You are on page 1of 29

Expert Review of Anti-infective Therapy

ISSN: 1478-7210 (Print) 1744-8336 (Online) Journal homepage: https://www.tandfonline.com/loi/ierz20

Extended spectrum β-lactamase producing


enterobacteriaceae: carbapenem sparing options

Abdullah Tarık Aslan & Murat Akova

To cite this article: Abdullah Tarık Aslan & Murat Akova (2019): Extended spectrum β-lactamase
producing enterobacteriaceae: carbapenem sparing options, Expert Review of Anti-infective
Therapy, DOI: 10.1080/14787210.2019.1693258

To link to this article: https://doi.org/10.1080/14787210.2019.1693258

Accepted author version posted online: 13


Nov 2019.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierz20
Publisher: Taylor & Francis & Informa UK Limited, trading as Taylor & Francis Group

Journal: Expert Review of Anti-infective Therapy

DOI: 10.1080/14787210.2019.1693258
Article type: Review

Extended spectrum β-lactamase producing enterobacteriaceae: carbapenem sparing


options

t
Abdullah Tarık Aslan1, Murat Akova*2

ip
cr
1
Hacettepe University Faculty of Medicine, Department of Internal Medicine, Sihhiye, Ankara
06100, Turkey

us
2
Hacettepe University Faculty of Medicine, Department of Infectious Diseases and Clinical
Microbiology, Hacettepe University Sıhhıye Campus, Sihhiye, Ankara 06100, Turkey
an
M
*Corresponding author:

Murat Akova
ed

Hacettepe University Faculty of Medicine

Department of Infectious Diseases and Clinical Microbiology


pt

Hacettepe University Sıhhıye Campus


ce

Sihhiye, Ankara 06100

Phone (312) 305-1279 Fax (312) 310-4179


Ac

Email: makova@hacettepe.edu.tr
Abstract

Introduction: Carbapenems have an important place in our antibiotic armamentarium and have been trusted
to effectively treat infections caused by ESBL-producing Enterobacteriaceae for many years. However, the
utility of carbapenems has been compromised by the emergence of resistance especially in
Enterobacteriaceae. Therefore, carbapenem-sparing alternative antibiotics are of extreme importance in
clinical practice.

Areas covered: We reviewed studies addressing currently available antibiotic options used as both empiric and

t
definitive therapy for the treatment of infections due to ESBL-producing Enterobacteriaceae published in the

ip
PubMed/MEDLINE, Web of Science and Scopus databases without any date restriction. Current treatment
alternatives included beta-lactam/beta-lactamase inhibitor combinations, cefepime, cephamycins,

cr
fluoroquinolones, aminoglycosides, fosfomycin, pivmecillinam, temocillin and, various oral alternative agents.
We also summarized the clinical and molecular epidemiology, early prediction methods and impact of initial

us
empirical therapy and de-escalation approach for ESBL-producing Enterobacteriaceae infections.

Expert opinion: The current literature would endorse the carbapenem utilization for patients with severe and
an
high inoculum-high risk infections. However, for milder infections particularly for urinary tract infections,
various carbapenem-sparing antibiotics can be considered in selected cases. For infections including easily
drainable intra-abdominal infections and catheter-related infections in which catheter removal is readily
M
available more reliable data are needed to recommend non-carbapenem antibiotics confidently.
ed
pt
ce
Ac
Article highlights

• Determining the appropriate empirical therapy for ESBL-producing Enterobacteriaceae infections is a


great challenge in modern era.
• The utility of carbapenems has been compromised by the emergence of resistance especially in
Enterobacteriaceae. Therefore, carbapenem-sparing alternative antibiotics are of extreme importance
in clinical practice.
• Currently, available scientific evidence indicates that type-2 carbapenems remain the best alternatives
for treating severe infections caused by ESBL-producing Enterobacteriaceae. However, this conclusion

t
is mostly derived by observational studies and only one randomized, prospective trial with some

ip
limitations supported this suggestion.

cr
Alternatives to carbapenems in this setting include PTZ, fosfomycin and amikacin and they seem to be
considered only for low risk-low inoculum infections such as UTIs.

us
For infections including easily drainable intra-abdominal infections and catheter-related infections in
which catheter removal is readily available more reliable data are needed to recommend non-
carbapenem antibiotics confidently.

an
Newer BLBLIs (ceftazidime-avibactam and ceftolozane-tazobactam) should be reserved only for
infections with carbapenem -resistant organisms and resistant P. aeruginosa infections respectively.
M
• Oral treatment alternatives merit further studies in community-onset infections caused by ESBL-
producing Enterobacteriacea..
ed
pt
ce
Ac
1. Introduction and global epidemiology of ESBL-producing bacteria

CTX-M type ESBLs in community acquired Escherichia coli and hospital acquired Klebsiella spp., particularly
blaCTX-M-15 have spread across the globe (1). Dissemination of this particular ESBL gene has been enabled by
highly virulent epidemic strains of E. coli (i.e. ST131) and plasmids such as epidemic IncFII plasmids (2,3). These
strains frequently carry multi-drug resistance elements with blaCTX-M-15 including resistance determinants for
fluoroquinolones, trimethoprim sulfamethoxazole (TMP-SMX) and gentamicin (4,5).

According to the 2017 EARS-Net surveillance; population weighted mean rate of the third generation

t
cephalosporin (3rd GC) resistance was 14.9% and 31.2% in invasive isolates of E. coli and Klebsiella

ip
penumoniae, respectively, in EU/EAA countries. More than 80% of isolates in both species were ESBL
producers. The resistance rates were highest in Eastern and Southern European countries as compared with

cr
Northern and Western Europe (6). In the US, the incidence of infections with ESBL-producing organisms have
steadily increased and reached 15% in Klebisellae and 12% in E. coli in 2013 (7,8). In Southeast and East Asia,

us
the detection rates of nosocomial ESBL-positive E. coli isolates were 20–40%, and the rates showed an
increasing tendency in many countries reaching 60–70% in China (9). In long-term care facilities, the ESBL
an
detection rates were reported to be 10–60% in Europe and ~50% in China (10). The ESBL fecal colonization
prevalence in the community was 14% as the global average rate; 4% in Europe, 2% in North America and the
rates were ~46% in Western Pacific (11). The bla CTX-M-15 is currently the most common ESBL gene worldwide
M
(12). The blaCTX-M-14 gene is also frequently detected in certain regions of Europe such as Spain and is prevalent
in East and Southeast Asia (12, 13).
ed

2. Clinical characteristics of infections caused by ESBL-producing bacteria

The World Health Organization prioritized Enterobacteriaceae resistant to the 3rd GCs as one of the most
pt

hazardous pathogens for public health because of easy transmission of ESBL genes leading rapid increase in
prevalence particularly in the community, and limitations in antibiotic choices for treatment (14).
ce

The residential care facilities are very important reservoirs for ESBL-producing pathogens in the community as
a result of various comorbidities, catheter use, greater transmission opportunities and antibiotic overuse
Ac

(15,16). Most of infections caused by these microorganisms involve urinary tract infections (UTIs) (13).

Life-threating bloodstream and intra-abdominal infections have also been encountered particularly with CTX-M
type ESBL-producing E. coli in both community- and in- hospital settings in several countries (17, 18). In
addition to previous exposure to broad spectrum antibiotics leading colonisation with these bacteria,
uncontrolled usage of antibiotics in veterinary medicine and food producing animals are also responsible for
emergence and rapid dissemination of ESBLs (19, 20). Other environmental sources such as urban
wastewaters, contaminated drinking water and spreading via international travel have been identified as
alternative acquisition modes of ESBL genes in the community (21-23).
3. Early prediction methods, impact of initial empirical antimicrobial therapy and de-escalation

Infections caused by ESBL-producing organisms are associated with high rate of mortality (24-27), prolonged
infection-related hospital stay (14,28,29) and increased healthcare associated costs (30-33). Therefore,
selecting appropriate antibiotic therapy in early empirical period is highly important but turns to be a challenge
in face of increased prevalence of antimicrobial resistance. Although some studies asserted no significant
impact of inappropriate initial empirical antimicrobial therapy on mortality (34,35), many others consistently
demonstrated otherwise (36-39). This controversy may be partly explained by the resistance profile of the
offending pathogen, the source and severity of infections, (not) achieving appropriate source control, baseline

t
ip
comorbidities and place of acquisition of infection (e.g. community-onset vs hospital-acquired).

Several ESBL-prediction scores have been developed and the decision analysis usually includes factors such as

cr
previous infection or colonization, age, recent hospitalization, presence of co-morbidities and previous
exposure to antibiotics (40,41). Using these scores may improve appropriateness of empirical antimicrobial

us
therapy and reduce carbapenem utilization. However, in the wake of an era with emerging community-
acquired ESBL-related infections, the predictability of these infections has further decreased (42).
an
Two different researchers investigated the effects of transition to oral step-down therapy between 2-5 days of
bacteremia and early de-escalation of therapy for infections due to Enterobacteriaceae, respectively. Both
M
found that oral step-down therapy and early de-escalation were not associated with higher rates of 30-day
mortality and clinical failure compared to controls. Switch to oral step-down therapy shortened the duration of
hospitalization significantly (43,44). These findings might indirectly suggest that the empirical therapy is
ed

important for clinically relevant outcomes such as mortality but definitive therapy is particularly related with
ecological impact and cost.
pt

4. Carbapenem sparing therapeutic options for infections caused by ESBL-producing bacteria

A systematic literature search using PubMed/MEDLINE, Scopus and Web of Science databases was performed
ce

without any date limitation. Key terms included ESBL or extended-spectrum β-lactamases, bacteremia,
bactereamia, bloodstream infection, urinary tract infection, complicated urinary tract infection, intra-
Ac

abdominal infection, biliary tract infection, sepsis, septic shock, pneumonia, high inoculum infection, low
inoculum infection, piperacillin-tazobactam, ciprofloxacin, levofloxacin, quinolones, fluoroquinolones, cefepime,
cephamycins, cefmetazole, cefoxitin, flomoxef, moxalactam, carbapenem, doripenem, meropenem,
ertapenem, fosfomycin, aminoglycosides, amikacin, plazomicin, mecillinam/pivmecillinam, amoxicillin-
clavulanate, cefoperazon-sulbactam, trimethoprim-sulfamethoxazole, nitrofurantion, ceftazidime-avibactam,
ceftolozane-tazobactam and temocillin. Full text articles in English language were considered. Articles were
screened by title and abstract for possible inclusion, and references within articles of interest were scanned to
capture additional sources. Priority was given for clinical studies, randomized controlled trials and meta-
analyses published after 2011.
4.1. Cephamycins

Cephamycins (e.g. cefoxitin, cefotetan, flomoxef, moxalactam and cefmetazole) appear to be a reasonable
option against ESBL-producing organisms owing to sufficient in vitro activity (45). However, clinical data
evaluating cephamycins for ESBL-producing Enterobacteriaceae are limited and usually included few patients.
In a multicenter retrospective cohort study, Matsumura et al. (46) compared patients receiving cefmetazole or
flomoxef with those given carbapenems for a bloodstream infection (BSI) with ESBL-producing E. coli.
Propensity score adjusted analysis showed that therapy with cefmetazole or flomoxef were not associated with
higher rate of mortality in empirical (adjusted HR, 0.87; CI, 0.11 to 6.52) and definitive treatment cohorts as

t
compared to those receiving a carbapenem (adjusted HR, 1.04; CI, 0.24 to 4.49). Clinical success rates were also

ip
similar for both treatment groups. However, there were some important limitations; (i) urinary tract infection
(UTI) as a bacteremia source was significantly more frequent in cephamycin group and primary bacteremia was

cr
more common in carbapenem group, (ii) cefmetazole and flomoxef were evaluated as a combined treatment
group. Thus, we cannot discriminate the activities of cefmetazole and flomoxef from each other, (iii) SOFA

us
score was significantly higher in those treated with carbapenems. Also, severe sepsis and septic shock were
more frequent in carbapenem receiving patients but this difference did not reach statistical significance (27%
an
vs 47% p:0.13), (iv) Patients receiving cephamycins were infected with highly susceptible pathogens (MIC50≤
1ug/ml for both cefmetazole and flomoxef), (v) the number of patients included in treatment arms were quite
few to provide a statistical power. Thus, the outcomes obtained from this study cannot be evaluated as a
M
robust evidence for adequacy of cephamycins in BSIs due to ESBL-producing Enterobacteriaceae. In another
retrospective single center study, Fukuchi and colleagues (47) found that 30-days mortality rates of patients
with BSIs caused by Enterobacteriaceae, mostly E. coli (93%), were similar for cefmetazole and a carbapenem
ed

when used as definitive therapies. This study had also similar limitations with the previous study. In addition,
the authors did not investigate the effects of confounding factors on study outcome. Other studies
demonstrated high rate of cefmetazole resistance (40%) in ESBL-producing Enterobacteriaceae isolates
pt

particularly having blaCTX-M-14 and blaCTX-M-15 genes (48), and significantly higher sepsis related mortality rates in
patients treated with flomoxef than those treated with a carbapenem (49). Furthermore, the combined
ce

expression of ESBLs and AmpC diminish susceptibility rates to many antibiotics involving cephamycins,
extended spectrum cephalosporins, β-lactam/β-lactamase inhibitor combinations, and even carbapenems (50).
AmpC is usually encoded by chromosomal genes, however acquisition of plasmid mediated AmpC is not
Ac

uncommon (51). Furthermore, cephamycins are not readily available for routine use in many countries. When
one considers these data in addition to the fact that optimum doses of cephamycins are not defined for various
type of infections and for varying MIC levels, these antibiotics might only be recommended for definitive
treatment of non-severe UTIs caused by ESBL-producing Enterobacteriaceae.

4.2. Cefepime

Cefepime, an oxyimino-cephalosporin, has enhanced activity against Enterobacteriaceae since it resists to


degradation by Amp-C β-lactamases and slightly more resistant against ESBLs (except CTX-M-10 and TEM-10)
than other cephalosporins (52). However, the activity of cefepime can be impaired by high inoculum infections
(53-56). The main pharmacokinetic/pharmacodynamic (PK/PD) target of cefepime, similar to other β-lactams, is
drug levels above the MIC for approximately 40-50% of dosing interval (57,58). Thus, rapid bacterial recovery
occurs when antibiotic concentrations fall below the MIC of the antibiotic. A study demonstrated that this
target attainment with 1-2 gr bid cefepime among gram negative organisms can be reached in less than 30% of
cases (59). Another study showed that if 67% fT>MIC is accepted as the target, the probability of target
attainment was achieved 90%, 80% and 45% for MICs of 2, 4 and 8 μg/mL respectively, with standard cefepime
dosing (i.e. 2 g bid) (60). In a retrospective cohort analysis, mortality rates for individuals treated with cefepime

t
1-2 g bid at an MIC 8 mg/L and >8 mg/L were 56.3% and 53.3%, respectively (61). These studies indicate that

ip
the success of cefepime therapy is possibly dictated by the low MICs and availability of target attainment of
serum concentrations of cefepime.

cr
The results from published studies comparing the efficacy of cefepime with carbapenems for the treatment of

us
ESBL-producing Enterobacteriaceae infections produced conflicting results. In a retrospective observational
study, sepsis related mortality rates due to monobacterial BSIs with ESBL-producing E. coli, K. pneumoniae or
Enterobacter cloacae were higher with cefepime than those treated with a carbapenem (47.1% vs. 11.9%
an
P=0.002). Thirty-day mortality rates were also higher in the cefepime group (58.8% vs 17.9%, P=0.001). In
multivariate analysis, definitive cefepime therapy was found as an independent risk factor for 30-day mortality.
Moreover, patients with a BSI caused by an isolate with cefepime MIC ≤1 mg/L had more clinical success and
M
less sepsis-related mortality as compared with those isolates with cefepime MIC 2-8 mg/L (62). In another
retrospective study, MIC of cefepime was identified as a significant prognostic factor for bacteriological
eradication but not for clinical failure. The possible explanation of this conflicting results is that
ed

aminoglycosides or fluoroquinolones were invariably administered with cefepime in the second study (63).
Furthermore, clinicians should be aware of the risk pertaining with unacceptably high rate of major error of
pt

cefepime susceptibility test result against ESBLs producers with Vitek2 microdilution method (64).

A retrospective cohort study with BSIs caused by ESBL-producing E. coli in patients with leukemia and
ce

hematopoietic stem cell transplantation explored efficacy of carbapenems, cefepime or piperacillin-


tazobactam. 14-day mortality rates were observed as 8%, 0% and 19% in cefepime, piperacillin-tazobactam and
Ac

carbapenem groups, respectively. In multivariate analysis, empiric treatment with cefepime was not associated
with increased 14-day or 30-day mortality (65). Although other studies showed evidence in the affirmative of
the results in previously mentioned studies, several methodological errors and low number of patients
analysed in these trials would prevent from constructing causal relationships (66-68).

Overall, administration of cefepime for severe infections caused by ESBL producers cannot be safely
recommended. However, one can consider prescribing cefepime regimen of 2 g bid or tid with prolonged
infusion (i.e. 3-4 hours) for non-severe infections (e.g. UTIs, non-bacteremic skin and soft tissue infections) with
ESBL producers with low cefepime MIC values. But even in such cases some caution would be appropriate.
4.3. β-lactam/β-lactamase inhibitor (BLBLI) combinations

Piperacillin-tazobactam (PTZ) has a good in vitro activity against ESBL-producing E. coli, but K. pneumoniae are
usually less susceptible (69-71). High inoculum infections such as bacteremia, pneumonia and osteoarticular
infections might adversely affect the activity of BLBLIs (55, 72). While amoxicillin-clavulanate (AMC) was able to
sustain bactericidal activity against ESBL-producing E. coli for 24 hours in case of high bacterial load, PTZ had a
substantial inoculum effect for both ESBL-producing and non-ESBL producing E. coli (73).

The PK-PD models proved a 99% success rate of achieving the target (time above the MIC 50%) against ESBL

t
producers by using 4.5 gram qid if the MIC of the isolate is ≤ 8 mg/L. However, success rate diminished to 57%

ip
when the MIC is 16 mg/L (74). Piperacillin-tazobactam dosage is particularly important in critically ill patients
since these patients often show different PKs through changes in key variables (e.g. renal clearance, increased

cr
capillary permeability, hypoalbuminemia and increased volume of distribution) (74). In many studies,
prolonged or continuous infusion of PTZ was found to be associated with better results in critically ill patients

us
and gram negative infections (76- 78) than those with standard usage, although studies challenging these
results were also published (79). an
Piperacillin-tazobactam did not show the same bactericidal activity to ESBL-producers and non-ESBL-producers
despite the presence of the same MIC (56). MICs of BLBLIs are dependent on type of ESBL (SHV group is more
resistant), porin mutations, the amount of β-lactamase produced, the rate of enzyme resynthesis, concomitant
M
expression of AmpC, presence of early OXA-type enzymes (e.g. OXA-1), simultaneous expression of multiple
type of ESBLs, inhibitor resistant ESBL co-production and methods used for MIC determination (80). In a survey
ed

including ESBL-producing isolates of Enterobacteriaceae from 72 US hospitals, PTZ susceptibility rates were
67.2%, 92.9% and 45.8% for isolates having CTX-M-15-like and CTX-M-14-like ESBLs and SHV-type enzymes,
respectively (81). Therefore, knowing molecular epidemiology of ESBLs and local resistance patterns might be
pt

useful before deciding empirical therapy with PTZ.

Several observational studies were published comparing PTZ and carbapenems as empirical or definitive
ce

therapy for infections caused by ESBL-producing Enterobacteriaceae with conflicting results.

In a post hoc analysis of six Spanish prospective cohort studies, authors investigated BLBLIs (AMC and PTZ) for
Ac

both the empirical and definitive treatment of ESBL-producing E. coli bacteremia. Mortality rates were similar
in both therapy groups receiving BLBLIs or carbapenems (82). Predominant type of ESBL was CTX-M-14 and the
prevalence of AmpC beta-lactamase production in Enterobacteriaceae isolates was reported as <5% in Spain at
that time (83). Main sources of bacteremia were low inoculum infections (e.g. UTIs) or infections in which
bacterial load can be reduced efficiently with surgical interventions (e.g. cholangitis) and >90% of patients were
treated with 4.5 gram PTZ qid. Mortality at 30 days in patients empirically treated with PTZ 4.5 grams qid was
only 4.5 % when the MIC was ≤4 mg/L, and 23% if the MIC was >4 mg/L (82). Similar results observed with 39
patients having bacteremia with ESBL- producing E. coli and treated with PTZ, empirically. None of the patients
with bacteremia originated from urinary tract were dead irrespective of PTZ MICs. However, in patients having
bacteremia from a non-urinary tract source, lower mortality rate was significantly associated with lower MIC (≤
2 mg/L). Of note, low MIC group mostly included patients with intra-abdominal infection in whom, source
control with surgical intervention might be a critical confounding factor (84). In contrast, Tamma et al. (85)
reported significantly lower 14-day mortality rate with empirical meropenem therapy than with PTZ empirical
therapy. These conflicting results may be due to the type of infections treated in different trials where efficacy
of PTZ may differ. Tazobactam is mainly excreted in the urine (86). Therefore, high concentrations of PTZ in
urine can kill ESBL-producing E. coli more efficiently since inoculum effects of gram negative organisms are low
in urinary tract infections. CTX-M type ESBLs can be inhibited more efficiently with tazobactam than SHV and
TEM type ESBLs which are encoded more frequently by Klebsiella spp (87). High dose PTZ (i.e. 4.5 gram qid)

t
ip
would help to attain PK/PD target (88). Thus, the failure of PTZ in the study by Tamma et al. (85) could be
explained by the fact that the study included bacteremia cases caused by E. coli, K. pneumoniae, Klebsiella

cr
oxytoca and Proteus mirabilis. Most of the patients were treated with 3.375 gram PTZ qid and only 39% of
patients received 4.5 gram of PTZ qid. Source of bacteremia cases were central line associated in 44%. The

us
second most common source was pneumonia which imply a high inoculum of bacteria in a compartment where
penetration of antibiotics may be impaired (85). Also, distribution of MIC values for PTZ were as follows:
2μg/ml (1%), 4μg/ml (39%), 8μg/ ml (46%), and 16μg/ml (14%). As compared with the study by Rodríguez-
an
Baño et al. (82), the MIC values of PTZ were higher in isolates included in the study by Tamma et al. (85).

In a multicenter, multinational, retrospective study (INCREMENT-ESBL), 365 and 601 patients were analysed in
M
an empirical and a targeted treatment cohort, respectively. CTX-M-, SHV- and TEM-type ESBLs were produced
in 77.3%, 10.6% and 12.1% of isolates, respectively. In the empirical therapy cohort, there were no difference
between BLBLIs and carbapenem groups in terms of 14-day clinical cure/improvement and 30-day crude
ed

mortality rates. In the targeted therapy cohort, the cure/improvement rates and 30-day mortality rates with
BLBLIs and carbapenems were 90.2% vs. 85.5% and 9.8% vs. 13.9%, respectively. The adjusted odds ratio (OR)
was 1.61 (95% confidence interval [CI]) value 0.58-4.86) for cure/improvement and 0.59 (0.19-1.71) for 30-day
pt

crude mortality. No difference can be detected between AMC and PTZ against carbapenems in subgroup
analysis. Only 11% of patients admitted to ICU, the most of the microorganisms were isolated from urinary or
ce

biliary sources and the vast majority of patients (83%) received 4.5 gram PTZ qid (89).

The outcomes in patients with BSIs from the original INCREMENT cohort revealed that 30-day mortality rate
Ac

was significantly higher in patients with ESBL-producing K. penumoniae BSI than ESBL-producing E. coli BSI
(33.7% vs 17.4% OR: 1.64, p=0.016). Seventy six per cent of 286 isolates in which PCR analyses for ESBL genes
were detected had CTX-M type ESBLs. Clinical characteristics and 30-day mortality rates were similar in CTX-M
and non-CTX-M groups (90). Similarly, in a single center Italian study; BSIs with ESBL-producing Proteus spp
were also found as an independent risk factor for 30-day mortality (91).

Gudiol et al. (92) conducted a multicentric retrospective cohort study involving neutropenic patients with an
hematological malignancy. One hundred and seventy four patients in empirical treatment cohort and 251
patients in definitive treatment cohort were included. In both crude analysis and propensity score matched
analysis, there were no significant difference in 30-day case fatality rates and in other secondary outcomes. In
propensity score matched empirical and definitive therapy cohorts, the 30-day mortality rates were 20% and
11% for the former and 13% and 7% for the latter for the BLBLIs and carbapenem groups, respectively. The
most common source of BSIs was endogenous and followed by catheter-related BSIs and urinary tract
infections.

Ng et al (93) conducted a multicentre retrospective cohort study in patients who had ESBL-producing E. coli and
K. pneumoniae bacteraemia treated with PTZ or a carbapenem. After adjusting for confounders, empiric PTZ
administration was not associated with increased 30-day mortality rate (OR 1.00, 95% CI; 0.45-2.17).

t
ip
Furthermore, empirical use of PTZ caused fewer multi-drug resistant bacterial and fungal infections as
compared with a carbapenem.

cr
A meta-analysis of 21 studies compared carbapenems with other alternatives for the treatment of bacteremia
with ESBL-producing Enterobacteriaceae. Mortality was not increased with BLBLIs in both empiric (relative risk

us
[RR]:0.91, 95% CI, 0.66-1.25) and definitive therapies (RR:0.52, 95% CI, 0.23-1.13) as compared with
carbapenems. However, the mortality rate was higher in patients treated with non-BLBLI carbapenem-sparing
an
options than those treated with a carbapenem alone in both therapy groups. But one should be aware that
several observational studies included in this meta-analysis did not perform multivariate and propensity score
analyses to eliminate effects of other confounders and potential bias in the selection of antibiotics. Also, many
M
patients who were treated with BLBLIs in empirical treatment period received a carbapenem as a definitive
therapy (94).
ed

Another two systematic reviews and meta-analyses revealed similar results by comparing BLBIs and
carbepenems in terms of mortality in BSIs caused by ESBL-producing Enterobacteriaceae (95,96).
pt

One possible advantage of prescribing PTZ may be reduction of selection pressure on ESBL producers. Several
investigators reported substantially lower frequency of ESBL-producing Enterobactericeae in patients with
ce

previous exposure to PTZ compared to oxyimino β-lactams and fluoroquinolones (4,97). Mclaughlin et al. (98)
also showed that PTZ was associated with lower rate of carbapenem resistance in Enterobactericeae spp. than
type-2 carbapenems . However, others pointed out that excessive use of PTZ can facilitate acquisition of OXA-
Ac

type β-lactamases and inhibitor-resistant TEM genes (99). Piperacillin-tazobactam might also exert broader
selective pressure than ertapenem given the anti-pseudomonal coverage of the former.

A recent multinational, multicenter, parallel group, randomized, prospective, non-inferiority clinical trial
(MERINO) included 391 participants from 26 centers in 9 countries and found that the 30-day mortality rate for
patients with ceftriaxone-nonsusceptible E. coli or K. pneumoniae BSIs treated with PTZ was 12.3%, compared
with 3.7% for patients who were treated with meropenem as definitive therapy (100). The difference in the 30-
day mortality rate exceeded the non-inferiority margin of 5%. Therefore, the authors recommended against
the use of PTZ to treat ceftriaxone-nonsusceptible E. coli or K. pneumoniae BSIs. However, there have been
several criticisms for this study that deserve mention here (101-103): There were imbalances in study arms
favouring meropenem (i.e. more UTIs in meropenem arm, cases with quick SOFA score >2 were more frequent
in PTZ arm). Randomization occurred after a 48-h empirical therapy and in 40.7% of enrolled patients the
infection was already partially resolved at the time of enrolment. A substantial proportion of patients
randomized to carbapenem arm received PTZ or other antibiotics and vice versa. Adequate source control was
not described, although, if present, it could affect mortality. All deaths were not associated with primary
infection but rather related with underlying comorbidities (mainly metastatic cancers). The geographical
differences in resistance rates would also have affected the results, as blaSHV- producing Enterobacteriaceae

t
have been common in many countries where the trial was conducted which accounted for 8 of the 23 deaths

ip
(35%) in the PTZ arm. Nonsusceptibility to PTZ might be higher than identified as 10% and 67% of isolates had
ampC and blaOXA-1 genes, respectively. Finally, a narrower-than-usual non- inferiority margin (i.e. -5% vs -15%

cr
used in similar trials) might have adversely affected non-inferiority of PTZ.

us
Cefoperazone-sulbactam is another BLBLI combination and has been widely used in Asia particularly in China
for the treatment of infections caused by Enterobacteriaceae. However; very limited clinical studies have been
published until now to use for ESBL producers (104). Additional data are needed for any suggestion in clinical
practice with this agent.
an
In conclusion, current available data indicate that type-2 carbapenems (meropenem or imipenem) seem to be
M
the preferred choice of therapy for high inoculum infections caused by ESBL-producing Enterobactericeae. The
role of PTZ, if any, in such infections would require further controlled studies for definitive therapy. Piperacillin
tazobactam can be suggested with appropriate dose and infusion regimens if MIC value of the culprit
ed

microorganism is low and adequate source control is provided or in those clinically stable patients with low
inoculum infections.
pt

4.4. Temocillin
ce

Temocillin, the 6-α-methoxy derivative of ticarcillin, which is active against ESBL- and derepressed AmpC-beta-
lactamases can be considered as a potential carbapenem-sparing agent (105). Optimal dose of temocillin (2
gram bid) was significantly associated with favourable outcomes in limited number of observational studies
Ac

(106). The fact that temocillin is only licensed in England, Belgium and France currently limits its use. Large
scale, randomized studies are required to establish the efficacy of this drug against ESBL-producing
Enterobacteriaceae among different type of infections.

4.5. Fosfomycin

Fosfomycin is a bactericidal, low molecular weight, broad-spectrum antibiotic and exerts its bactericidal activity
against several multi-drug resistant gram negative bacteria by irreversible inhibition of early steps of
peptidoglycan synthesis (107). Fosfomycin, primarily excreted by kidneys and has very good tissue penetration
in soft tissues, lungs, bone, cerebrospinal fluid, and heart valves (108). Especially for the urinary tract, the drug
achieves high concentrations for a prolonged period of time. Optimal PK/PD index has not been determined,
yet. During monotherapy with fosfomycin, rapid resistance may develop. In vitro studies also demonstrated
appearance of resistant subpopulations within 30-40 h of drug exposure (107). Therefore, other than acute
cystitis the drug should be used in combinations for treating any other infections. The fosA3 gene which is
responsible for plasmid-mediated fosfomycin resistance is usually co-harboured with blaCTX-M gene in E. coli
thus compromising the efficacy of the drug against ESBL producers (109). Bi et al (109) from China
demonstrated fosfomycin resistance rate among ESBL-producing E. coli as 6.7%. Seventy per cent of these
isolates acquired fosfomycin resistance via plasmid-mediated fos A3 gene. On the other hand, frequencies of

t
ip
chromosomal mutations in both genes of mur A and glpT-uhpT membrane transporters were 12.5% in the
same E. coli isolates. Lu et al. (110) reported that fosfomycin resistance rate among ESBL-producing K.

cr
pneumoniae isolates were 27.8%. Among fosfomycin resistant K. pneumoniae isolates, mutations rates in
murA and membrane transporter genes were 70% and 97%, respectively. Fosfomycin resistance can also be

us
affected by the in vitro sensitivity method used. Approved MIC detection method is agar dilution using agar
media supplemented with 25 ugr/ml of glucose-6-phosphate (111). Resistance rates are higher in studies in
which disk diffusion or microbroth dilution susceptibility testing were used than in studies with a reference
an
agar dilution (112).

In a randomized controlled trial fosfomycin was non-inferior to piperacillin-tazobactam in complicated UTIs


M
including some ESBL-producing Enterobacteriaceae (113). Oral formulation (fosfomycin trometamol) has been
used to treat cystitis (114) or chronic bacterial prostatitis due to ESBL-producing Enterobacteriaceae (115). In a
retrospective single institution study, fosfomycin and ertapenem were compared as step down therapy in
ed

outpatients with UTI caused by ESBL-producing Enterobacteriaceae and 30 day re-admission/re-visit rates were
evaluated as primary outcome (116). Fosfomycin was primarily administered as 3 gram/every 72 hours (62%)
or 3 gram/every 48 hours orally (23%). There were no difference between fosfomycin and ertapenem in terms
pt

of 30 day re-admission/re-visit rates. Şenol et al. (113) compared fosfomycin vs carbapenems


(imipenem/cilastatin or meropenem) for complicated UTIs caused by ESBL-producing E. coli. Twenty seven
ce

patients were treated with 3 gram fosfomycin orally every other day for 3 doses and 20 patients with
conventional doses of imipenem/cilastatin or meropenem. Clinical success and relapse/re-admission rates
Ac

were similar in the two treatment groups.

Fosfomycin therapy can be recommended for uncomplicated UTIs with ESBL-producing E. coli as a step-down
outpatient therapy. However, complicated UTIs, prostatitis, UTIs with K. pneumoniae would require more data
from well-designed studies. A randomized clinical trial (“FOREST”) comparing the safety and efficacy of
fosfomycin versus meropenem in bacteremic UTIs caused by ESBL-producing E. coli have been ongoing (117).

4.6. Fluoroquinolones
Fluoroquinolones appear to have lost a great part of their activity against Enterobacteriaceae prior to the
emergence of contemporary ESBL types such as CTX-M-15 in specific molecular epidemiological investigations
(118). These early evolutionary changes that were seen in Enterobacteriaceae in 1980s are still prevailing in
most E. coli and K. pneumoniae strains today (119,120). In parallel with in vitro susceptibility studies, empirical
therapy with carbapenems was found superior for treatment outcomes than fluoroquinolones in many clinical
studies (121- 123). Nonetheless, it may be noteworthy reviewing its efficacy in susceptible isolates. Tumbarello
et al. (124) demonstrated that, 8 of the 16 patients with bacteraemia caused by ESBL-producing
Enterobacteriaceae who were treated with ciprofloxacin, died. Among these patients, the ciprofloxacin MIC
was 0.5-1 mg/L. In contrast to this study, a meta-analysis showed that fluoroquinolones were associated with

t
ip
increased mortality compared with carbapenems in empirical treatment, but not as a targeted therapy (in
infections with susceptible isolates) (94). Thus, these drugs cannot be recommended for the treatment for

cr
severe infections caused by ESBL-producing Enterobacteriaceae. However, fluoroquinolones may be
considered in targeted therapy or in oral de-escalating therapy for mild infections (e.g. UTIs) caused by

us
quinolone-susceptible ESBL-producing Enterobacteriaceae with limited supporting data.

4.7. Aminoglycosides
an
Aminoglycosides are other potential options as carbapenem-sparing therapy for UTIs in monotherapy and in
monotherapy or combination therapy of intra-abdominal infections (125). A study from Korea evaluated in
M
vitro activities of 21 different antimicrobial agents alone or in combination with aminoglycosides against ESBL-
producing E. coli isolates. Among 291 isolates, amikacin resistance was 11.7% in contrast 51.9% for gentamicin.
Importantly, combination of amikacin with PTZ elevated PTZ susceptibility rate from 57.0% to 92.8% (126). In
ed

some clinical studies, aminoglycosides were reported as a reasonable carbapenem-sparing options for UTIs
with ESBL-producing Enterobacteriaceae (127-129). Enterobacteriacea have developed resistance to
aminoglycosides by various mechanisms among which the most common is enzymatic modification by
pt

aminoglycoside-modifying enzymes (AMEs). These enzymes can be frequently co-produced by various ESBLs,
but with mostly CTX-M types on the same plasmid (130). A novel aminoglycoside, plazomicin, derived from
ce

sisomicin is an appealing alternative antibiotic against multidrug-resistant organisms (131). Plazomicin is


completely resistant to inactivation by AMEs (132). Also, it is currently approved for complicated UTIs caused
by plazomicin-susceptible gram-negative bacteria including those with carbapenem resistance.
Ac

4.8. Mecillinam/Pivmecillinam

Pivmecillinam is a pro-drug of mecillinam which is an amidinopenicillin with selective activity against gram-
negative bacteria. Raja et al (133) reported pivmecillinam susceptibility rate against ESBL-producing
Enterobacteriaceae as >95%. In another series, this rate was found as 85% (134). In a Norwegian prospective
multicenter observational study, 400 mg tid pivmecillinam therapy was equally effective against community
acquired UTIs caused by ESBL-producing E. coli vs. non-ESBL-producing strains (135). However, 200 mg tid
regimen was associated with significantly higher clinical failure rate when it was administered ≤ 5 days in
patients infected with ESBL (+) E. coli. Other small scale studies produced similar results for this indication
(136,137). Unfortunately, mecillinam/pivmecillinam is not available in several countries.

The use of pivmecillinam can be considered in lower UTI with ESBL-producing Enterobacteriaceae but should
not be utilized in high inoculum infections because of absence of supporting data.

4.9. New β-lactam/β-lactamase Inhibitor (BLBLIs) Combinations

Ceftolozane is particularly active against P. aeruginosa but can be degraded by ESBL enzymes. Combination

t
with tazobactam extends its activity against ESBLs including TEM, SHV, CTX-M types (138,139). In vitro

ip
susceptibility rates for ESBL-producing E. coli and K. pneumoniae were reported as 92.8% and 73.5%,
respectively (140). Pooled analysis of two double-blind, randomized, controlled trials (ASPECT-cUTI) and

cr
(ASPECT-cIAI) compared efficacy of ceftolozane-tazobactam with levofloxacin and meropenem, respectively. In
vitro susceptibility rates for ESBL-producing E. coli and K. pnemoniae were 95% and 56.7% according to FDA

us
criteria, respectively. The clinical cure rates were 97.4% (76/78) for ceftolozane-tazobactam [ESBL-E. coli,
98.0%; ESBL-K. pneumoniae, 94.4%], 82.6% for levofloxacin and 88.5% for meropenem (141). The results may
be supportive for ceftolozane-tazobactam to be used in these infections as a carbapenem sparing drug.
an
Ceftazidime-avibactam is a combination of ceftazidime with a novel non-BLBLI within the diazabicyclooctane
chemical class and protects ceftazidime from hydrolysis by class A (TEM, SHV, CTX-M, KPC), class C (AmpC) and
M
some class D (OXA) beta-lactamases (142,143). Ceftazidime-avibactam is usually more active in vitro against
ESBL producers than ceftolozane-tazobactam. (144). However, so far there has been no clinical study
ed

investigating clinical efficacy of this combination for infections caused by ESBL-producing Enterobacteriaceae.
For antimicrobial stewardship purposes, this combination should be reserved for carbapenem-resistant Gram-
negative bacteria (141).
pt

4.10. Oral antibiotic alternatives of carbapenems for the treatment of ESBL-producing Enterobacteriaceae
infections
ce

Increasing prevalence of infections with community-onset ESBL-producing Enterobacteriaceae revived


Ac

forgotten old antibiotics as possible candidates for these infections, particularly in UTIs. In some clinical and in-
vitro susceptibility studies, fosfomycin, pivmecillinam and nitrofurantion were evaluated against ESBL-
producing isolates and infections with these pathogens. Fosfomycin is licenced for intravenous use only in a
few countries. Its main limitation is the development of overt resistance or hetero-resistance during treatment.
Use of fosfomycin in clinical setting was described above in detail. However, there is currently no convincing
data about the efficacy and safety of fosfomycin for the treatment of severe infections and/or infections with
high bacterial loads even with iv formulation.

In a study with 238 eligible cases, E. coli (83.6%) was the most common causative pathogen. Sixty patients
received oral antimicrobial treatment after a median of four days of appropriate iv therapy, and 178 patients
completed treatment with iv therapy. Fluoroquinolones (58.3%) were the most commonly prescribed oral
agents, followed by TMP-SMX and AMC. Oral antimicrobial treatment was not associated with treatment
failure (adjusted OR 0.66; 95% CI 0.18–2.44) and hospitalization length was shorter in the oral treatment group
(6.2 days versus 10.7 days; P < 0.01). Acute pyelonephritis recurrence caused by ESBL-producing
Enterobacteriaceae infection within 2 months was not associated with oral antimicrobial treatment (adjusted
HR 0.56; 95% CI 0.16–2.00). Using oral agents reduced hospital stay without adverse effects on treatment
outcome. However, the number of cases was insufficient to analyse the clinical efficacy of each oral
antimicrobial agent individually (145).

t
ip
As previously mentioned, fluoroquinolone resistance is highly prevalent among ESBL-producing
Enterobacteriaceae and only few data available for treating infections with these bacteria. Targeted therapy for

cr
infections caused by susceptible isolates could be an option with these antibiotics. In future studies,
fluoroquinolones should be evaluated in definitive treatment of infections caused by susceptible isolates as an

us
oral switch option, since there are few specific data about their effectiveness (146). Furthermore, recent
cautionary data about fluoroquinolone toxicity should be considered while prescribing fluoroquinolones (147).
an
Regarding TMP-SMX, although some ESBL isolates may be susceptible, co-resistance against TMP-SMX is quite
common and there are no studies in the literature that have been analysed this treatment option.
M
Amoxicillin-clavulanate is a good option for susceptible isolates albeit susceptibility rate is low in many
countries (82). As a favourable feature, it does not suffer from the inoculum effect (73), and it may be used for
oral switch treatment. But, again more robust data are required to recommend oral treatment with AMC for
ed

infections with ESBL-producing Enterobacteriaceae.

Nitrofurantoin is active in-vitro against common causes of UTI including E. coli and Citrobacter spp.. However,
pt

Klebsiellae and Enterobacter spp. are less susceptible. Nitrofurantoin suggested as a preferred drug in an
international consensus guideline for UTI in 2010 (148). It may lead to major hepatic and pulmonary toxicity,
ce

but the true incidence is unclear (149). For the short-term treatment of uncomplicated urinary tract infection in
otherwise healthy young women, nitrofurantoin is a safe and effective choice. However, reliable clinical data to
utilize nitrofurantoin for UTIs with ESBL-producing Enterobacteriaceae are lacking.
Ac

5. Conclusion

Currently, available scientific evidence indicates that type-2 carbapenems remain the best alternatives for
treating severe infections caused by ESBL-producing Enterobacteriaceae. However, this conclusion is mostly
derived by observational studies and only one randomized, prospective trial with some limitations supported
this suggestion. Alternatives to carbapenems in this setting include PTZ, fosfomycin and amikacin and they
seem to be considered only for low risk-low inoculum infections such as UTIs. On the other hand
fluoroquinolones and cefepime appear to be inappropriate options as carbapenem-sparing antibiotics.
Additional data are needed to establish the exact role of cefoperazone-sulbactam and temocillin which are not
widely available molecules. The new BLBLI combinations such as ceftolozane-tazobactam and ceftazidime-
avibactam should not be used as primary agents for these infections.

6. Expert opinion

Extended spectrum beta-lactamase producing gram-negative bacteria are frequent culprits in different types of
infections in clinical practice worldwide. Although the data are limited to determine the best available therapy
for infections caused by these pathogens, type-2 carbapenems seem to be the best option. The available

t
ip
clinical evidence dictates that carbapenem-sparing antibiotics should not be administered for high inoculum
infections, critically ill (e.g. septic shock) and immunocompromised patients. Also, level of evidence is

cr
insufficient to offer alternative antibiotics to carbapenems for non-biliary intra-abdominal infections and
catheter-related infections. ESBL-producing organisms are frequently resistant to non-beta-lactam antibiotics

us
including fluoroquinolones, aminoglycosides and TMP-SMX due to co-expression of resistance determinants
against these antibiotics. Another important threat to public health is the rapid dissemination of ESBL genes in
the community and increasing prevalence of community-onset infections caused by ESBL-producing organisms.
an
Not only UTIs but also BSIs and other invasive infections can be seen in the community setting with these
bacteria. International travel, urban wastewaters, food animals, companion animals and contaminated drinking
waters have been identified as possible sources for transmission of ESBL-producing organisms to humans in the
M
community. Predictability of infections caused by ESBL producers has been further mitigated due to
widespread dissemination of these organisms in the community. Therefore, administration of appropriate
empirical therapy for these infections is a great challenge in modern era. The following suggestions for future
ed

research should be taken into account for ameliorating the problem caused by ESBL-producing bacterial
infections:
pt

1- Rapid diagnostic methods should be explored to shorten time interval between taking sample for culture
and identification of species, in-vitro susceptibilities and resistance determinants. Rapid diagnostics will
ce

provide relevant information not only for determination of appropriate therapy much earlier but also for
implementing the appropriate infection control measures. Therefore, affordable, user-friendly and accurate
Ac

point-of-care tests are urgently needed in routine practice.


2- In the literature, non-carbapenem antibiotics are usually compared with carbapenems as monotherapy for
the treatment of infections with ESBL-producing organisms. However, combination of antibiotics could be
investigated as carbapenem sparing alternatives in well-designed clinical studies. Beta-lactam/beta-lactamase
inhibitor combinations plus aminoglycosides or cefepime plus aminoglycosides can be considered as most
suitable options for comparison with carbapenems in clinical studies for severe infections with ESBL producers.
3- Newer BLBLIs (ceftazidime-avibactam and ceftolozane-tazobactam) should be reserved for infections with
carbapenem-resistant organisms and resistant P. aeruginosa infections, respectively, but their role in infections
with ESBL-producing pathogens should also be established.
4- Efficacy of BLBLIs and other alternatives should be compared with carbapenems in infections where an
effective source control can be provided. Examples are intra-abdominal infections that can be drained and
catheter-related infections in which catheter removal is performed.
5- Efficacy of cephamycins, fosfomycin, pivmecillinam, temocillin and aminoglycosides in complicated UTIs
should be investigated and also optimal dose and duration of therapies with these antibiotics according to
varied MIC values should be determined by future studies.
6- Local molecular epidemiologic data and resistance patterns should be screened periodically to determine
optimal therapies in a particular region.
7- Prediction score models or decision tree algorithms may be useful for early prediction of infections caused

t
ip
by ESBL-producing organisms and decreasing unnecessary carbapenem prescription. Validation studies of
previous prediction score models and decision tree algorithms or constructing new ones can be performed in

cr
future studies.
8- Oral treatment alternatives merit further studies including infections caused by ESBL-producing

us
Enterobacteriacea particularly as options for ambulatory therapy of community-acquired, mild infections.

an
Funding

This work had no funding


M

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a
ed

financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This
includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or
patents received or pending, or royalties.
pt

Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.
ce
Ac
References

Papers of special note have been highlighted as:

* of interest

** of considerable interest

1- Livermore DM. Current epidemiology and growing resistance of gram-negative pathogens. Korean J Intern
Med 2012; 27:128–42.

t
ip
2- Novais A, Canton R, Moreira R, et al. Emergence and dissemination of Enterobacteriaceae isolates producing
CTX-M-1-like enzymes in Spain are associated with IncFII (CTX-M-15) and broad host range (CTX-M-1, -3, and -
32) plasmids. Antimicrob Agents Chemother 2004; 48:3758-64.

cr
3- Livermore DM, Canton R, Gniadkowski M, et al. CTX-M: changing the face of ESBLs in Europe. J Antimicrob
Chemother 2007; 59:165-74.

us
4- Harris PNA, Tambyah PA, Paterson DL. β-lactam and β-lactamase inhibitor combinations in the treatment of
extended-spectrum β-lactamase producing Enterobacteriaceae: time for a reappraisal in the era of few
antibiotic options? Lancet Infect Dis 2015; 15:475-85.
an
5- Merino I, Shaw E, Horcajada JP, et al. CTX-M-15-H30Rx-ST131 subclone is one of the main causes of
healthcare-associated ESBL-producing Escherichia coli bacteraemia of urinary origin in Spain. J Antimicrob
Chemother 2016; 71:2125-30.
M

6- European Centre for Disease Prevention and Control (ECDC). Surveillance of antimicrobial resistance in
Europe 2017. Annual report of the European Antimicrobial Resistance Surveillance Network (EARS-Net).
Stockholm, Sweden: ECDC; 2018.
ed

7- Castanheira M, Mills JC, Costello SE, et al. Ceftazidime-avibactam activity tested against Enterobacteriaceae
isolates from US hospitals (2011 to 2013) and characterization of β-lactamase-producing strains. Antimicrob
Agents Chemother 2015; 59:3509–17.
pt

8- McDanel J, Schweizer M, Crabb V, et al. Incidence of extended-spectrum β-Lactamase (ESBL)-producing


Escherichia coli and Klebsiella infections in the United States: A systematic literature review. Infect Control
ce

Hosp Epidemiol 2017; 38:1209-15.

9- Jean SS, Hsueh PR, SMART Asia-Pacific Group. Distribution of ESBLs, AmpC β-lactamases and
carbapenemases among Enterobacteriaceae isolates causing intra-abdominal and urinary tract infections in the
Ac

Asia-Pacific region during 2008-14: results from the study for monitoring antimicrobial resistance trends
(SMART). J Antimicrob Chemother 2017; 72:166–71.

10- Flokas ME, Alevizakos M, Shehadeh F, et al. Extended- spectrum β-lactamase-producing Enterobacteriaceae
colonisation in long-term care facilities: a systematic review and meta-analysis. Int J Antimicrob Agents 2017;
50:649–56.

11- Karanika S, Karantanos T, Arvanitis M, et al. Fecal colonization with extended-spectrum beta-lactamase-
producing Enterobacteriaceae and risk factors among healthy individuals: a systematic review and
metaanalysis. Clin Infect Dis 2016; 63:310-18.

12- Bevan ER, Jones AM, Hawkey PM. Global epidemiology of CTX-M β-lactamases: temporal and geographical
shifts in genotype. J Antimicrob Chemother 2017; 72:2145-55.
13- Rodríguez-Baño J and Navarro MD. Extended-spectrum β-lactamases in ambulatory care: a clinical
perspective. Clin Microbiol Infect 2008; 14:104–10.

**14- Tacconelli E, Carrara E, Savoldi A, et al. WHO Pathogens Priority List Working Group. Discovery, research,
and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis.
Lancet Infect Dis 2018; 18:318–27.

This report describes ESBL-producing Enterobacteriaceae as one of the most critical pathogen for public health
and calling urgent research for development of new antibiotics

15- Stuart RL, Kotsanas D, Webb B, et al. Prevalence of antimicrobial-resistant organisms in residential aged
care facilities. Med J Aust 2011; 195:530–33.

t
ip
16- Rooney PJ, O’Leary MC, Loughrey AC, et al. Nursing homes as a reservoir of extended-spectrum β-
lactamase (ESBL)-producing ciprofloxacin-resistant Escherichia coli. J Antimicrob Chemother 2009; 64:635-41.

cr
17- Rodríguez-Baño J, Paterson DL. A change in the epidemiology of infections due to extended-spectrum β-
lactamase-producing organisms. Clin Infect Dis 2006; 42:935–37.

us
18- Quan J, Zhao D, Liu L, et al. High prevalence of ESBL-producing Escherichia coli and Klebsiella pneumoniae in
community-onset bloodstream infections in China. J Antimicrob Chemother 2017; 72:273–80.

19- Singer RS, Finch R, Wegener HC, et al. Antibiotic resistance-the interplay between antibiotic use in animals
and human beings. Lancet Infect Dis 2003; 3:47–51.
an
20- Paterson DL, Bonomo RA. Extended- spectrum beta-lactamases: a clinical update. Clin Microbiol Rev 2005;
18:657–86.
M
21- Doi Y, Lovleva A, Bonomo RA. The ecology of extended-spectrum β-lactamases (ESBLs) in the developed
World. J Travel Med 2017; 24:44-51.
ed

22- Bréchet C, Plantin J, Sauget M, et al. Wastewater treatment plants release large amounts of extended-
spectrum β-lactamase-producing Escherichia coli into the environment. Clin Infect Dis 2014; 58:1658-65.

23- Hawkey PM. Multidrug-resistant gram-negative bacteria: a product of globalization. J Hosp Infect 2015; 89:
241-47.
pt

24- Kim YK, Pai H, Lee HJ, et al. Bloodstream infections by extended-spectrum beta-lactamase-producing
Escherichia coli and Klebsiella pneumoniae in children: epidemiology and clinical outcome. Antimicrob Agents
ce

Chemother 2002; 46:1481-91.

25- Ha YE, Kang CI, Cha MK, et al. Epidemiology and clinical outcomes of bloodstream infections caused by
extended-spectrum β-lactamase-producing Escherichia coli in patients with cancer. Int J Antimicrob Agents
Ac

2013; 42:403-09.

26- Kang CI, Chung DR, Ko KS, et al. Korean Network for Study of Infectious Diseases. Risk factors for infection
and treatment outcome of extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella
pneumoniae bacteremia in patients with hematologic malignancy. Ann Hematol 2012; 91: 115-21.

27- Qureshi ZA, Paterson DL, Peleg AY, et al. Clinical characteristics of bacteraemia caused by extended-
spectrum β-lactamase-producing Enterobacteriaceae in the era of CTX-M-type and KPC-type β-lactamases. Clin
Microbiol Infect 2012; 18:887-93.

28- Lautenbach E, Patel JB, Bilker WB, et al. Extended-spectrum beta-lactamase-producing Escherichia coli and
Klebsiella pneumoniae: risk factors for infection and impact of resistance on outcomes. Clin Infect Dis 2001;
32:1162–71.
29- Stewardson A, Fankhauser C, De Angelis G, et al. Burden of bloodstream infection caused by extended-
spectrum β-lactamase-producing enterobacteriaceae determined using multistate modelling at a Swiss
University Hospital and a nationwide predictive model. Infect Control Hosp Epidemiol 2013; 34:133-43.

30- Nieminen O, Korppi M, Helminen M. Healthcare costs doubled when children had urinary tract infections
caused by extended-spectrum β-lactamase-producing bacteria. Acta Paediatr 2017; 106:327-33.

31- Tumbarello M, Spanu T, Di Bidino R, et al. Costs of bloodstream infections caused by Escherichia coli and
influence of extended-spectrum-beta-lactamase production and inadequate initial antibiotic therapy.
Antimicrob Agents Chemother 2010; 54:4085–91.

32- MacVane SH, Tuttle LO, Nicolau DP. Impact of extended-spectrum β-lactamase-producing organisms on

t
clinical and economic outcomes in patients with urinary tract infection. J Hosp Med 2014; 9:232-38.

ip
33- Schwaber MJ, Venezia SN, Kaye KS, et al. Clinical and Economic Impact of Bacteremia with Extended-

cr
Spectrum-β-Lactamase-Producing Enterobacteriaceae. Antimicrob Agents Chemother 2006; 50:1257-62

34- De Rosa FG, Pagani N, Fossati L, et al. The effect of inappropriate therapy on bacteremia by ESBL-producing
bacteria Infection 2011; 39:555–61.

us
35- Tsai HY, Chen YH, Tang HJ, et al. Carbapenems and piperacillin/tazobactam for the treatment of bacteremia
caused by extended-spectrum β-lactamase-producing Proteus mirabilis. Diagn Microb and Infect Dis 2014; 80:
222-26.
an
36- Tacconelli E, Cataldo MA, Mutters NT, et al. Role of place of acquisition and inappropriate empirical
antibiotic therapy on the outcome of extended-spectrum β-lactamase-producing Enterobacteriaceae
M
infections. Int J Antimicrob Agents 2019; 54(1):49-54.

37- Tumbarello M, Sanguinetti M, Montuori E, et al. Predictors of mortality in patients with bloodstream
infections caused by extended-spectrum-beta-lactamase-producing Enterobacteriaceae: importance of
inadequate initial antimicrobial treatment. Antimicrob Agents Chemother 2007; 51:1987–94.
ed

38- Endimiani A, Luzzaro F, Brigante G, et al. Proteus mirabilis bloodstream infections: risk factors and
treatment outcome related to the expression of extended-spectrum beta-lactamases. Antimicrob Agents
Chemother 2005; 49:2598–05.
pt

39- Lee JC, Lee NY, Lee HC, et al. Clinical characteristics of urosepsis caused by extended-spectrum beta-
lactamase-producing Escherichia coli or Klebsiella pneumoniae and their emergence in the community. J
ce

Microbiol Immunol Infect 2012; 45:127–33.

40- Augustine MR, Testerman TL, Justo JA, et al. Clinical risk score for prediction of extended-spectrum β-
Ac

lactamase-producing Enterobacteriaceae in bloodstream isolates. Infect Control Hosp Epidemiol 2017; 38:266-
72

41- Goodman KE, Lessler J, Cosgrove SE, et al. A clinical decision tree to predict whether a bacteremic patient is
infected with an extended-spectrum β-lactamase-producing organism. Clin Infect Dis 2016; 63:896-03.

42- Thaden TJ, Fowler WG, Sexton DJ, et al. Increasing incidence of extended-spectrum β-lactamase-producing
Escherichia coli in community hospitals throughout the southeastern United States. Infect Control Hosp
Epidemiol 2016; 37:49-54.

43- Palacios-Baena ZR, Delgado-Valverde M, Valiente Méndez A, et al. Impact of de-escalation on prognosis of
patients with bacteraemia due to Enterobacteriaceae: a post-hoc analysis from a multicenter prospective
cohort. Clin Infect Dis 2019; 69(6):956-62
44- Tamma PD, Conley AT, Cosgrove SE, et al. Association of 30-Day Mortality With Oral Step-Down
vs Continued Intravenous Therapy in Patients Hospitalized With Enterobacteriaceae Bacteremia. JAMA Intern
Med 2019; 179:316-23.

45- Matsumura Y, Yamamoto M, Nagao M, et al. In vitro activities and detection performances of cefmetazole
and flomoxef for extended-spectrum β-lactamase and plasmid-mediated AmpC β-lactamase-producing
Enterobacteriaceae. Diagn Microbiol Infect Dis 2016; 84:322–27.

46- Matsumura Y, Yamamoto M, Nagao M, et al. Multicenter retrospective study of cefmetazole and flomoxef
for treatment of extended-spectrum-β-lactamase-producing Escherichia coli bacteremia. Antimicrob Agents
Chemother 2015; 59:107-13.

t
47- Fukuchi T, Iwata K, Kobayashi S, et al. Cefmetazole for bacteremia caused by ESBL-producing

ip
enterobacteriaceae comparing with carbapenems. BMC Infect Dis 2016; 16(1):427-32.

48- Lee CH, Su LH, Chen FJ, et al. Clinical and microbiological characteristics of adult patients with recurrent

cr
bacteraemia caused by extended-spectrum β-lactamase-producing Escherichia coli or Klebsiella pneumoniae.
Clin Microbiol Infect 2015; 21(12): 1105.e1-1105.e8.

us
49- Lee CH, Su LH, Chen FJ, et al. Comparative effectiveness of flomoxef versus carbapenems in the treatment
of bacteraemia due to extended-spectrum β-lactamase-producing Escherichia coli or Klebsiella pneumoniae
with emphasis on minimum inhibitory concentration of flomoxef: a retrospective study. Int J Antimicrob Agents
2015; 46:610-15.
an
50- Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev. 2009; 22(1):161-82.

51- Alvarez M, Tran JH, Chow N, et al. Epidemiology of conjugative plasmid-mediated AmpC β-lactamases in
M
the United States. Antimicrob Agents Chemother 2004; 48: 533–37.

52-Perez F, Bonomo RA. Bloodstream infection caused by extended-spectrum β-lactamase–producing gram-


negative bacteria: How to define the best treatment regimen? Clin Infect Dis 2015; 60:1326-29.
ed

53- Bedenic B, Beader N, Zagar Z. Effect of inoculum size on the antibacterial activity of cefpirome and
cefepime against Klebsiella pneumoniae strains producing SHV extended-spectrum beta-lactamases. Clin
Microbiol Infect 2001; 7:626-35.
pt

54- Jett BD, Ritchie DJ, Reichley R, et al. In vitro activities of various beta-lactam antimicrobial agents against
clinical isolates of Escherichia coli and Klebsiella spp. resistant to oxyimino cephalosporins. Antimicrob Agents
ce

Chemother 1995; 39:1187-90.

55- Thomson KS, and Moland ES. Cefepime, piperacillin-tazobactam and the inoculum effect in tests with
extended-spectrum beta-lactamase-producing Enterobacteriaceae. Antimicrob. Agents Chemother 2001;
Ac

45:3548-54.

56- Burgess DS, Hall RG. In vitro killing of parenteral beta-lactams against standard and high inocula of
extended-spectrum beta-lactamase and non-ESBL producing Klebsiella pneumoniae. Diagn Microbiol Infect Dis
2004; 49:41-46.

57- Craig WA. Interrelationship between pharmacokinetics and pharmacodynamics in determining dosage
regimens for broad-spectrum cephalosporins. Diagn Microbiol Infect Dis 1995; 22:89-96.

58- Jacobs MR. Optimisation of antimicrobial therapy using pharmacokinetic and pharmacodynamic
parameters. Clin Microbiol Infect 2001; 7:589-96.
59- Roos FJ, Bulitta J, Lipman J, et al. Pharmacokinetic-pharmacodynamic rationale for cefepime dosing
regimens in intensive care units. J Antimicrob Chemother 2006; 58:987-93.

60- Lodise TP, Lomaestro BM, and Drusano GL, et al. Application of Antimicrobial Pharmacodynamic Concepts
into Clinical Practice: Focus on β-Lactam Antibiotics. Pharmacotherapy 2006; 26:1320-32.

61- Bhat SV, Peleg AY, Lodise TP, et al. Failure of current cefepime breakpoints to predict clinical outcomes of
bacteremia caused by gram negative organisms. Antimicrob Agents Chemother 2007; 51:4390-95.

62- Lee NY, Lee CC, Huang WH, et al. Cefepime therapy for monomicrobial bacteremia caused by cefepime-
susceptible extended-spectrum beta-lactamase-producing Enterobacteriaceae: MIC matters. Clin Infect Dis
2013; 56:488-95.

t
ip
63- Goethaert K, Looveren MV, Lammens C, et al. High-dose cefepime as an alternative treatment for infections
caused by TEM-24 ESBL-producing Enterobacter aerogenes in severely-ill patients. Clin Microbiol Infect 2006;
12:56-62.

cr
64- Rhodes NJ, Richardson CL, Heraty R, et al. Unacceptably high error rates in Vitek 2 testing of cefepime
susceptibility in extended spectrum β-lactamase producing Escherichia coli. Antimicrob Agents Chemother

us
2014; 58:3757–61.

65- Benanti GE, Brown AR, Shigle TL, et al. Carbapenem versus cefepime or piperacillin-tazobactam for empiric
treatment of bacteremia due to extended-spectrum-β-lactamase-producing Escherichia coli in patients with
an
hematologic malignancy. Antimicrob Agents Chemother 2019; 63:813-18.

66- Kim SA, Altshuler J, Paris D, et al. Cefepime versus carbapenems for the treatment of urinary tract
infections caused by extended-spectrum β-lactamase-producing Enterobacteriaceae. Int J Antimicrob Agents
M
2018; 51:155-158.

67- Wang R, Cosgrove SE, Sutter ST, et al. Cefepime Therapy for Cefepime- Susceptible Extended-Spectrum β-
Lactamase-Producing Enterobacteriaceae Bacteremia. Open Forum Infect Dis 2016; (4). DOI:10.1093.
ed

68- Chopra T, Marchaim D, Veltman J, et al. Impact of cefepime therapy on mortality among patients with
bloodstream infections caused by extended-spectrum-beta-lactamase-producing Klebsiella pneumoniae and
Escherichia coli. Antimicrobial Agents Chemother 2012; 56: 3936-42.
pt

69- Pagani L, Migliavacca R, Luzzaro F, et al. Comparative activity of piperacillin/tazobactam against clinical
isolates of extended-spectrum beta-lactamase-producing Enterobacteriaceae. Chemotherapy 1998; 44:377–84.
ce

70- Canoui E, Tankovic J, Bige N, et al. Which proportion of extended-spectrum beta-lactamase producing
strains could be treated by non-carbapenem beta-lactams? Med Mal Infect 2014; 44:235-37.
Ac

71- Jean SS, Coombs G, Ling T, et al. Epidemiology and antimicrobial susceptibility profiles of pathogens causing
urinary tract infections in the Asia-Pacific region: Results from the Study for Monitoring Antimicrobial
Resistance Trends (SMART), 2010–2013. Int J Antimicrob Agents 2016; 47:328–34

72- Bonfiglio G, Livermore DM. Inoculum effects on E-tests and agar dilution minimum inhibitory
concentrations. Piperacillin and piperacillin-tazobactam against Staphylococcus aureus. Diagn Microbiol Infect
Dis 1994; 19:163-66.

73- López-Cerero L, Picón E, Morillo C, et al. Comparative assessment of inoculum effects on the antimicrobial
activity of amoxycillin-clavulanate and piperacillin-tazobactam with extended-spectrum β-lactamase-producing
and extended-spectrum β-lactamase-non-producing Escherichia coli isolates. Clin Microbiol Infect 2010;
16:132–36.
74- MacGowan A. Breakpoints for extended-spectrum beta-lactamase–producing Enterobacteriacae:
pharmacokinetic/pharmacodynamic considerations. Clin Microbiol Infect 2008; 14:166-68.

75- Carlier M, Carrette S, Roberts JA, et al. Meropenem and piperacillin/tazobactam prescribing in critically ill
patients: does augmented renal clearance affect pharmacokinetic/pharmacodynamic target attainment when
extended infusions are used? Crit Care 2013; 17(3): R84.

76- Zelenitsky SA, Ariano RE, Zhanel GG. Pharmacodynamics of empirical antibiotic monotherapies for an
intensive care unit (ICU) population based on Canadian surveillance data. J Antimicrob Chemother 2011; 66:
343-49.

77- Yost RJ, Cappelletty DM. RECEIPT Study group. The Retrospective Cohort of Extended-Infusion Piperacillin-

t
Tazobactam (RECEIPT) study: a multicenter study. Pharmacotherapy 2011; 31:767–75.

ip
78- Bartoletti M, Giannella M, Lewis RE, et al. Extended infusion of β-lactams for bloodstream infection in
patients with liver cirrhosis: An observational multicenter study. Clin Infect Dis 2019; 69(10):1731-39.

cr
79- Dulhunty JM, Roberts JA, Davis JS, et al. A multicenter randomized trial of continuous versus intermittent β-
lactam infusion in severe sepsis. Am J Respir Crit Care Med 2015; 192:1298-05.

us
80- Babini GS, Yuan M, Hall LM, et al. Variable susceptibility to piperacillin/tazobactam amongst Klebsiella spp.
with extended-spectrum β-lactamases. J Antimicrob Chemother 2003; 51: 605-12.
an
81- Castanheira M, Farrell SE, Krause KM, et al. Contemporary diversity of β-lactamases among
Enterobacteriaceae in the nine US census regions and ceftazidime-avibactam activity tested against isolates
producing the most prevalent β-lactamase groups. Antimicrob Agents Chemother 2014; 58:833-38.
M
82- Rodríguez-Baño J, Navarro MD, Retamar P, et al. β-lactam/β-lactamase inhibitor combinations for the
treatment of bacteremia due to extended-spectrum β -lactamase–producing Escherichia coli: A post hoc
analysis of prospective cohorts. Clin Infect Dis 2012; 54:167-74.
ed

83- Oteo J, Cercenado E, Cuevas O, et al. AmpC beta-lactamases in Escherichia coli: emergence of CMY-2-
producing virulent phylogroup D isolates belonging mainly to STs 57, 115, 354, 393, and 420, and phylogroup
B2 isolates belonging to the international clone O25b- ST131. Diagn Microbiol Infect Dis 2010; 67:270-76.
pt

84- Retamar P, López-Cerero L, Muniain MA, et al. Impact of the MIC of Piperacillin-tazobactam on the
outcome of patients with bacteremia due to extended-spectrum- β-lactamase-producing Escherichia coli.
Antimicrob Agents Chemother 2013; 57:3402-04.
ce

85- Tamma PD, Han JH, Rock C, et al. Carbapenem therapy is associated with improved survival compared with
piperacillin-tazobactam for patients with extended- spectrum β-lactamase bacteremia. Clin Infect Dis 2015;
60:1319-25.
Ac

86- Miller B, Hershberger E, Benziger D. et al. Pharmacokinetics and safety of intravenous ceftolozane-
tazobactam in healthy adult subjects following single and multiple ascending doses. Antimicrob Agents
Chemother 2012; 56:3086-91.

87- Drawz SM, Bonomo RA. Three decades of beta-lactamase inhibitors. Clin Microbiol Rev 2010; 23:160–201.

88- Perez F, Bonomo RA. Can we really use β-lactam/ β-lactam inhibitor combinations for the treatment of
infections caused by extended-spectrum β-lactamase–producing bacteria? Clin Infect Dis 2012; 54:175-77.

89- Gutiérrez-Gutiérrez B, Pérez-Galera S, Salamanca E, et al. A multinational, preregistered cohort study of β-


lactam/ β-lactamase inhibitor combinations for treatment of bloodstream infections due to extended-
spectrum- β-lactamase-producing Enterobacteriaceae. Antimicrob Agents Chemother 2016; 60:4159-69.
90- Scheuerman O, Schechner V, Carmeli Y, et al. Comparison of predictors and mortality between bloodstream
infections caused by ESBL-producing Escherichia coli and ESBL-producing Klebsiella pneumoniae. Infect Control
Hosp Epidemiol 2018; 39:660-67.

91- Giannella M, Pascale R, Ferraro G, et al. Risk factors for treatment failure in patients receiving β-lactam/β-
lactamase inhibitor combinations for Enterobacteriaceae bloodstream infection: A retrospective, single-centre,
cohort study. Int J Antimicrob Agents 2019; 53:574-81.

92- Gudiol C, Royo-Cebrecos C, Abdala E, et al. Efficacy of β lactam/ β-lactamase inhibitor combinations for the
treatment of bloodstream infection due to extended-spectrum- β-lactamase- producing Enterobacteriaceae in
hematological patients with neutropenia. Antimicrob Agents Chemother 2017; 61(8): e00164-17.

t
93- Ng TM, Khong WX, Harris PN, et al. Empiric piperacillin-tazobactam versus carbapenems in the treatment of

ip
bacteraemia due to extended- spectrum β-lactamase-producing Enterobacteriaceae. PLoS One 2016; 11:
e0153696.

cr
94- Vardakas KZ, Tansarli GS, Rafailidis PI, et al. Carbapenems versus alternative antibiotics for the treatment of
bacteraemia due to Enterobacteriaceae producing extended-spectrum β-lactamases:
a systematic review and meta-analysis. J Antimicrob Chemother 2012; 67:2793–03.

us
95- Mohammed M, Flokas ME, Detsis M, et al. Comparison between carbapenems and β-lactam/β-lactamase
inhibitors in the treatment for bloodstream infections caused by extended-spectrum β-lactamase-producing
Enterobacteriaceae: A systematic review and meta-analysis. Open Forum Infect Dis 2017; (8). DOI:10.1093.
an
*96- Sfeir MM, Askin G, Christos P, et al. Beta-lactam/beta-lactamase inhibitors versus carbapenem for
bloodstream infections due to extended-spectrum beta-lactamase-producing Enterobacteriaceae: Systematic
review and meta-analysis. Int J Antimicrob Agents 2018; 52:554-70.
M

In this systematic review and meta-analysis, totally 3842 participants in 25 observational studies were
analyzed. The 30-day mortality rates were similar for BLBLI or PTZ and carbapenem for the treatment of ESBL-
producing Enterobacteriaceae infections.
ed

97- Lee CH, Su LH, Li CC, et al. Microbiologic and clinical implications of bacteremia due to extended-spectrum-
β-lactamase-producing Klebsiella pneumoniae with or without plasmid-mediated AmpC β-lactamase DHA-1.
Antimicrob Agents Chemother 2010; 54: 5395-98.
pt

98- McLaughlin M, Advincula MR, Malczynski M, et al. Correlations of antibiotic use and carbapenem resistance
in Enterobacteriaceae. Antimicrob Agents Chemother 2013; 57:5131-33.
ce

99- Livermore DM. Fourteen years in resistance. Int J Antimicrob Agents 2012; 39:283–94.

**100- Harris PNA, Tambyah PA, Lye DC, et al. Effect of piperacillin-tazobactam vs meropenem on 30-day
Ac

mortality for patients with E coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance: a
randomized clinical trial. JAMA 2018; 320:984–94.

This is the only randomized controlled trail comparing the efficacy of PTZ with meropenem as definitive
therapy in patients with BSI due to cephalosporin- resistant Enterobacteriaceae showing the inferiority of PTZ
as compared with meropenem in terms of 14-day mortality rate. Several limitations of the study is described in
the text.

101- Sfeir MM. Post-MERINO trial: Any role for piperacillin-tazobactam in treating bloodstream infections
caused by extended-spectrum beta-lactamase producing Enterobacteriaceae? Int J Antimicrob Agents 2019;
53:557–58.
102- Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Kahlmeter G. Antibiotics for ceftriaxone-resistant gram-negative
bacterial bloodstream infections. JAMA 2019; 321:612-13.

103- Prevel R, Berdaï D, Boyer A. Antibiotics for ceftriaxone-resistant gram-negative bacterial bloodstream
infections. JAMA 2019; 321:613.

104- Su J, Guo Q, Li Y, et al. Comparison of empirical therapy with cefoperazone/sulbactam or a carbapenem


for bloodstream infections due to ESBL-producing Enterobacteriaceae. J Antimicrob Chemother 2018; 73:3176-
80.

105- Livermore DM, Tulkens PM. Temocillin revived. J Antimicrob Chemother 2009; 63:243-45.

t
106- Balakrishnan I, Awad-El-Kariem FM, Aali A, et al. Temocillin use in England: clinical and microbiological

ip
efficacies in infections caused by extended-spectrum and/or derepressed AmpC β-lactamase-producing
Enterobacteriaceae. J Antimicrob Chemother 2011; 66:2628-31.

cr
107- Dijkmans AC, Ortiz Zacarias NV, Burggraaf J, et al. Fosfomycin: Pharmacological, clinical and future
perspectives. Antibiotics 2017;6,24.

us
108- Roussos N, Karageorgopoulos DE, Samonis G, et al. Clinical significance of the pharmacokinetic and
pharmacodynamic characteristics of fosfomycin for the treatment of patients with systemic infections. Int J
Antimicrob Agents. 2009; 34:506-15.
an
109- Wenz Bi, Bin Li, Jianging Song et al. Antimicrobial susceptibility and mechanisms of fosfomycin resistance
in extended-spectrum β-lactamase-producing Escherichia coli strains from urinary tract infections in Wenzhou,
China. Int J Antimicrob Agents 2017; 50:29-34.
M
110- Lu PL, Hsieh YJ, Lin JE, et al. Characterisation of fosfomycin resistance mechanisms and molecular
epidemiology in extended-spectrum β-lactamase-producing Klebsiella pneumoniae isolates. Int J Antimicrob
Agents 2016; 48(5):564-68.
ed

111- Falagas ME, Kastoris AC, Kapaskelis AM, et al. Fosfomycin for the treatment of multidrug-resistant,
including extended-spectrum β-lactamase-producing, Enterobacteriaceae infections: a systematic review.
Lancet Infect Dis 2010; 10:43-50.
pt

112- Aris P, Boroumand MA, Rahbar M, et al. The activity of fosfomycin against extended-spectrum-beta-
lactamase-producing isolates of Enterobacteriaceae recovered from urinary tract infections: a single-center
ce

study over a period of 12 years. Microbial Drug Resistance 2018; 24:607-12.

113- Kaye K, Rice L, Dane A, et al. Fosfomycin for injection (ZTI-01) vs piperacillin-tazobactam (PIP-TAZ) for the
treatment of complicated urinary tract infection (cUTI) including acute pyelonephritis (AP): ZEUS, a phase 2/3
Ac

randomized trial. Clin Infect Dis 2019. pii: ciz181.

114 Senol S, Tasbakan M, Pullukcu H, et al. Carbapenem versus fosfomycin trometamol in the treatment of
extended-spectrum β-lactamase-producing Escherichia coli-related complicated lower urinary tract infection. J
Chemother 2010; 22:355-57.

115- Los-Arcos I, Pigrau C, Rodríguez-Pardo D, et al. Long-Term fosfomycin-tromethamine oral therapy for
difficult-to-treat chronic bacterial prostatitis. Antimicrob Agents Chemother 2016; 60:1854-52.

116- Veve MP, Wagner JL, Kenney RM, et al. Comparison of fosfomycin to ertapenem for outpatient or step-
down therapy of extended-spectrum β-lactamase urinary tract infections. Int J Antimicrob Agents 2016; 48:46-
60.
*117- Rosso-Fernández C, Sojo-Dorado J, Barriga A, et al. Fosfomycin versus meropenem in bacteraemic
urinary tract infections caused by extended- spectrum β-lactamase-producing Escherichia coli (FOREST): Study
protocol for an investigator-driven randomised controlled trial. BMJ Open. 2015; 5(3): e007363.

This eagerly awaited RCT will demonstrate utility of fosfomycin in bacteraemic UTIs with ESBL-producing E. coli.
The results of this study may change our routine practice substantially in near future.

118- Ben Zakour NL, Alsheikh-Hussain AS, Ashcroft MM, et al. Sequential acquisition of virulence and
fluoroquinolone resistance has shaped the evolution of Escherichia coli ST131. mBio 2016; 7(2): e00347-16.

119- Merino I, Shaw E, Horcajada JP, et al. CTX-M-15-H30Rx-ST131 subclone is one of the main causes of
healthcare-associated ESBL-producing Escherichia coli bacteraemia of urinary origin in Spain. J Antimicrob

t
Chemother 2016; 71: 2125–30.

ip
120- Mathers AJ, Peirano G, Pitout JDD. The role of epidemic resistance plasmids and international high- risk
clones in the spread of multidrug-resistant Enterobacteriaceae. Clin Microbiol Rev 2015; 28:565-91

cr
121- Zhang H, Johnson A, Zhang G, et al. Susceptibilities of Gram-negative bacilli from hospital- and
community-acquired intra-abdominal and urinary tract infections: a 2016–2017 update of the Chinese SMART

us
study. Infection and Drug Resistance 2019; 12:905-14.

122- Rodríguez-Baño J, Navarro MD, Romero L. Bacteremia due to extended-spectrum β-lactamase-producing


Escherichia coli in the CTX-M era: A new clinical challenge. Clin Infect Dis 2006; 43:1407-14.
an
123- Paterson DL, Ko WC, Gottberg AV, et al. Antibiotic therapy for Klebsiella pneumoniae bacteremia:
implications of production of extended-spectrum-β-lactamases. Clin Infect Dis 2003; 39:31-37.
M
124- Tumbarello M, Sanguinetti M, Montuori E, et al. Predictors of mortality in patients with bloodstream
infections caused by extended-spectrum-beta-lactamase-producing Enterobacteriaceae: importance of
inadequate initial antimicrobial treatment. Antimicrob Agents Chemother 2007; 51:1987-94.
ed

125- Bassetti M, Peghin M, Pecori D. The management of multidrug-resistant Enterobacteriaceae. Curr Opin
Infect Dis. 2016; 29:583-94.

126- Cha MK, Kang C, Kim SH, et al. In vitro activities of 21 antimicrobial agents alone and in combination with
pt

aminoglycosides or fluoroquinolones against extended-spectrum-β-lactamase-producing Escherichia coli


isolates causing bacteremia. Antimicrob Agents Chemother 2015; 59:5834-37.
ce

127- Cho SY, Choi SM, Park SH, et al. Amikacin therapy for urinary tract infections caused by extended-
spectrum- β-lactamase-producing Escherichia coli. Korean J Intern Med 2016; 31:156-61.

128- Ipekci T, Seyman D, Berk H, et al. Clinical and bacteriological efficacy of amikacin in the treatment of lower
Ac

urinary tract infection caused by extended-spectrum beta-lactamase-producing Escherichia coli or Klebsiella


pneumoniae. J Infect Chemother 2014; 20:762-67.

129- Polat M, Kara SS. Once-daily intramuscular amikacin for outpatient treatment of lower urinary tract
infections caused by extended-spectrum β-lactamase-producing Escherichia coli in children. Infect Drug Resist
2017; 10:393-99.

130- Ramirez, MS., and Tolmasky ME. Aminoglycoside modifying enzymes. Drug Resist Updat 2010; 13:151-71.

131- Pana ZD, Zaoutis T. Treatment of extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBLs)


infections: what have we learned until now? F1000Research 2018, 7(F1000 Faculty Rev):1347.
132- Zhanel GG, Lawson CD, Zelenitsky S, et al. Comparison of the next-generation aminoglycoside plazomicin
to gentamicin, tobramycin and amikacin. Expert Rev Anti Infect Ther 2012; 10:459-73.

133- Raja NS. Oral treatment options for patients with urinary tract infections caused by extended spectrum
Beta-lactamase (ESBL) producing Enterobacteriaceae. J Infect Public Health 2019, Jun 5. pii: S1876-
0341(19)30180-7. doi: 10.1016/j.jiph.2019.05.012. [Epub ahead of print].

134- Auer S, Wojna A, Hell M. Oral treatment options for ambulatory patients with urinary tract infections
caused by extended-spectrum-beta-lactamase-producing Escherichia coli. Antimicrob Agents Chemother 2010;
54:4006-08.

135- Bollestad M, Grude N, Solhaug S, et al. Clinical and bacteriological efficacy of pivmecillinam treatment for

t
uncomplicated urinary tract infections caused by ESBL-producing Escherichia coli: a prospective, multicentre,

ip
observational cohort study. J Antimicrob Chemother 2018; 73:2503–09.

136- Titelman E, Iversen A, Kalin M, et al. Efficacy of pivmecillinam for treatment of lower urinary tract

cr
infection caused by extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella pneumoniae.
Microb Drug Resist 2012; 18:189-92.

us
137- Jansaker F, Frimodt-Moller N, Sjogren I, et al. Clinical and bacteriological effects of pivmecillinam for ESBL-
producing Escherichia coli or Klebsiella pneumoniae in urinary tract infections. J Antimicrob Chemother 2014;
69:769-72.
an
138- Sucher AJ, Chahine EB, Cogan P, et al. Ceftolozane/tazobactam: A new cephalosporin and β-lactamase
inhibitor combination. Ann Pharmacother. 2015; 49:1046–56.

130- Hawkey PM, Warren RE, Livermore DM, et al. Treatment of infections caused by multidrug-resistant
M
Gram-negative bacteria: report of the British Society for Antimicrobial Chemotherapy/Healthcare Infection
Society/British Infection Association Joint Working Party. J Antimicrob Chemother 2018; 73 Suppl 3: iii2–iii78.

140- Shortridge D, Pfaller MA, Castanheira M, et al. Antimicrobial activity of ceftolozane-tazobactam tested
ed

against Enterobacteriaceae and Pseudomonas aeruginosa collected from patients with bloodstream infections
isolated in United States hospitals (2013–2015) as part of the Program to Assess Ceftolozane-Tazobactam
Susceptibility (PACTS) surveillance program. Diagn Microbiol Infect Dis 2018; 92:158-63.
pt

*141- Popejoy MW, Paterson DL, Cloutier D, et al. Efficacy of ceftolozane/tazobactam against urinary tract and
intra-abdominal infections caused by ESBL-producing Escherichia coli and Klebsiella pneumoniae: a pooled
analysis of Phase 3 clinical trials. J Antimicrob Chemother 2017; 72:268-72.
ce

This pooled analysis of RCTs demonstrated the high clinical efficacy of ceftolozane-tazobactam in UTIs and IAIs
caused by ESBL-producing Enteroacteriaceae. This result should not be interpreted as ceftolozane-tazobactam
is a potential carbapenem sparing alternative for infections with ESBL-producing Enterobacteriaceae. This drug
Ac

should be strictly reserved for MDR/XDR-Pseudomonas aeruginosa infections.

142- Bush K. A resurgence of beta-lactamase inhibitor combinations effective against multidrug-resistant Gram-
negative pathogens. Int J Antimicrob Agents 2015; 46:483-93.

*143- Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Machuca I, et al. Treatment of infections caused by extended-


spectrum-beta-lactamase-, AmpC-, and carbapenemase-producing Enterobacteriaceae. Clin Microbiol Rev
2018; 31: pii e00079-17.

This extensive review contains comprehensive information for treatment of infections caused by
Enterobacteriaceae producing ESBL, AmpC and carbapenemase.
144- Hackel M, Kazmierczak KM, Hoban DJ, et al. Assessment of the in vitro activity of ceftazidime-avibactam
against multidrug-resistant Klebsiella spp. Collected in the INFORM Global Surveillance Study, 2012 to 2014.
Antimicrob Agents Chemother 2016; 60:4677-83.

145- Kim SH, Lim KR, Lee H et al. Clinical effectiveness of oral antimicrobial therapy for acute pyelonephritis
caused by extended-spectrum β-lactamase-producing Enterobacteriales. Eur J Clin Microbiol Infect Dis 2019;
(9) https://doi.org/10.1007/s10096-019-03705-w

146- Lo CL, Lee CC, Li CW, Li MC, Hsueh PR, Lee NY, et al. Fluoroquinolone therapy for bloodstream infections
caused by extended-spectrum beta-lactamase- producing Escherichia coli and Klebsiella pneumoniae. J
Microbiol Immunol Infect 2017; 50:355-61.

t
147- FDA. FDA updates warnings for fluoroquinolone antibiotics on risks of mental health and low blood sugar

ip
adverse reactions Title [Internet]. Available at:
https://www.fda.gov/newsevents/newsroom/pressannouncements/ ucm612995.htm.

cr
148- GuptaK, HootonTM, NaberKG, et al. Infectious Diseases Society of America; European Society for
Microbiology and Infectious Diseases. International clinical practice guidelines for the treatment of acute
uncomplicated cystitis and pyelonephritis in women: a 2010 update by the Infectious Diseases Society of

us
America and the European Society for Microbiology and Infectious Diseases. Clin Infect Dis 2011; 52:103-20.

149- Grayson ML, Cosgrove SE, Crowe SM, et al. editors. Kucers’ the use of antibiotics: a clinical review of
antibacterial, antifungal, antiparasitic and antiviral drugs. 7th ed. Boca Raton (FL): CRC Press; 2017.
an
M
ed
pt
ce
Ac

You might also like