escherichia-Aerobacter organ- 2005. Emergence of plasmid-mediated quinolone resistance in Esche-
isms responsible for some defects in dairy products. J Bacteriol 25: richia coli in Europe. Antimicrob Agents Chemother 49:71–76. https://
461– 467. doi.org/10.1128/AAC.49.1.71-76.2005.
309. Werkman CH, Gillen GF. 1932. Bacteria producing trimethylene glycol. 328. Poirel L, Leviandier C, Nordmann P. 2006. Prevalence and genetic
J Bacteriol 23:167–182. analysis of plasmid-mediated quinolone resistance determinants QnrA
310. Jacoby GA, Corcoran MA, Hooper DC. 2015. Protective effect of Qnr on and QnrS in Enterobacteriaceae isolates from a French university hos-
agents other than quinolones that target DNA gyrase. Antimicrob Agents pital. Antimicrob Agents Chemother 50:3992–3997. https://doi.org/10
Chemother 59:6689–6695. https://doi.org/10.1128/AAC.01292-15. .1128/AAC.00597-06.
311. Rodríguez-Martínez JM, Velasco C, Briales A, García I, Conejo MC, 329. Potter RF, D’Souza AW, Wallace MA, Shupe A, Patel S, Gul D, Kwon
Pascual A. 2008. Qnr-like pentapeptide repeat proteins in Gram- JH, Andleeb S, Burnham CA, Dantas G. 2017. Draft genome sequence
positive bacteria. J Antimicrob Chemother 61:1240 –1243. https://doi of the blaOXA-436- and blaNDM-1-harboring Shewanella putrefaciens
.org/10.1093/jac/dkn115. SA70 isolate. Genome Announc 5(29):e00644-17. https://doi.org/10
312. Cattoir V, Poirel L, Mazel D, Soussy CJ, Nordmann P. 2007. Vibrio .1128/genomeA.00644-17.
splendidus as the source of plasmid-mediated QnrS-like quinolone 330. Kim HB, Park CH, Gavin M, Jacoby GA, Hooper DC. 2011. Cold shock
resistance determinants. Antimicrob Agents Chemother 51:2650 –2651. induces qnrA expression in Shewanella algae. Antimicrob Agents Che-
https://doi.org/10.1128/AAC.00070-07. mother 55:414 – 416. https://doi.org/10.1128/AAC.00991-10.
313. Poirel L, Liard A, Rodriguez-Martinez J-M, Nordmann P. 2005. Vibrion- 331. Melvold JA, Wyrsch ER, McKinnon J, Roy Chowdhury P, Charles IG,
aceae as a possible source of Qnr-like quinolone resistance determi- Djordjevic SP. 2017. Identification of a novel qnrA allele, qnrA8, in
nants. J Antimicrob Chemother 56:1118 –1121. https://doi.org/10.1093/ environmental Shewanella algae. J Antimicrob Chemother 72:
jac/dki371. 2949 –2952. https://doi.org/10.1093/jac/dkx226.
314. Doublet B, Boyd D, Mulvey MR, Cloeckaert A. 2005. The Salmonella 332. Zhou MM, Tu HX, Zhou TL, Fei JX, Li C, Zhao Y, Bao Q. 2010. Detection
genomic island 1 is an integrative mobilizable element. Mol Microbiol of a new qnrA7 genotypes in Shewanella algae. Chin J Microbiol
55:1911–1924. https://doi.org/10.1111/j.1365-2958.2005.04520.x. Immunol 30:593–596.
315. Colomer-Lluch M, Jofre J, Muniesa M. 2014. Quinolone resistance genes 333. Cimmino T, Olaitan AO, Rolain JM. 2016. Whole genome sequence to
(qnrA and qnrS) in bacteriophage particles from wastewater samples decipher the resistome of Shewanella algae, a multidrug-resistant
and the effect of inducing agents on packaged antibiotic resistance bacterium responsible for pneumonia, Marseille, France. Expert Rev
Anti Infect Ther 14:269 –275. https://doi.org/10.1586/14787210.2016 AL, Lozniewski A, Riahi J, Vernet-Garnier V, Cambau E. 2014. Mobile
.1106936. insertion cassette elements found in small non-transmissible plasmids
334. Aigle A, Michotey V, Bonin P. 2015. Draft-genome sequence of She- in Proteeae may explain qnrD mobilization. PLoS One 9:e87801. https://
wanella algae strain C6G3. Stand Genomic Sci 10:43. https://doi.org/10 doi.org/10.1371/journal.pone.0087801.
.1186/s40793-015-0022-0. 350. Kraychete GB, Campana EH, Picão RC, Bonelli RR. 2019. qnrD-harboring
335. Jacoby GA, Walsh KE, Mills DM, Walker VJ, Oh H, Robicsek A, Hooper plasmids in Providencia spp. recovered from food and environmental
DC. 2006. qnrB, another plasmid-mediated gene for quinolone resis- Brazilian sources. Sci Total Environ 646:1290 –1292. https://doi.org/10
tance. Antimicrob Agents Chemother 50:1178 –1182. https://doi.org/10 .1016/j.scitotenv.2018.07.378.
.1128/AAC.50.4.1178-1182.2006. 351. Pathirana HNKSP, Shin G-W, Wimalasena SHMP, De Silva BCJ, Hossain S,
336. Richter SN, Frasson I, Bergo C, Manganelli R, Cavallaro A, Palù G. 2010. Heo G-J. 2018. Prevalence and characterization of quinolone resistance
Characterisation of qnr plasmid-mediated quinolone resistance in En- genes in Proteus species isolated from pet turtles. J Exot Pet Med
terobacteriaceae from Italy: association of the qnrB19 allele with the 27:67–73. https://doi.org/10.1053/j.jepm.2017.10.026.
integron element ISCR1 in Escherichia coli. Int J Antimicrob Agents 352. Bueno MFC, Francisco GR, Cerdeira LT, Ienne S, Souza TA, Lincopan N,
35:578 –583. https://doi.org/10.1016/j.ijantimicag.2010.02.015. Garcia DO. 2016. Sequenciamento completo de plasmídeo que carrega
337. Cattoir V, Nordmann P, Silva-Sanchez J, Espinal P, Poirel L. 2008. blaCTX-M-8 de Klebsiella pneumoniae multirresistente, abstr 8573. Ab-
ISEcp1-mediated transposition of qnrB-like gene in Escherichia coli. str 52nd Congr Soc Bras Med Trop.
Antimicrob Agents Chemother 52:2929 –2932. https://doi.org/10.1128/ 353. Soares FB, Camargo CH, Cunha MPV, de Almeida EA, Bertani AMJ,
AAC.00349-08. Carvalho E, de Paiva JB, Fernandes SA, Tiba-Casas MR. 2019. Co-
338. Venter JC, Remington K, Heidelberg JF, Halpern AL, Rusch D, Eisen JA, occurrence of qnrE1 and blaCTX-M-8 in IncM1 transferable plasmids
Wu D, Paulsen I, Nelson KE, Nelson W, Fouts DE, Levy S, Knap AH, contributing to MDR in different Salmonella serotypes. J Antimicrob
Lomas MW, Nealson K, White O, Peterson J, Hoffman J, Parsons R, Chemother 74:1155–1156. https://doi.org/10.1093/jac/dky516.
Baden-Tillson H, Pfannkoch C, Rogers Y-H, Smith HO. 2004. Environ- 354. Kehrenberg C, Friederichs S, de Jong A, Michael GB, Schwarz S. 2006.
mental genome shotgun sequencing of the Sargasso Sea. Science Identification of the plasmid-borne quinolone resistance gene qnrS in
304:66 –74. https://doi.org/10.1126/science.1093857. Salmonella enterica serovar Infantis. J Antimicrob Chemother 58:18 –22.
339. Sánchez MB, Hernández A, Rodríguez-Martínez JM, Martínez-Martínez https://doi.org/10.1093/jac/dkl213.
L, Martínez JL. 2008. Predictive analysis of transmissible quinolone 355. Lavilla S, González-López JJ, Sabaté M, García-Fernández A, Larrosa MN,
resistance indicates Stenotrophomonas maltophilia as a potential Bartolomé RM, Carattoli A, Prats G. 2008. Prevalence of qnr genes
source of a novel family of Qnr determinants. BMC Microbiol 8:148. among extended-spectrum -lactamase-producing enterobacterial iso-
https://doi.org/10.1186/1471-2180-8-148. lates in Barcelona, Spain. J Antimicrob Chemother 61:291–295. https://
340. Wang M, Guo Q, Xu X, Wang X, Ye X, Wu S, Hooper DC, Wang M. 2009. doi.org/10.1093/jac/dkm448.
New plasmid-mediated quinolone resistance gene, qnrC, found in a 356. Yue L, Jiang HX, Liao XP, Liu JH, Li SJ, Chen XY, Chen CX, Lü DH, Liu YH.
clinical isolate of Proteus mirabilis. Antimicrob Agents Chemother 53: 2008. Prevalence of plasmid-mediated quinolone resistance qnr genes
1892–1897. https://doi.org/10.1128/AAC.01400-08. in poultry and swine clinical isolates of Escherichia coli. Vet Microbiol
341. Azargun R, Sadeghi MR, Barhaghi MHS, Kafil HS, Yeganeh F, Oskouee 132:414 – 420. https://doi.org/10.1016/j.vetmic.2008.05.009.
MA, Ghotaslou R. 2018. The prevalence of plasmid-mediated quinolone 357. Hu FP, Xu XG, Zhu DM, Wang MG. 2008. Coexistence of qnrB4 and qnrS1
resistance and ESBL-production in Enterobacteriaceae isolated from in a clinical strain of Klebsiella pneumoniae. Acta Pharmacol Sin 29:
urinary tract infections. Infect Drug Resist 11:1007–1014. https://doi 320 –324. https://doi.org/10.1111/j.1745-7254.2008.00757.x.
.org/10.2147/IDR.S160720. 358. Poirel L, Cattoir V, Soares A, Soussy CJ, Nordmann P. 2007. Novel
342. El-Badawy MF, Tawakol WM, El-Far SW, Maghrabi IA, Al-Ghamdi SA, Ambler class A -lactamase LAP-1 and its association with the plasmid-
Mansy MS, Ashour MS, Shohayeb MM. 2017. Molecular identification of mediated quinolone resistance determinant QnrS1. Antimicrob Agents
aminoglycoside-modifying enzymes and plasmid-mediated quinolone Chemother 51:631– 637. https://doi.org/10.1128/AAC.01082-06.
resistance genes among Klebsiella pneumoniae clinical isolates recov- 359. Kim HB, Wang M, Ahmed S, Park CH, LaRocque RC, Faruque ASG, Salam
ered from Egyptian patients. Int J Microbiol 2017:8050432. https://doi MA, Khan WA, Qadri F, Calderwood SB, Jacoby GA, Hooper DC. 2010.
.org/10.1155/2017/8050432. Transferable quinolone resistance in Vibrio cholerae. Antimicrob Agents
343. Das A, Natarajan M, Mandal J. 2016. The emergence of quinolone Chemother 54:799 – 803. https://doi.org/10.1128/AAC.01045-09.
resistant Shigella sonnei, Pondicherry, India. PLoS One 11:e0160290. 360. Rajpara N, Patel A, Tiwari N, Bahuguna J, Antony A, Choudhury I,
https://doi.org/10.1371/journal.pone.0160290. Ghosh A, Jain R, Ghosh A, Bhardwaj AK. 2009. Mechanism of drug
344. Cavaco LM, Hasman H, Xia S, Aarestrup FM. 2009. qnrD, a novel gene resistance in a clinical isolate of Vibrio fluvialis: involvement of
conferring transferable quinolone resistance in Salmonella enterica multiple plasmids and integrons. Int J Antimicrob Agents 34:
serovar Kentucky and Bovismorbificans strains of human origin. 220 –222. https://doi.org/10.1016/j.ijantimicag.2009.03.020.
Antimicrob Agents Chemother 53:603– 608. https://doi.org/10.1128/ 361. Xia R, Guo X, Zhang Y, Xu H. 2010. qnrVC-like gene located in a novel
AAC.00997-08. complex class 1 integron harboring the ISCR1 element in an Aeromonas
345. Guillard T, Cambau E, Neuwirth C, Nenninger T, Mbadi A, Brasme L, punctata strain from an aquatic environment in Shandong province,
Vernet-Garnier V, Bajolet O, de Champs C. 2012. Description of a China. Antimicrob Agents Chemother 54:3471–3474. https://doi.org/10
2,683-base-pair plasmid containing qnrD in two Providencia rettgeri .1128/AAC.01668-09.
isolates. Antimicrob Agents Chemother 56:565–568. https://doi.org/10 362. Wu K, Wang F, Sun J, Wang Q, Chen Q, Yu S, Rui Y. 2012. Class 1
.1128/AAC.00081-11. integron gene cassettes in multidrug-resistant Gram-negative bacteria
346. Zhang S, Sun J, Liao XP, Hu QJ, Liu BT, Fang LX, Deng H, Ma J, Xiao X, in southern China. Int J Antimicrob Agents 40:264 –267. https://doi.org/
Zhu HQ, Liu YH. 2013. Prevalence and plasmid characterization of the 10.1016/j.ijantimicag.2012.05.017.
qnrD determinant in Enterobacteriaceae isolated from animals, retail 363. Zhang Y, Zheng Z, Chan EW, Dong N, Xia X, Chen S. 2018. Molecular
meat products, and humans. Microb Drug Resist 19:331–335. https:// characterization of qnrVC genes and their novel alleles in Vibrio spp.
doi.org/10.1089/mdr.2012.0146. isolated from food products in China. Antimicrob Agents Chemother
347. Adeolu M, Alnajar S, Naushad S, Gupta RS. 2016. Genome-based phy- 62:e00529-18. https://doi.org/10.1128/AAC.00529-18.
logeny and taxonomy of the ‘Enterobacteriales’: proposal for Enterobac- 364. Bado I, Ezdra RP, Cordeiro N, Outeda M, Caiata L, García-Fulgueiras V,
terales ord. nov. divided into the families Enterobacteriaceae, Erwini- Seija V, Vignoli R. 2018. Detection of qnrVC6, within a new genetic
aceae fam. nov., Pectobacteriaceae fam. nov., Yersiniaceae fam. nov., context, in a NDM-1-producing Citrobacter freundii clinical isolate from
Hafniaceae fam. nov., Morganellaceae fam. nov., and Budviciaceae fam. Uruguay. J Glob Antimicrob Resist 14:95–98. https://doi.org/10.1016/j
nov. Int J Syst Evol Microbiol 66:5575–5599. https://doi.org/10.1099/ .jgar.2018.02.023.
ijsem.0.001485. 365. Janvier F, Otto MP, Jové T, Mille A, Contargyris C, Meaudre E, Brisou P,
348. Chen L, Zhang Y, Du J, Zhang X, Li M, Chen H, Yu X, Sun Y, Zhou T. 2018. Plésiat P, Jeannot K. 2017. A case of multiple contamination with
Description and plasmid characterization of the qnrD determinant in methylase ArmA-producing pathogens. J Antimicrob Chemother 72:
Proteeae in Wenzhou, Southern China. J Microbiol Immunol Infect 618 – 620. https://doi.org/10.1093/jac/dkw418.
51:115–122. https://doi.org/10.1016/j.jmii.2016.02.001. 366. Tacão M, Moura A, Correia A, Henriques I. 2014. Co-resistance to
349. Guillard T, Grillon A, de Champs C, Cartier C, Madoux J, Berçot B, Lebreil different classes of antibiotics among ESBL-producers from aquatic
systems. Water Res 48:100 –107. https://doi.org/10.1016/j.watres.2013 386. Wang M, Jacoby GA, Mills DM, Hooper DC. 2009. SOS regulation of qnrB
.09.021. expression. Antimicrob Agents Chemother 53:821– 823. https://doi.org/
367. Jacoby GA, Hooper DC. 2013. Phylogenetic analysis of chromosomally 10.1128/AAC.00132-08.
determined qnr and related proteins. Antimicrob Agents Chemother 387. Hau HH, Gralnick JA. 2007. Ecology and biotechnology of the genus
57:1930 –1934. https://doi.org/10.1128/AAC.02080-12. Shewanella. Annu Rev Microbiol 61:237–258. https://doi.org/10.1146/
368. Shimizu K, Kikuchi K, Sasaki T, Takahashi N, Ohtsuka M, Ono Y, Hira- annurev.micro.61.080706.093257.
matsu K. 2008. Smqnr, a new chromosome-carried quinolone resistance 388. Tran JH, Jacoby GA, Hooper DC. 2005. Interaction of the plasmid-
gene in Stenotrophomonas maltophilia. Antimicrob Agents Chemother encoded quinolone resistance protein Qnr with Escherichia coli DNA
52:3823–3825. https://doi.org/10.1128/AAC.00026-08. gyrase. Antimicrob Agents Chemother 49:118 –125. https://doi.org/10
369. Gracia-Paez JI, Ferraz JR, Silva IA, Rossi F, Levin AS, Costa SF. 2013. .1128/AAC.49.1.118-125.2005.
Smqnr variants in clinical isolates of Stenotrophomonas maltophilia in 389. Tran JH, Jacoby GA, Hooper DC. 2005. Interaction of the plasmid-
Brazil. Rev Inst Med Trop Sao Paulo 55:417– 420. https://doi.org/10 encoded quinolone resistance protein QnrA with Escherichia coli topo-
.1590/S0036-46652013000600008. isomerase IV. Antimicrob Agents Chemother 49:3050 –3052. https://doi
370. Kanamori H, Yano H, Tanouchi A, Kakuta R, Endo S, Ichimura S, Ogawa .org/10.1128/AAC.49.7.3050-3052.2005.
M, Shimojima M, Inomata S, Ozawa D, Aoyagi T, Weber DJ, Kaku M. 390. Kim ES, Chen C, Braun M, Kim HY, Okumura R, Wang Y, Jacoby GA,
2015. Prevalence of Smqnr and plasmid-mediated quinolone resistance Hooper DC. 2015. Interactions between QnrB, QnrB mutants, and DNA
determinants in clinical isolates of Stenotrophomonas maltophilia from gyrase. Antimicrob Agents Chemother 59:5413–5419. https://doi.org/
Japan: novel variants of Smqnr. New Microbes New Infect 7:8 –14. 10.1128/AAC.00771-15.
https://doi.org/10.1016/j.nmni.2015.04.009. 391. Cattoir V, Poirel L, Nordmann P. 2007. In-vitro mutagenesis of qnrA and
371. Shah S, Heddle JG. 2014. Squaring up to DNA: pentapeptide repeat qnrS genes and quinolone resistance in Escherichia coli. Clin Microbiol
proteins and DNA mimicry. Appl Microbiol Biotechnol 98:9545–9560. Infect 13:940 –943. https://doi.org/10.1111/j.1469-0691.2007.01778.x.
https://doi.org/10.1007/s00253-014-6151-3. 392. Rodríguez-Martínez JM, Briales A, Velasco C, Conejo MC, Martínez-
372. Vetting MW, Hegde SS, Fajardo JE, Fiser A, Roderick SL, Takiff HE, Martínez L, Pascual A. 2009. Mutational analysis of quinolone resistance
Blanchard JS. 2006. Pentapeptide repeat proteins. Biochemistry 45: in the plasmid-encoded pentapeptide repeat proteins QnrA, QnrB and
1–10. https://doi.org/10.1021/bi052130w. QnrS. J Antimicrob Chemother 63:1128 –1134. https://doi.org/10.1093/
373. Hegde SS, Vetting MW, Roderick SL, Mitchenall LA, Maxwell A, Takiff HE, jac/dkp111.
Blanchard JS. 2005. A fluoroquinolone resistance protein from Myco- 393. Guo Q, Weng J, Xu X, Wang M, Wang X, Ye X, Wang W, Wang M. 2010.
bacterium tuberculosis that mimics DNA. Science 308:1480 –1483. A mutational analysis and molecular dynamics simulation of quinolone
https://doi.org/10.1126/science.1110699. resistance proteins QnrA1 and QnrC from Proteus mirabilis. BMC Struct
374. Montero C, Mateu G, Rodríguez R, Takiff H. 2001. Intrinsic resistance of Biol 10:33. https://doi.org/10.1186/1472-6807-10-33.
Mycobacterium smegmatis to fluoroquinolones may be influenced by 394. Po KH, Chan EW, Chen S. 2016. Mutational analysis of quinolone
new pentapeptide protein MfpA. Antimicrob Agents Chemother 45: resistance protein QnrVC7 provides novel insights into the structure-
3387–3392. https://doi.org/10.1128/AAC.45.12.3387-3392.2001. activity relationship of Qnr proteins. Antimicrob Agents Chemother
375. Garrido MC, Herrero M, Kolter R, Moreno F. 1988. The export of the 60:1939 –1942. https://doi.org/10.1128/AAC.01805-15.
DNA replication inhibitor microcin B17 provides immunity for the 395. Martens R, Wetzstein HG, Zadrazil F, Capelari M, Hoffmann P, Schmeer
host cell. EMBO J 7:1853–1862. https://doi.org/10.1002/j.1460-2075 N. 1996. Degradation of the fluoroquinolone enrofloxacin by wood-
.1988.tb03018.x. rotting fungi. Appl Environ Microbiol 62:4206 – 4209.
376. Vetting MW, Hegde SS, Wang M, Jacoby GA, Hooper DC, Blanchard JS. 396. Wetzstein HG, Schmeer N, Karl W. 1997. Degradation of the fluoroquin-
2011. Structure of QnrB1, a plasmid-mediated fluoroquinolone resis- olone enrofloxacin by the brown rot fungus Gloeophyllum striatum:
tance factor. J Biol Chem 286:25265–25273. https://doi.org/10.1074/jbc identification of metabolites. Appl Environ Microbiol 63:4272– 4281.
.M111.226936. 397. Wetzstein HG, Stadler M, Tichy HV, Dalhoff A, Karl W. 1999. Degradation
377. Xiong X, Bromley EH, Oelschlaeger P, Woolfson DN, Spencer J. 2011. of ciprofloxacin by basidiomycetes and identification of metabolites
Structural insights into quinolone antibiotic resistance mediated by generated by the brown rot fungus Gloeophyllum striatum. Appl Envi-
pentapeptide repeat proteins: conserved surface loops direct the ac- ron Microbiol 65:1556 –1563.
tivity of a Qnr protein from a gram-negative bacterium. Nucleic Acids 398. Rusch M, Kauschat A, Spielmeyer A, Römpp A, Hausmann H, Zorn H,
Res 39:3917–3927. https://doi.org/10.1093/nar/gkq1296. Hamscher G. 2015. Biotransformation of the antibiotic danofloxacin by
378. Hooper DC, Jacoby GA. 2016. Topoisomerase inhibitors: fluoroquin- Xylaria longipes leads to an efficient reduction of its antibacterial
olone mechanisms of action and resistance. Cold Spring Harb Perspect activity. J Agric Food Chem 63:6897– 6904. https://doi.org/10.1021/acs
Med 6:a025320. https://doi.org/10.1101/cshperspect.a025320. .jafc.5b02343.
379. Park KS, Lee JH, Jeong DU, Lee JJ, Wu X, Jeong BC, Kang CM, Lee SH. 399. Chen Y, Rosazza JP, Reese CP, Chang HY, Nowakowski MA, Kiplinger JP.
2011. Determination of pentapeptide repeat units in Qnr proteins by 1997. Microbial models of soil metabolism: biotransformations of dano-
the structure-based alignment approach. Antimicrob Agents Che- floxacin. J Ind Microbiol Biotechnol 19:378 –384. https://doi.org/10
mother 55:4475– 4478. https://doi.org/10.1128/AAC.00041-11. .1038/sj.jim.2900409.
380. Vetting MW, Hegde SS, Zhang Y, Blanchard JS. 2011. Pentapeptide-repeat 400. Kim DW, Thawng CN, Lee K, Cha CJ. 2018. Revisiting polymorphic
proteins that act as topoisomerase poison resistance factors have a com- diversity of aminoglycoside N-acetyltransferase AAC(6=)-Ib based on
mon dimer interface. Acta Crystallogr Sect F Struct Biol Cryst Commun bacterial genomes of human, animal, and environmental origins. Front
67:296–302. https://doi.org/10.1107/S1744309110053315. Microbiol 9:1831. https://doi.org/10.3389/fmicb.2018.01831.
381. Hegde SS, Vetting MW, Mitchenall LA, Maxwell A, Blanchard JS. 2011. 401. Albornoz E, Lucero C, Romero G, Quiroga MP, Rapoport M, Guerriero L,
Structural and biochemical analysis of the pentapeptide repeat protein Andres P, Rodriguez C, Galas M, Centrón D, Corso A, Petroni A. 2017.
EfsQnr, a potent DNA gyrase inhibitor. Antimicrob Agents Chemother Prevalence of plasmid-mediated quinolone resistance genes in clinical
55:110 –117. https://doi.org/10.1128/AAC.01158-10. Enterobacteria from Argentina. Microb Drug Resist 23:177–187. https://
382. Ellington MJ, Woodford N. 2006. Fluoroquinolone resistance and plas- doi.org/10.1089/mdr.2016.0033.
mid addiction systems: self-imposed selection pressure? J Antimicrob 402. Ruiz E, Ocampo-Sosa AA, Alcoba-Flórez J, Román E, Arlet G, Torres C,
Chemother 57:1026 –1029. https://doi.org/10.1093/jac/dkl110. Martínez-Martínez L. 2012. Changes in ciprofloxacin resistance levels in
383. Kwak YG, Jacoby GA, Hooper DC. 2015. Effect of Qnr on plasmid gyrase Enterobacter aerogenes isolates associated with variable expression of
toxins CcdB and ParE. Antimicrob Agents Chemother 59:5078 –5079. the aac(6=)-Ib-cr gene. Antimicrob Agents Chemother 56:1097–1100.
https://doi.org/10.1128/AAC.00524-15. https://doi.org/10.1128/AAC.05074-11.
384. Rodríguez-Martínez JM, Machuca J, Cano ME, Calvo J, Martínez- 403. Chang CY, Fang YT, Tsai SM, Chang LL, Yu WL. 2009. Characterization
Martínez L, Pascual A. 2016. Plasmid-mediated quinolone resistance: of class 1 integrons and gene cassettes in clinical isolates of Klebsiella
two decades on. Drug Resist Updat 29:13–29. https://doi.org/10.1016/ pneumoniae from Taiwan. Diagn Microbiol Infect Dis 65:214 –216.
j.drup.2016.09.001. https://doi.org/10.1016/j.diagmicrobio.2009.06.005.
385. Da Re S, Garnier F, Guérin E, Campoy S, Denis F, Ploy MC. 2009. The SOS 404. Moura A, Pereira C, Henriques I, Correia A. 2012. Novel gene cassettes and
response promotes qnrB quinolone-resistance determinant expression. integrons in antibiotic-resistant bacteria isolated from urban wastewaters.
EMBO Rep 10:929 –933. https://doi.org/10.1038/embor.2009.99. Res Microbiol 163:92–100. https://doi.org/10.1016/j.resmic.2011.10.010.
405. Pérez-Moreno MO, Centelles-Serrano MJ, Cortell-Ortolá M, Fort-Gallifa I, 422. Veleba M, Higgins PG, Gonzalez G, Seifert H, Schneiders T. 2012.
Ruiz J, Llovet-Lombarte MI, Picó-Plana E, Jardí-Baiges AM. 2011. Molec- Characterization of RarA, a novel AraC family multidrug resistance
ular epidemiology and resistance mechanisms involved in reduced regulator in Klebsiella pneumoniae. Antimicrob Agents Chemother 56:
susceptibility to amoxicillin/clavulanic acid in Klebsiella pneumoniae 4450 – 4458. https://doi.org/10.1128/AAC.00456-12.
isolates from a chronic care centre. Int J Antimicrob Agents 37: 423. Furlan JPR, Sanchez DG, Gallo IFL, Stehling EG. 2019. Characterization
462– 466. https://doi.org/10.1016/j.ijantimicag.2010.12.010. of acquired antimicrobial resistance genes in environmental
406. Kim DW, Thawng CN, Choi JH, Lee K, Cha CJ. 2018. Polymorphism of Stenotrophomonas maltophilia isolates from Brazil. Microb Drug Resist
antibiotic-inactivating enzyme driven by ecology expands the environ- 25:475– 479. https://doi.org/10.1089/mdr.2018.0216.
mental resistome. ISME J 12:267–276. https://doi.org/10.1038/ismej 424. Chen X, Zhang W, Pan W, Yin J, Pan Z, Gao S, Jiao X. 2012. Prevalence
.2017.168. of qnr, aac(6=)-Ib-cr, qepA, and oqxAB in Escherichia coli isolates from
407. Maurice F, Broutin I, Podglajen I, Benas P, Collatz E, Dardel F. 2008. humans, animals, and the environment. Antimicrob Agents Chemother
Enzyme structural plasticity and the emergence of broad-spectrum 56:3423–3427. https://doi.org/10.1128/AAC.06191-11.
antibiotic resistance. EMBO Rep 9:344 –349. https://doi.org/10.1038/ 425. Dotto G, Giacomelli M, Grilli G, Ferrazzi V, Carattoli A, Fortini D, Piccirillo
embor.2008.9. A. 2014. High prevalence of oqxAB in Escherichia coli isolates from
408. Wang D, Wang H, Qi Y, Liang Y, Zhang J, Yu L. 2012. Characteristics of domestic and wild lagomorphs in Italy. Microb Drug Resist 20:118 –123.
Klebsiella pneumoniae harboring QnrB32, Aac(6=)-Ib-cr, GyrA and CTX- https://doi.org/10.1089/mdr.2013.0141.
M-22 genes. Folia Histochem Cytobiol 50:68 –74. https://doi.org/10 426. Janecko N, Halova D, Jamborova I, Papousek I, Masarikova M, Dolejska
.2478/18698. M, Literak I. 2018. Occurrence of plasmid-mediated quinolone resis-
409. de Toro M, Rodríguez I, Rojo-Bezares B, Helmuth R, Torres C, Guerra B, tance genes in Escherichia coli and Klebsiella spp. recovered from
Sáenz Y. 2013. pMdT1, a small ColE1-like plasmid mobilizing a new Corvus brachyrhynchos and Corvus corax roosting in Canada. Lett Appl
variant of the aac(6=)-Ib-cr gene in Salmonella enterica serovar Typhi- Microbiol 67:130 –135. https://doi.org/10.1111/lam.12993.
murium. J Antimicrob Chemother 68:1277–1280. https://doi.org/10 427. Li L, Liao X, Yang Y, Sun J, Li L, Liu B, Yang S, Ma J, Li X, Zhang Q, Liu
.1093/jac/dkt001. Y. 2013. Spread of oqxAB in Salmonella enterica serotype Typhimurium
410. Vetting MW, Park CH, Hegde SS, Jacoby GA, Hooper DC, Blanchard JS. predominantly by IncHI2 plasmids. J Antimicrob Chemother 68:
2008. Mechanistic and structural analysis of aminoglycoside 2263–2268. https://doi.org/10.1093/jac/dkt209.
N-acetyltransferase AAC(6)-Ib and its bifunctional, fluoroquinolone- 428. Manageiro V, Félix D, Jones-Dias D, Sampaio DA, Vieira L, Sancho L,
active AAC(6)-Ib-cr variant. Biochemistry 47:9825–9835. https://doi.org/ Ferreira E, Caniça M. 2017. Genetic background and expression of the
10.1021/bi800664x. new qepA4 gene variant recovered in clinical TEM-1- and CMY-2-
411. Jones GL, Warren RE, Skidmore SJ, Davies VA, Gibreel T, Upton M. 2008. producing Escherichia coli. Front Microbiol 8:1899. https://doi.org/10
Prevalence and distribution of plasmid-mediated quinolone resistance .3389/fmicb.2017.01899.
genes in clinical isolates of Escherichia coli lacking extended-spectrum 429. Rahman Z, Islam A, Rashid MU, Johura FT, Monira S, Watanabe H,
beta-lactamases. J Antimicrob Chemother 62:1245–1251. https://doi
Ahmed N, Camilli A, Alam M. 2017. Existence of a novel qepA variant in
.org/10.1093/jac/dkn406.
quinolone resistant Escherichia coli from aquatic habitats of Bangla-
412. Cervantes-Vega C, Chávez J, Córdova NA, de la Mora P, Amador Velasco
desh. Gut Pathog 9:58. https://doi.org/10.1186/s13099-017-0207-8.
J. 1986. Resistance to metals by Pseudomonas aeruginosa clinical iso-
430. Wang D, Huang X, Chen J, Mou Y, Li H, Yang L. 2015. Characteriza-
lates. Microbios 48:159 –163.
tion of genetic structures of the QepA3 gene in clinical isolates of
413. Ramírez-Díaz MI, Díaz-Magaña A, Meza-Carmen V, Johnstone L, Cer-
Enterobacteriaceae. Front Microbiol 6:1147. https://doi.org/10.3389/
vantes C, Rensing C. 2011. Nucleotide sequence of Pseudomonas
fmicb.2015.01147.
aeruginosa conjugative plasmid pUM505 containing virulence and
431. Machuca J, Briales A, Díaz-de-Alba P, Martínez-Martínez L, Pascual Á,
heavy-metal resistance genes. Plasmid 66:7–18. https://doi.org/10
Rodríguez-Martínez JM. 2015. Effect of the efflux pump QepA2 com-
.1016/j.plasmid.2011.03.002.
bined with chromosomally mediated mechanisms on quinolone resis-
414. Ruiz J. 23 July 2019. CrpP, a passenger or a hidden stowaway in the
tance and bacterial fitness in Escherichia coli. J Antimicrob Chemother
Pseudomonas aeruginosa genome? J Antimicrob Chemother https://doi
70:2524 –2527. https://doi.org/10.1093/jac/dkv144.
.org/10.1093/jac/dkz316.
415. Kung VL, Ozer EA, Hauser AR. 2010. The accessory genome of Pseu- 432. Littlejohn TG, Paulsen IT, Gillespie MT, Tennent JM, Midgley M, Jones IG,
domonas aeruginosa. Microbiol Mol Biol Rev 74:621– 641. https://doi Purewal AS, Skurray RA. 1992. Substrate specificity and energetics of
.org/10.1128/MMBR.00027-10. antiseptic and disinfectant resistance in Staphylococcus aureus. FEMS
416. Klockgether J, Reva O, Larbig K, Tümmler B. 2004. Sequence analysis of Microbiol Lett 74:259 –265. https://doi.org/10.1111/j.1574-6968.1992
the mobile genome island pKLC102 of Pseudomonas aeruginosa C. J .tb05376.x.
Bacteriol 186:518 –534. https://doi.org/10.1128/JB.186.2.518-534.2004. 433. Paulsen IT, Brown MH, Littlejohn TG, Mitchell BA, Skurray RA. 1996.
417. Chávez-Jacobo VM, Hernández-Ramírez KC, Silva-Sánchez J, Garza- Multidrug resistance proteins QacA and QacB from Staphylococcus
Ramos U, Barrios-Camacho H, Ortiz-Alvarado R, Cervantes C, Meza- aureus: membrane topology and identification of residues involved in
Carmen V, Ramírez-Díaz MI. 2019. Prevalence of the crpP gene confer- substrate specificity. Proc Natl Acad Sci U S A 93:3630 –3635. https://
ring decreased ciprofloxacin susceptibility in enterobacterial clinical doi.org/10.1073/pnas.93.8.3630.
isolates from Mexican hospitals. J Antimicrob Chemother 74: 434. Miyajima E, Harada D, Nakaminami H, Kitamura Y, Tamura T, Kawakubo T,
1253–1259. https://doi.org/10.1093/jac/dky562. Noguchi N. 9 April 2019. Decreased prevalence of qacA-positive
418. Horna G, López M, Guerra H, Saénz Y, Ruiz J. 2018. Interplay between methicillin-resistant Staphylococcus aureus in hospitalized patients in To-
MexAB-OprM and MexEF-OprN in clinical isolates of Pseudomonas aerugi- kyo, Japan. Microb Drug Resist https://doi.org/10.1089/mdr.2018.0351.
nosa. Sci Rep 8:16463. https://doi.org/10.1038/s41598-018-34694-z. 435. Infante-Martínez V, Rodríguez-Martínez JM, Velasco C, Pascual A. 2012.
419. Lomovskaya O, Warren MS, Lee A, Galazzo J, Fronko R, Lee M, Blais J, Ausencia del sistema de expulsión de quinolonas mediado por
Cho D, Chamberland S, Renau T, Leger R, Hecker S, Watkins W, Hoshino plásmido QacBIII en aislados bacteriémicos de Staphylococcus aureus
K, Ishida H, Lee VJ. 2001. Identification and characterization of inhibi- en el área hospitalaria Virgen Macarena de Sevilla. Enferm Infecc
tors of multidrug resistance efflux pumps in Pseudomonas aeruginosa: Microbiol Clin 30:51. https://doi.org/10.1016/j.eimc.2011.07.019.
novel agents for combination therapy. Antimicrob Agents Chemother 436. Szczepanowski R, Krahn I, Linke B, Goesmann A, Pühler A, Schlüter A.
45:105–116. https://doi.org/10.1128/AAC.45.1.105-116.2001. 2004. Antibiotic multiresistance plasmid pRSB101 isolated from a
420. Chen Y, Hu D, Zhang Q, Liao XP, Liu YH, Sun J. 2017. Efflux pump wastewater treatment plant is related to plasmids residing in phyto-
overexpression contributes to tigecycline heteroresistance in Salmo- pathogenic bacteria and carries eight different resistance determinants
nella enterica serovar Typhimurium. Front Cell Infect Microbiol 7:37. including a multidrug transport system. Microbiology 150:3613–3630.
https://doi.org/10.3389/fcimb.2017.00037. https://doi.org/10.1099/mic.0.27317-0.
421. Bialek-Davenet S, Lavigne JP, Guyot K, Mayer N, Tournebize R, Brisse S, 437. Dolejska M, Villa L, Poirel L, Nordmann P, Carattoli A. 2013. Complete
Leflon-Guibout V, Nicolas-Chanoine MH. 2015. Differential contribution sequencing of an IncHI1 plasmid encoding the carbapenemase NDM-1,
of AcrAB and OqxAB efflux pumps to multidrug resistance and viru- the ArmA 16S RNA methylase and a resistance-nodulation-cell division/
lence in Klebsiella pneumoniae. J Antimicrob Chemother 70:81– 88. multidrug efflux pump. J Antimicrob Chemother 68:34 –39. https://doi
https://doi.org/10.1093/jac/dku340. .org/10.1093/jac/dks357.
438. Abraham EP, Chain E. 1940. An enzyme from bacteria able to destroy 457. Ma X, Lü X, Chen H, Gao Y, Mu L, Yu R, Du R, Niu F. 2009. Study on
penicillin. Nature 146:837. https://doi.org/10.1038/146837a0. qnrB-mediated quinolone resistance among Enterobacteriaceae spe-
439. Appelt S, Fancello L, Le Bailly M, Raoult D, Drancourt M, Desnues C. cies. Chin J Antibiot 2:114 –119.
2014. Viruses in a 14th-century coprolite. Appl Environ Microbiol 80: 458. Ma J, Zeng Z, Chen Z, Xu X, Wang X, Deng Y, Lü D, Huang L, Zhang Y,
2648 –2655. https://doi.org/10.1128/AEM.03242-13. Liu J, Wang M. 2009. High prevalence of plasmid-mediated quinolone
440. Petrova M, Gorlenko Z, Mindlin S. 2011. Tn5045, a novel integron- resistance determinants qnr, aac(6=)-Ib-cr, and qepA among ceftiofur-
containing antibiotic and chromate resistance transposon isolated resistant Enterobacteriaceae isolates from companion and food-
from a permafrost bacterium. Res Microbiol 162:337–345. https://doi producing animals. Antimicrob Agents Chemother 53:519 –524. https://
.org/10.1016/j.resmic.2011.01.003. doi.org/10.1128/AAC.00886-08.
441. Hall BG, Barlow M. 2004. Evolution of the serine beta-lactamases: past, 459. Lenaerts AJ, Bitting C, Woolhiser L, Gruppo V, Marietta KS, Johnson CM,
present and future. Drug Resist Updat 7:111–123. https://doi.org/10 Orme IM. 2008. Evaluation of a 2-pyridone, KRQ-10018, against Myco-
.1016/j.drup.2004.02.003. bacterium tuberculosis in vitro and in vivo. Antimicrob Agents Che-
442. Mosher RH, Camp DJ, Yang K, Brown MP, Shaw WV, Vining LC. 1995. mother 52:1513–1515. https://doi.org/10.1128/AAC.00897-07.
Inactivation of chloramphenicol by O-phosphorylation. A novel resis- 460. McCullough PA, Bennett-Guerrero E, Chawla LS, Beaver T, Mehta RL,
tance mechanism in Streptomyces venezuelae ISP5230, a chloramphen- Molitoris BA, Eldred A, Ball G, Lee HJ, Houser MT, Khan S. 2016. ABT-719
icol producer. J Biol Chem 270:27000 –27006. https://doi.org/10.1074/ for the prevention of acute kidney injury in patients undergoing high-
jbc.270.45.27000. risk cardiac surgery: a randomized phase 2b clinical trial. J Am Heart
443. Uchiyama H, Weisblum B. 1985. N-Methyl transferase of Streptomyces Assoc 5:e003549. https://doi.org/10.1161/JAHA.116.003549.
erythraeus that confers resistance to the macrolide-lincosamide- 461. Park-Wyllie LY, Juurlink DN, Kopp A, Shah BR, Stukel TA, Stumpo C,
streptogramin B antibiotics: amino acid sequence and its homology to Dresser L, Low DE, Mamdani MM. 2006. Outpatient gatifloxacin therapy
cognate R-factor enzymes from pathogenic bacilli and cocci. Gene and dysglycemia in older adults. N Engl J Med 354:1352–1361. https://
38:103–110. https://doi.org/10.1016/0378-1119(85)90208-2. doi.org/10.1056/NEJMoa055191.
444. Van Goethem MW, Pierneef R, Bezuidt OKI, Van De Peer Y, Cowan DA, 462. Blum MD, Graham DJ, McCloskey CA. 1994. Temafloxacin syndrome:
Makhalanyane TP. 2018. A reservoir of ‘historical’ antibiotic resistance review of 95 cases. Clin Infect Dis 18:946 –950. https://doi.org/10.1093/
genes in remote pristine Antarctic soils. Microbiome 6:40. https://doi clinids/18.6.946.
.org/10.1186/s40168-018-0424-5. 463. Crowell EL, Koduri VA, Groat RS, Lee DA. 2017. Cost comparison of
445. Duchesne E. 1897. Contribution à l’etude de la concurrence vitale chez commonly used postoperative topical ophthalmic antibiotics. J Cata-
les microorganismes. Antagonisme entre les moisissures et le microbes. ract Refract Surg 43:1322–1327. https://doi.org/10.1016/j.jcrs.2017.07
PhD thesis. Faculté de Médecine et de Pharmacie de Lyon, Lyon,
.028.
France.
464. Wang D, Yang J, Fang M, He W, Zhang Y, Liu C, Zhou D. 2017.
446. Garre C. 1887. Ueber antagonisten unter den Bakterien. Korresponden-
Characterization of the novel In1059 harbouring VIM gene cassette.
zbl Schweiz Aerzte 17:385–392.
Antimicrob Resist Infect Control 6:50. https://doi.org/10.1186/s13756
447. Ruiz J. 2005. La prehistoria de los antimicrobianos, p 41– 44. In Bar-
-017-0204-1.
riobero Neiva JI, Soldevilla Armas A (ed), Un breve viaje por la ciencia.
465. Wei Q, Jiang X, Yang Z, Chen N, Chen X, Li G, Lu Y. 2009. dfrA27, a
Universidad de La Rioja, Logroño, Spain.
new integron-associated trimethoprim resistance gene from Esche-
448. Tiberio V. 1895. Sugli estratti di alcune muffe. An Ig Sper 5:91–103.
richia coli. J Antimicrob Chemother 63:405– 406. https://doi.org/10
449. Emmerich R, Löw O. 1899. Bakteriolytische enzyme als ursache der
.1093/jac/dkn474.
erworbenen immunität und die heilung von infectionskrankheiten
466. Pallecchi L, Riccobono E, Sennati S, Mantella A, Bartalesi F, Trigoso C,
durch dieselben. Z Hyg Infektionskr 31:1–56.
Gotuzzo E, Bartoloni A, Rossolini GM. 2010. Characterization of small
450. Jones HW. 1911. Report of a series of cases of syphilis treated by
Ehrlich’s arsenobenzol at Walter Reed General Hospital. District of ColE-like plasmids mediating widespread dissemination of the qnrB19
Columbia. Boston Med Surg J 164:381–383. https://doi.org/10.1056/ gene in commensal enterobacteria. Antimicrob Agents Chemother
NEJM191103161641103. 54:678 – 682. https://doi.org/10.1128/AAC.01160-09.
451. Domagk G. 1935. Ein beitrag zur chemotherapie der bakteriellen infek- 467. Hering J, Dunevall E, Ek M, Brändén G. 2018. Structural basis for
tionen. Dtsch Med Wochenschr 61:250 –253. https://doi.org/10.1055/s selective inhibition of antibacterial target MraY, a membrane-bound
-0028-1129486. enzyme involved in peptidoglycan synthesis. Drug Discov Today 23:
452. Higuchi S, Onodera Y, Chiba M, Hoshino K, Gotoh N. 2013. Potent in 1426 –1435. https://doi.org/10.1016/j.drudis.2018.05.020.
vitro antibacterial activity of DS-8587, a novel broad-spectrum quino- 468. Li R, Xie M, Lv J, Wai-Chi Chan E, Chen S. 2017. Complete genetic
lone, against Acinetobacter baumannii. Antimicrob Agents Chemother analysis of plasmids carrying mcr-1 and other resistance genes in an
57:1978 –1981. https://doi.org/10.1128/AAC.02374-12. Escherichia coli isolate of animal origin. J Antimicrob Chemother 72:
453. Pucci MJ, Bush K. 2013. Investigational antimicrobial agents of 2013. 696 – 699. https://doi.org/10.1093/jac/dkw509.
Clin Microbiol Rev 26:792– 821. https://doi.org/10.1128/CMR.00033-13. 469. Shen Y, Zhou H, Xu J, Wang Y, Zhang Q, Walsh TR, Shao B, Wu C, Hu Y,
454. Balwan A, Nicolau DP, Wungwattana M, Zuckerman JB, Waters V. 2016. Yang L, Shen Z, Wu Z, Sun Q, Ou Y, Wang Y, Wang S, Wu Y, Cai C, Li J,
Clinafloxacin for treatment of Burkholderia cenocepacia infection in a Shen J, Zhang R, Wang Y. 2018. Anthropogenic and environmental
cystic fibrosis patient. Antimicrob Agents Chemother 60:1–5. https:// factors associated with high incidence of mcr-1 carriage in humans
doi.org/10.1128/AAC.01428-15. across China. Nat Microbiol 3:1054 –1062. https://doi.org/10.1038/
455. Madurga S, Sánchez-Céspedes J, Belda I, Vila J, Giralt E. 2008. Mecha- s41564-018-0205-8.
nism of binding of fluoroquinolones to the quinolone resistance- 470. Wang Y, Zhang R, Li J, Wu Z, Yin W, Schwarz S, Tyrrell JM, Zheng Y,
determining region of DNA gyrase: towards an understanding of the Wang S, Shen Z, Liu Z, Liu J, Lei L, Li M, Zhang Q, Wu C, Zhang Q,
molecular basis of quinolone resistance. Chembiochem 9:2081–2086. Wu Y, Walsh TR, Shen J. 2017. Comprehensive resistome analysis
https://doi.org/10.1002/cbic.200800041. reveals the prevalence of NDM and MCR-1 in Chinese poultry pro-
456. Hata H, Natori T, Mizuno T, Kanazawa I, Eldesouky I, Hayashi M, Miyata duction. Nat Microbiol 2:16260. https://doi.org/10.1038/nmicrobiol
M, Fukunaga H, Ohji S, Hosoyama A, Aono E, Yamazoe A, Tsuchikane K, .2016.260.
Fujita N, Ezaki T. 2016. Phylogenetics of family Enterobacteriaceae and 471. Lascols C, Robert J, Cattoir V, Bébéar C, Cavallo JD, Podglajen I, Ploy MC,
proposal to reclassify Escherichia hermannii and Salmonella subterranea Bonnet R, Soussy CJ, Cambau E. 2007. Type II topoisomerase mutations
as Atlantibacter hermannii and Atlantibacter subterranea gen. nov., in clinical isolates of Enterobacter cloacae and other enterobacterial
comb. nov. Microbiol Immunol 60:303–311. https://doi.org/10.1111/ species harbouring the qnrA gene. Int J Antimicrob Agents 29:402– 409.
1348-0421.12374. https://doi.org/10.1016/j.ijantimicag.2006.11.008.