Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

CE: Namrta; QCO/320601; Total nos of Pages: 11;

QCO 320601

REVIEW

CURRENT
OPINION Carbapenem-sparing strategy: carbapenemase,
treatment, and stewardship
Silvia Corcione a, Tommaso Lupia a, Alberto Enrico Maraolo b
Simone Mornese Pinna a, Ivan Gentile b, and Francesco G. De Rosa a

Purpose of review
describing the current role of carbapenems and carbapenem-sparing strategies in the setting of
antimicrobial stewardship programs.
Downloaded from https://journals.lww.com/co-infectiousdiseases by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3HsbbxaSipcNThgg4eS87naQ1M9h5WkSgQUjy6hCmfEk= on 10/23/2019

Recent findings
sparing carbapenems with other drugs appears to be an interesting perspective for a variety of reasons in
the current context of the multidrug-resistant (MDR) pandemic. Specific algorithms should also be precisely
investigated to define better how to spare carbapenems within empiric and targeted regimens, with
combination treatment or monotherapies, aiming at the best use of the new drugs and improving de-
escalation as soon as possible for most of the patients.
Summary
stewardship programs may be useful in reducing probable misuse and overuse of antibiotics, which has
probably contributed to the emergence of carbapenem-resistant bacteria worldwide. The proposal of
carbapenem-sparing strategies has then generated substantial scientific debate and, overall, the concept of
sparing these drugs is well advocated together with judicious use of novel drugs, appropriate measures of
infection control and prevention as well as in stewardship programs to curb the spread of MDR and XDR-
strains in healthcare facilities.
Keywords
Acinetobacter, antimicrobial stewardship, carbapenem, carbapenem sparing, carbapenemase, carbapenem-
resistant Enterobacteriaceae, Pseudomonas

INTRODUCTION infection control, prevention, and stewardship pro-


Antimicrobial resistance is one of the most serious grams [9,10].
public health menaces worldwide [1,2]. Among the The present review is aimed at describing the
biggest threats, multidrug-resistant (MDR) or exten- current role of carbapenems and carbapenem-
sively drug-negative (XDR) Gram-negative bacteria sparing strategies in the setting of infections and
such as carbapenem-resistant Enterobacteriaceae proper antimicrobial therapy management dealing
(CRE), extended-spectrum b-lactamase producing with difficult-to-treat Gram-negative infections.
(ESBL) Enterobacteriaceae, Pseudomonas aeruginosa
and Acinetobacter baumannii have been labeled as
MECHANISMS OF RESISTANCE
the most serious or urgent [3–5].
With regard to MDR and XDR-Gram-negative Several mechanisms of antibiotic resistance have
bacteria, the use of carbapenems, alone or in combi- been described for Gram-negative bacteria (Table 1):
nation with other agents, is one the most controver-
sial issue: on the one hand their efficacy is well
established in many scenarios (for instance, infec- a
Department of Medical Sciences, Infectious Diseases, University of
tions by ESBL-Enterobacteriaceae), on the other hand, Turin, Turin and bDepartment of Clinical Medicine and Surgery, Section
their misuse and overuse (both as empiric or targeted of Infectious Disease, University of Naples Federico II, Naples, Italy
therapy) has resulted in the emergence of CRE which Correspondence to Francesco G. De Rosa, MD, Department of Medical
represent a paramount therapeutic challenge [6–8]. Sciences, Infectious Diseases, University of Turin, Turin, Italy.
Furthermore, carbapenem-sparing strategies have E-mail: francescogiuseppe.derosa@unito.it
generated substantial debate and are advocated with Curr Opin Infect Dis 2019, 32:000–000
judicious use of novel antibiotics, proper measures of DOI:10.1097/QCO.0000000000000598

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Gram-negative infections

(1) Enzymatic inactivation:


KEY POINTS (a) Class A b-lactamases include penicillinases
 Antimicrobial Stewardship Programs were pivotal to and cephalosporinases, such as (i.e., TEM,
reduce excessive or misuse of antibiotics in the struggle SHV, CTX-M, PER, VEB, GES, and IBC) with
against MDR and XDR-bacteria. development of resistance to carboxypeni-
cillins, ureidopenicillins, and aztreonam
 Carbapenem-sparing strategy concept has the aim to
[11,12]. This class also comprises Klebsiella
profess to save of these drugs and judicious use of
novel drugs not forgetting appropriate measures of pneumoniae carbapenemases (KPC) enzymes
infection control and prevention. and GES-2 that can hydrolyze carbapenems
[12,13].
 Antibiotic resistance passes through several (b) Class B are defined metallo-b-lactamases
mechanisms: enzymatic inactivation, efflux pumps,
(MBL), including IMP, verone integron-
reducing permeability, and target modifications.
encoded metallo-beta-lactamase (VIM),
 The rediscovery of molecules set aside, the synergistic New Delhi metallo-beta-lactamases (NDM),
association between drugs and new weapons in the SPM, and GIM, born in Asian countries and
pipeline will be the present and the hopeful future of after disseminated among Enterobacteriaceae
difficult-to-treat pathogens such as ESBLs, CR-E, and
worldwide [13–15].
resistant Pseudomonas and Acinetobacter spp.
(c) Class C comprises intrinsic and inducible
 Improving de-escalation, as soon as possible, is another resistance mechanisms, like AmpC-type
focal point that does not worsen clinical outcome in the cephalosporinase, not inhibited by clavu-
selected population. lanic acid, tazobactam, and sulbactam.
The structural modifications of AmpC,
can confer reduced susceptibilities to new

Table 1. Enzymes involved in carbapenems resistance in Enterobacteriaceae, Pseudomonas spp., and Acinetobacter spp.
Susceptibility to carbapenems
Mechanism of Resistance CRE Pseudomonas spp. Acinetobacter spp.

Enzymatic inactivation Between ‘()’ the most Between ‘()’ the most Between ‘()’ the most
important of the class important of the class important of the class
Class ‘A’ enzymes

(KPC, GES) (GES) (KPC and GES)


Class ‘B’ enzymes

(IMP, VIM, and NDM) (IMP, VIM, NDM, SPM, (IMP, VIM, NDM,
and GIM) and SIM)
Class ‘C’ enzymes

(AmpC together with (AmpC together with other (AmpC together with other
other mechanism mechanism reported, mechanism reported,
reported, e.g. Porins) e.g. Porins) e.g. Porins)
Class ‘D’ enzymes

(OXA, e.g. OXA-48, 181) (OXA, e.g. OXA-198) (OXA, e.g. OXA-51, 48)
Efflux pumps

(RND) (RND, e.g. MexAB-OprM) (RND, e.g. AdeABC)


Porins

(Omp33-36 OmpA, OmpK) (OprD, e.g. OprD2) (Omp33-36)


Target modifications

(PBPs alterations) (PBPs alterations) (PBPs alterations)

CR-E, carbapenem resistant Enterobacteriaceae; KPC, Klebsiella pneumoniae carbapenemases; NDM, New Delhi metallo-beta-lactamases; OMP, outer membrane
porin; OXA, oxacillin; PBP, penicillin-binding protein; RND, resistance-nodulation-division; VIM, verone integron-encoded metallo-beta-lactamase.

2 www.co-infectiousdiseases.com Volume 32  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Carbapenems sparing strategy Corcione et al.

§ C/T +/- Fosfomycin or AG If Suscepble Strains Consider:


§ C/A +/- Fosfomycin or AG § PTZ +/- AG
§ PTZ +/- AG § PTZ +/- Fosfomycin
For less severe infecons § Ceazidime +/- AG
§ Cefepime +/- AG § C/T +/- AG
§ Tigecycline +/- AG § C/A +/- AG
§ Tigecycline +/- FQ
For UTI no complicated If High Risk of MDR/XDR Strains Consider:
§ Fosfomycin
§ AG ESBL PA §
§
C/T + Colisn (plus/or Fosfomycin)
C/A + Colisn (plus/or Fosfomycin)
§ Nitrofurantoin

CR-E AB
§ C/A + Fosfomycin
§ C/A + Tigecycline § Colisn+ Fosfomycin
§ C/A + AG
§ Colisn + Tigecycline
§ C/A + Aztreonam
§ Colisn + Fosfomycin (plus/or Tigecycline) § Colisn + Rifampin
§ Tigecycline + Fosfomicin (plus/or AG) § Colisn + Aztreonam

FIGURE 1. Carbapenem sparing strategies in the setting of suspected infections by extended-spectrum b-lactamase producing,
P. aeruginosa (PA), A. baumannii (AB), carbapenem-resistant Enterobacteriaceae (CR-E). AG, aminoglycoside; C/A,
ceftazdime/avibactam; C/T, ceftolozane/tazobactam; FQ, fluoroquinolones.

b-lactam-b-lactamase inhibitor (BLBLI), proteins alterations), quinolones (mutations


ceftolozane/tazobactam, and ceftazidime/ in DNA gyrase and topoisomerases IV), and
avibactam [13,16]. polymixins (LPS alterations) [21].
(d) Class D oxacillinases provide resistance to
all penicillins, 3 and 4-generation cephalo-
sporines (3GC) and aztreonam [13,17].
Other OXA enzymes, such as OXA-198,
CARBAPENEMS IN THE SETTING OF
are known mechanisms of carbapenem
EXTENDED-SPECTRUM b-LACTAMASE
resistance [13,17], other OXAs b-lacta-
PRODUCING INFECTIONS
mases, mainly OXA-48, express low activity Carbapenem utilization has been fueled because of
against the carbapenems, better hydrolyz- the spread of ESBL, inciting a dangerous vicious
ing imipenem than meropenem [13,17]. circle in empiric and targeted treatment because
(2) Efflux pump and reduced permeability: of their efficacy against ESBL, but the quest for safe
(a) Resistance-nodulation-division (RND) fam- and effective alternatives has been representing a
ily are membrane proteins, including the &
significant area of research [22 ,23]. BLBLI combi-
main efflux pumps responsible for increased nations (e.g., piperacillin-tazobactam (PTZ) and
resistance of P. aeruginosa to a wide range of amoxicillin-clavulanic acid), cephamycins, amino-
agents commonly used. They rarely occur glycosides, fosfomycin, tigecycline, may show
individually but are often flanked by other potential activity: nevertheless, many of these
resistance mechanisms [18]. Lost or modifi- options can be used just under specific circumstan-
cation of OprD2 may increase the MIC for &&
ces [24 ,25] (Fig. 1). Among long-standing drugs in
carbapenems, especially imipenem [19,20]. the antibiotic armamentarium, cefepime is probably
(3) Target modifications: a useful option for not severe scenarios, for instance,
(a) Mainly, four classes of antimicrobials are urinary tract infections, whereas conflicting results
involved in mechanisms of resistance because are reported in case of severe clinical picture with
of change of the molecule target for P. aeru- &&
high bacterial inoculum [24 ,26]. In this specific
ginosa such as aminoglycosides (methylation setting, if strains are susceptible, valuable options
of 16S), b-lactams (penicillin-binding are fosfomycin and nitrofurantoin [27].

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com 3

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Gram-negative infections

’Old’ BLBLIs (primarily upon PTZ) in observa- as a carbapenem-sparing strategy for complicated
tional studies definitely did not show their inferior- abdominal or urinary tract ESBL infections, with
ity when compared with carbapenems against ESBL caution against Klebsiella spp. and encouraged versus
& &
infections in terms of mortality [28,29,30 ,31], and P. aeruginosa [40 ]. Ceftolozane/tazobactam may hap-
recently the missing piece in the ESBL puzzle was pen also as empiric therapy when there is a high
&
studied by a randomized controlled trial (RCT), the likelihood of serious infections by ESBL or CRE [40 ].
&&
MERINO study [32 ] in which 391 patients with BSI Ceftazidime/avibactam, whose spectrum of activ-
by Escherichia coli or K. pneumoniae not susceptible to ity also covers CRE (except metallo-b-Lactamases) may
ceftriaxone were assigned (ratio 1:1) either to the serve more appropriately as an alternative to carbape-
meropenem arm (1 g each 8 h) or to the PTZ arm nems for severe and invasive infections with ESBL or
(4.5 g each 6 h). The MERINO trial showed higher also with AmpC-producing Enterobacteriaceae [41].
mortality in the latter group (12.3%) than in the
former (3.7%); therefore the results do not support
the noninferiority of PTZ to meropenem [32 ].
&&
CARBAPENEMS FOR THE MANAGEMENT
Does the MERINO trial put an end to the use of OF P. aeruginosa INFECTIONS
‘old’ BLBLI against ESBL-E? It did, although some Acute invasive infections caused by P. aeruginosa are
points deserve further clarification [33]. associated with high mortality rates, above 40%,
First, PTZ was administered at intermittent especially in the case of MDR/XDR strains [42].
intervals, in contrast with meta-analytic evidence Generally, there has been considerable debate
of significantly better outcomes with extended/con- regarding the benefit of combination therapy over
tinuous infusion and the same goes to meropenem, monotherapy against invasive infections by P. aer-
especially in critically ill patients. [34,35]. uginosa [43]. Data from literature highlight no dif-
Second, generic lots of drugs may widely vary in ference between the two options, but it is difficult to
terms of potency in comparison with branded prod- draw inferences regarding MDR and XDR-isolates
&
ucts and could have influenced the trial’s results [36]. [44,45,46 ].
Third, ESBLs are not a homogenous group with A great problem is posed by carbapenem-resis-
relevant differences among the different ß-lacta- tant P. aeruginosa (CR-PA): death risk is significantly
mases: the prevalence of some types, poorly inhib- higher in patients infected by CR-PA than among
ited by tazobactam, may undermine the efficacy of patients infected by their sensitive counterparts, as
&
PTZ [37 ,38]. demonstrated by meta-analytic data (crude
Perhaps the main question regarding the OR ¼ 1.64; 95% CI ¼ 1.40, 1.93; adjusted OR ¼ 2.38;
MERINO trial is the higher mortality in the PTZ 95% CI ¼ 1.53, 3.69) [47].
arm, albeit the randomized sample showed, as base- Zusman et al. [48] reported in-vitro data about
line characteristics, low Pitt and Charlson Comorbid- combination therapy for MDR P. aeruginosa with a
&&
ity Index scores [32 ,33]. Furthermore, two-thirds of synergistic effect of carbapenems (especially doripe-
patients randomized to PTZ received an appropriate nem), with polymixins. Bactericidal effects (>3 logs
treatment according to susceptibility testing within reduction) increased from 10% to 49% with this
&&
5.5 h after collecting blood culture [32 ,33]; in the combination, and the result persisted even when
same arm the proportion of infections from urinary the isolate was both resistant to colistin and the
source was high (54.8%), which is traditionally the carbapenem [49,50]. New insights in the use of
main clinical scenario of ESBL invasive infections and carbapenem regimens is based on optimizing anti-
the ideal setting for PTZ [29]. In contrast, the median biotic combinations: Yadav et al. [51] reported that
MIC of PTZ was 2 mg/l, whereas values above this clinically relevant concentrations of imipenem and
threshold have been associated with the poor out- an aminoglycoside provided synergistic bacterial
&
come under PTZ therapy [37 ,38]. Of note, randomi- killing and suppression of resistance against a high
zation was performed at day 3 in patients with severe inoculum of clinical isolates of carbapenem-resis-
infections, a frame time in which the response to tant P. aeruginosa. A precious help has been repre-
empirical treatment is clinically evaluable. sented by the introduction in clinical practice of the
However, the new frontier of noncarbapenem b- new BLBLI ceftolozane/tazobactam and ceftazi-
Lactams for ESBL invasive infections, to pursuit a dime/avibactam: the first is ranked first as antipseu-
carbapenem-sparing strategy, is nowadays repre- domonal b-lactam in recent Spanish guidelines
sented by novel BLBLIs, ceftolozane/tazobactam specifically addressing acute invasive infections by
& &
and ceftazidime/avibactam [39 ,40 ,41]. P. aeruginosa [52]. Ceftolozane/tazobactam is con-
Ceftolozane/tazobactam shows good activity in sidered the main backbone of the empiric approach
vitro against ESBL, barring carbapenem-resistant phe- to suspected MDR-PA infections because of its
& &
notypes [39 ,40 ]. Recent guidelines underline its role marked stability versus many mechanisms of

4 www.co-infectiousdiseases.com Volume 32  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Carbapenems sparing strategy Corcione et al.

resistances employed by P. aeruginosa, including than IV colistin alone was fosfomycin and IV colis-
resistance to carbapenems, as long as not mediated tin; the concomitant use of IV also showed a better
by carbapenemases [53–55]. Montero et al. [56] clinical outcome than the comparator and inhaled
reported bacterial suppression using a combination colistin [67]. Fosfomycin does not display activity
of ceftolozane/tazobactam and high dose merope- per se against A. baumannii, but its synergistic effects
nem against a strain of XDR P. aeruginosa recovered with other drugs turn out to be very useful [69].
from a Spanish patient, whereas microbiological fail- The second NMA [68], compared efficacy and
ure occurred when molecules were individually safety of treatment options for MDR and XDR-AB
tested. A recent study found a synergism in vitro with infections of any anatomical site: about the clinical
a combination of ceftolozane/tazobactam with colis- cure, the combination of colistin, tigecycline and
tin or fosfomycin against MDR P. aeruginosa [57]. sulbactam was not superior to IV colistin monother-
apy. Regarding all-cause mortality, triple combination
therapy based on colistin, tigecycline and a third drug
CARBAPENEMS FOR THE MANAGEMENT ranked first, but it was not superior to IV colistin
OF Acinetobacter baumannii INFECTIONS monotherapy (RR ¼ 0.27; 95% CI ¼ 0.02–4.09); more-
Carbapenems, the cornerstone of anti-acinetobacter over, tigecycline based-therapies were far less effective
baumannii susceptible strains [58,59], show highly when considering microbiological eradication (in this
bactericidal activity in many clinical scenarios case sulbactam monotherapy had the highest ranking)
(pneumonia, bloodstream infections – BSI) but also or important subgroup such as BSI patients [68].
reported vital factors for the development of CR-AB A very recent multicentre RCT had tested the
strains [60]. superiority of meropenem and colistin versus colistin
Colistin probably represents the paramount alone against carbapenem-resistant Gram-negative
weapon in the armamentarium in these cases, but bacteria as a cause of severe infections [70]. Overall,
its toxicity often poses relevant challenges [61] on no difference in terms of clinical failure was observed
the other hand tigecycline, is intriguing alternative, between the two arms, and this was confirmed in the
but its use has several limitations [62]: bacteriosta- subgroup (widely prevalent, 312 patients) of CR-AB
tism, suboptimal lung penetration and it received a infections [70]. Interestingly, in a subsequent analysis
black box warning by the Food and Drug Adminis- of this trial only concerning CR-AB cases, the death
tration [62,63]. rate was significantly lower in patients with infections
Currently, carbapenems are often employed in by isolates resistant also to colistin (OR ¼ 0.285; 95%
the case of CR-AB infection in the framework of CI ¼ 0.118–0.686), probably because of a loss of fitness
combination regimens, using the synergistic activity and virulence by the bacterial strains [71]. The real
with other drugs such as colistin [64,65]. The advan- unexpected result was the higher mortality, in this
tage of combination therapies over monotherapies subset of patients, associated with the addition of
against MDR and XDR-AB has not been established, meropenem to colistin, compared with colistin mono-
as showed, for instance, by the seminal Italian RCT therapy (OR ¼ 3.065; 95% CI ¼ 1.021–9.202) [70,71].
comparing colistin versus colistin and rifampin for The explanation was unclear: the authors suggested a
life-threatening infections because of XDR-AB: in possible modification in gene expression induced by
spite of an higher microbiological eradication rate, carbapenems, thereby restoring bacterial virulence
the addition of rifampin did not reduce 30-day mor- overcoming the fitness cost of colistin-resistance
tality [65]. Lopes-Cortes et al. [66], in their multi- [70,71].
centre prospective cohort did not find benefits in
clinical outcome from combination therapy.
Recently two network meta-analyses (NMA), have CARBAPENEMS FOR THE TREATMENT OF
tried to identify the best antimicrobial regimen against CARBAPENEM-RESISTANT
difficult-to-treat A. baumannii infections [67,68]. Enterobacteriaceae
The first one, focusing on pneumonia in criti-
cally ill patients by MDR or XDR-AB was developed Carbapenem paradox: high dosage and
upon intravenous (IV) colistin as a common com- double carbapenem
parator and data showed that the best treatment for The time the plasma drug concentration is main-
reducing all-cause mortality was the treatment with tained above the MIC (t>MIC) is the most crucial
sulbactam monotherapy [67]. Furthermore, sulbac- pharmacodynamics parameter predicting in-vivo
tam was significantly superior to the comparator efficacy of b-lactams, including carbapenems [72].
for lowering all-cause mortality (OR ¼ 0.27; 95% In general, for a bacteriostatic and bactericidal
CI ¼ 0.06–0.91). Of note, the only combination effect, the carbapenems require a t more than
therapy based on two different drugs ranking higher MIC of approximately 20% and 40%, respectively

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com 5

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Gram-negative infections

[72,73]. For CRE exhibiting MIC more than 4 mg/l, Carbapenems as part of combination therapy
the extended infusion of meropenem over 3–4 h at for carbapenem-resistant
the standard dose of 1 g every 8 h provides higher Enterobacteriaceae
probabilities of target attainment than conven- The role of carbapenems in the treatment of CRE has
tional 30 min infusion [74]. Furthermore, the same long been debated: some studies reported lower
dosing infused over 4 h may provide adequate expo- mortality in patients treated with high dose mero-
sure for organisms with MICs more than 8 mg/l penem combined with other agents for CR-Kp infec-
[74,75]. These studies suggested that prolonging tions when the carbapenem MIC was more than
the infusion time for over 1 h allowed target attain- 8 mg/l [87,88]. Results by Del Bono et al. [79] suggest
ment 1 dilution higher than shorter infusions [76]. that meropenem might retain some bactericidal
High-dose meropenem (i.e., 2 g every 8 h) may be activity on CR-Kp strain with up to 32 mg/l in some
able to achieve better PK/pharmacodynamics target cases. Synergisms between carbapenem and co-
than standard dose [77,78]. One study reporting 4 h administered agents could have supported this use
infusions of this regimen have shown to achieve even if PK/pharmacodynamics targets were not
adequate exposure for organisms with MICs up to reached, in line with previous reports of synergistic
16 mg/l [78]. Continuous infusions strategies of mer- effects with meropenem [89].
openem following a 3 g daily dose have been found to The introduction of the new b-lactamase inhib-
achieve median concentrations of 7 mg/l. By increas- itor ceftazidime/avibactam has changed the clinical
ing this dosage to 6 g in three doses may provide scenario (Fig. 1). Several case series described a better
adequate exposure for organisms with MICs up to survival at 30 and 90 days in patients treated with
16 mg/l [79]. Giannella et al. [80] pointed out that ceftazidime/avibactam-based regimens than carba-
high-dose carbapenem-containing regimens were penem and other agents, including patients treated
associated with better outcomes in CRE-related infec- with ceftazidime/avibactam monotherapy [90].
tions. In 595 patients with CR-Kp BSI, analyzed, 77% Tumbarello et al. [91] recently found similar results
of isolates showed a carbapenem MIC at least 16 mg/l, in a retrospective series of patients with CP-Kp
428 (71.9%) received HD carbapenem-based combi- infections treated with ceftazidime/avibactam
nation therapy. HD carbapenem use (HR ¼ 0.69, 95% within a compassionate-access-program. Neverthe-
CI ¼ 0.47–1.00, P ¼ 0.05) was protective factor and less, in a study of 37 patients infected by CRE,
adjusted for the propensity score, showed a greater Schields et al. [92] described low clinical success,
protective effect (HR ¼ 0.64, 95% CI ¼ 0.43–0.95, especially in patients with pneumonia because of
P ¼ 0.03). After stratification benefit of HD carbape- CRE treated by ceftazidime/avibactam without dif-
nem was also observed for strains with carbapenem ference between monotherapy and combination
MIC at least 16 mg/l [80]. regimen. Ceftazidime/avibactam use produced
Following the first reports of patients with infec- encouraging results in the CR-Kp BSIs with thirty-
tions because of carbapenemases-producing Entero- day survival rates of 59.1% in monotherapy versus
bacteriaceae treated with a double carbapenem 63.1% with a carbapenem associated [91,92].
combination, a growing number of case series have
been published, accompanied by observational stud-
ies conducted in Europe and in the United States [81– NEW CARBAPENEM COMBINATIONS
84] despite that the sample size is frequently small The introduction of the novel b-lactamase inhibitors
and RCTs are lacking. The rationale of such a combi- vaborbactam and relebactam, has provided additional
nation is based on the possibility that the adminis- therapeutic options for CRE infections [93]. Of note,
tration of ertapenem, will serve as a suicidal inhibitor, these new b-lactamase inhibitors are not active against
allowing the second carbapenem to reach concen- all major carbapenemases, but these novel b-lacta-
trations exceeding the MIC or a sort of ‘double-hit’ in mase inhibitor combinations will also provide carba-
which the first antibiotic should reduce bacterial load penem-sparing options for the treatment of MDR
in the site of infection, allowing the second medica- bacteria. The combination of meropenem with a
tion acting on a decreased bacterial volume [81,82]. boronic acid beta-lactamase, vaborbactam (M/V),
The proposed two carbapenems-based schemes may has shown an increase in-vitro activity against class
be supported by a third antibiotic from a different A and class C serine b-lactamases, although Class B
class (polymyxins, aminoglycosides, tigecycline, and (e.g., NDM, VIM) and class D (e.g., OXA-48) carbape-
fluoroquinolones) [85,86]. With the advent of new nemases are not inhibited by vaborbactam [93]. The
molecules, double-carbapenem regimens have been efficacy, tolerability and safety of M/V was first
partly set aside, and it can be considered as salvage assessed in TANGO I [94] a multicenter, double-blind,
therapy when no other options are available. randomized, phase III, noninferiority trial versus P/T

6 www.co-infectiousdiseases.com Volume 32  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Carbapenems sparing strategy Corcione et al.

for the treatment of cUTIs and TANGO II [95], in and imipenem/cilastatin (COL þ IMI) in patients
which M/V was compared to best available treatment with imipenem-nonsusceptible bacterial infec-
(BAT) in a series of infections because of suspected or tions, including HABP, VABP, cIAI and cUTIs [98].
known CRE (notably cUTI, hospital-acquired or ven- In the RESTORE-IMI 2 trial I/R was compared to PTZ
tilator-associated pneumonia – HABP/VABP, compli- (both arms with the addition of Linezolid) in HABP/
cated intrabdominal infections – cIAI, bacteremia): VABP [99].
because of superiority of M/V compared to BAT this Nacubactam is a novel member of the diaza-
study was terminated prematurely. A phase III trial bicyclooctane inhibitor family with a dual-mode
called TANGO III, in which M/V was compared to PTZ of action, acting as a b-lactamase inhibitor and an
for the treatment of HABP/VABP, was withdrawn by antibacterial agent employing PBP2 inactivation
sponsor decision [96]. [100]. The combination of meropenem with nacu-
Imipenem/cilastatin with relebactam (I/R), bactam is still in a phase I status but is potentially
combines an approved carbapenem with a novel active against carbapenem-resistant Enterobacter-
b-lactamase inhibitor [93]. The in-vitro addition of iaceae isolates expressing serine b-lactamases
relebactam showed increased activity of imipenem (theoretically also ceftazidime/avibactam resis-
against ESBLs, serine b-lactamases, and P. aerugi- tant strains), OXA-48 and metallo b-lactamases
nosa; however, relebactam cannot inhibit Class D [101].
OXA-48 [93]. A phase II study assessed efficacy,
tolerability and safety I/R versus imipenem/cilasta-
tin in cIAI and cUTI [97], furthermore two phase III ANTIMICROBIAL STEWARDSHIP OF
study, RESTORE-IMI 1 [98] and RESTORE-IMI 2 [99], CARBAPENEMS
are currently evaluating I/R in different settings. Carbapenems are a powerful weapon against Gram-
RESTORE-IMI 1 is a multicenter, randomized, dou- negative bacteria but sparing carbapenems usage
ble-blind, comparator-controlled trial, comparing can have interesting microbiological effects as well
the efficacy and safety of IMI/REL versus colistin [102] (Fig. 2).

Empiric Treament
ESBL
Targeted Treament
ESBL

Empiric Treament
CR-E
If High Risk Of CR-E
Colonizaon Empiric Treament
CR-E
If low risk of CR-E
Colonizaon

Targeted Treament
CR-E

Sparing Favourable
Carbapenems

FIGURE 2. Role of carbapenem in the setting of empiric or target therapy for extended-spectrum b-lactamase producing and
carbapenem-resistant Enterobacteriaceae (CR-E).

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com 7

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Gram-negative infections

Gut colonization is the main reservoir of CR-Kp CONCLUSION


among patients, and intestinal carriage is associated In the current context of the MDR pandemic and
with increased risk for developing successive CRE increasing opportunities for carbapenem-sparing
infections [103]. Not surprisingly, carbapenems use initiatives, the respective impacts of relevant clinical
is among the critical determinant for CRE acquisi- parameters, such as dosing regimen, combination
tion, is associated with a higher rate of superinfec- therapies with nonb-lactam drugs, length of treat-
tion compared with noncarbapenem therapies and a ment exposure and the sequence of antimicrobial
higher risk of developing resistance through selec- use, should be precisely investigated to better define
tive pressure that gives resistant strains the chance the positioning of carbapenem-sparing b-lactams
to overwhelm their sensitive counterparts. [104]. in antimicrobial stewardship programs and de-
Several strategies for wiser use of carbapenem are escalation algorithms for patients.
needed: antimicrobial stewardship program should
be focused on the education of physicians in
addressing the indication for using carbapenems Acknowledgements
and when and how treatment can be de-escalated None.
[105].
From an antimicrobial stewardship perspective, Financial support and sponsorship
the lower 30-day mortality with carbapenems
described in the MERINO trial could support the None.
hypothesis of saving carbapenems as a target ther-
apy for patients at high risk for ESBL infections, Conflicts of interest
therefore reducing their use as empiric therapy, a There are no conflicts of interest.
&&
setting which PTZ may still have a role [32 ].
Another focal point is therapy duration and the
role of de-escalation. Recent data point out that REFERENCES AND RECOMMENDED
short-course treatment (< 10 days) for Enterobacter- READING
iaceae BSI does not worsen clinical outcomes [106] Papers of particular interest, published within the annual period of review, have
been highlighted as:
and this has also been confirmed in a setting & of special interest
wherein the proportion of ESBL was more than && of outstanding interest

25% [107]. A Japanese study in another setting 1. Bassetti M, Poulakou G, Ruppe E, et al. Antimicrobial resistance in the next
endemic for ESBL and investigating a cohort of 30 years, humankind, bugs and drugs: a visionary approach. Intensive Care
Med 2017; 43:1464–1475.
300 patients showed that a de-escalation strategy 2. Naylor NR, Atun R, Zhu N, et al. Estimating the burden of antimicrobial
from carbapenems, whenever possible according to resistance: a systematic literature review. Antimicrob Resist Infect Control
2018; 7:58.
predefined criteria related to either empiric or tar- 3. Cassini A, Högberg LD, Plachouras D, et al. Attributable deaths and dis-
geted treatment, did not impair clinical outcomes, ability-adjusted life-years caused by infections with antibiotic-resistant bac-
teria in the EU and the European Economic Area in 2015: a population-level
achieving instead a reduction of CDI and of CR-AB modelling analysis. Lancet Infect Dis 2019; 56–66.
acquisition [108]. 4. Centers for Disease Control and Prevention (CDC) Biggest threats. Avail-
able at https://www.cdc.gov/drugresistance/biggest_threats.html
Experiences from other countries all converge 5. Bassetti M, Russo A, Carnelutti A, et al. Antimicrobial resistance and
on the feasibility of carbapenem-sparing strategies, treatment: an unmet clinical safety need. Expert Opin Drug Saf 2018;
17:669–680.
displaying a favorable impact on clinical, economic, 6. Magiorakos AP, Srinivasan A, Carey RB, et al. Multidrug-resistant, extensively
and ecological outcomes [109,110]. Of note, the drug-resistant and pandrug-resistant bacteria: an international expert pro-
posal for interim standard definitions for acquired resistance. Clin Microbiol
studies mentioned above were carried out before Infect 2012; 18:268–281.
the marketing of new drugs that can act as valid 7. Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic steward-
ship program: guidelines by the Infectious Diseases Society of America and
alternatives to carbapenems in serious infections the Society for Healthcare Epidemiology of America. Clin Infect Dis 2016;
wherein the latter are the benchmark as therapeutic 62:e51–e77.
8. de With K, Allerberger F, Amann S, et al. Strategies to enhance rational use of
options, namely invasive infections by ESBL. antibiotics in hospital: a guideline by the German Society for Infectious
Moreover, the selective pressure exerted by Diseases. Infection 2016; 44:395–439.
9. Viale P, Giannella M, Bartoletti M, et al. Considerations about antimicrobial
antimicrobials on the gut microbiota promotes stewardship in settings with epidemic extended-spectrum b-lactamase-pro-
the carriage of MDR bacteria and recent data ducing or carbapenem-resistant enterobacteriaceae. Infect Dis Ther 2015;
4(Suppl 1):65–83.
described as carbapenems are a stronger driver of 10. WHO Guidelines Approved by the Guidelines Review Committee. Guide-
intestinal dysbiosis [111,112]: saving carbapenems lines for the prevention and control of carbapenem-resistant enterobac-
teriaceae, Acinetobacter baumannii and Pseudomonas aeruginosa in
in favor of certain nonb-lactam drugs with limited Healthcare Facilities. Geneva: World Health Organization; 2017. Avail-
impact on the intestinal microbiota, when feasible, able at: https://www.who.int/infection-prevention/publications/guide-
lines-cre/en/
may also have a role in the management of MDR 11. Wilson H, Török ME. Extended-spectrum b-lactamase-producing and car-
infections. bapenemase-producing Enterobacteriaceae. Microb Genom 2018; 4.

8 www.co-infectiousdiseases.com Volume 32  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Carbapenems sparing strategy Corcione et al.

12. Porreca AM, Sullivan KV, Gallagher JC. The epidemiology, evolution, and 35. Yu Z, Pang X, Wu X, et al. Clinical outcomes of prolonged infusion
treatment of KPC-producing organisms. Curr Infect Dis Rep 2018; 20:13. (extended infusion or continuous infusion) versus intermittent bolus of
13. Bush K. Past and present perspectives on b-lactamases. Antimicrob Agents meropenem in severe infection: a meta-analysis. PLoS One 2018;
Chemother 2018; 62:e01076-18. 13:e0201667.
14. Walsh TR. The emergence and implications of metallo-beta-lactamases in 36. Jones RN, Sader HS, Flamm RK, et al. More potency assay results for generic
Gram-negative bacteria. Clin Microbiol Infect 2005; 11(Suppl 6):2–9. non-USA lots of piperacillin/tazobactam and initial reports for generic mer-
15. Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-beta-lactamases: the openem compounds marketed in the USA. Diagn Microbiol Infect Dis 2013;
quiet before the storm? Clin Microbiol Rev 2005; 18:306–325. 76:110–112.
16. Jacoby GA. AmpC B-Lactamases. Clin Microbiol Rev 2009; 22:161–182. 37. Scheuerman O, Schechner V, Carmeli Y, et al. Comparison of predictors and
17. Evans BA, Amyes SG. OXA b-lactamases. Clin Microbiol Rev 2014; & mortality between bloodstream infections caused by ESBL-producing Es-
27:241–263. cherichia coli and ESBL-producing Klebsiella pneumoniae. Infect Control
18. Poole K. Efflux-mediated antimicrobial resistance. J Antimicrob Chemother Hosp Epidemiol 2018; 39:660–667.
2005; 56:20–51. Comparing the epidemiology, clinical characteristics, and mortality of patients with
19. Livermore DM. Multiple mechanisms of antimicrobial resistance in Pseudo- BSI caused by ESBL-producing Escherichia coli (ESBL-EC) versus ESBL-produ-
monas aeruginosa: our worst nightmare? Clin Infect Dis 2002; 34:634–640. cing Klebsiella pneumoniae (ESBL-KP) and to examine the differences in clinical
20. Pagès JM, James CE, Winterhalter M. The porin and the permeating anti- characteristics and outcome between BSIs caused by isolates with CTX-M versus
biotic: a selective diffusion barrier in Gram-negative bacteria. Nat Rev other ESBL genotypes.
Microbiol 2008; 6:893–903. 38. Rodriguez-Bano J, Navarro MD, Retamar P, et al. Beta-lactam/beta-lactam
21. Iredell J, Brown J, Tagg K. Antibiotic resistance in Enterobacteriaceae: inhibitor combinations for the treatment of bacteremia due to extended-
mechanisms and clinical implications. BMJ 2016; 352:h6420. spectrum beta-lactamase-producing Escherichia coli: a post hoc analysis of
22. Rodrı́guez-Baño J. The times they are a-changing: carbapenems for ex- prospective cohorts. Clin Infect Dis 2012; 54:167–174.
& tended-spectrum-b-lactamase-producing bacteria. Antimicrob Agents Che- 39. Gentile I, Maraolo AE, Borgia G. What is the role of the new b-lactam/
mother 2015; 59:5095–5096. & b-lactamase inhibitors ceftolozane/tazobactam and ceftazidime/avibactam?
Several antimicrobial agents are being investigated as alternatives to carbapenems Expert Rev Anti Infect Ther 2016; 14:875–878.
in the treatment of infections caused by ESBL-producing Enterobacteriaceae, Place in therapy, strengths, and weakness of new b-lactam/b-lactamases inhibi-
which may be useful in avoiding overuse of carbapenems in the context of recent tors.
global spread of carbapenem-resistant Enterobacteriaceae. The most promising 40. Montravers P, Bassetti M. The ideal patient profile for new beta-lactam/beta-
candidates for invasive infections so far are b-lactam/b-lactamase inhibitor com- & lactamase inhibitors. Curr Opin Infect Dis 2018; 31:587–593.
binations and cephamycins. The purpose of this study was to provide insight concerning the patients who
23. Wilson AP. Sparing carbapenem usage. J Antimicrob Chemother 2017; would constitute ideal candidates to receive these new BLBLI combinations.
72:2410–2417. 41. Sternbach N, Leibovici-Weissman Y, Avni T, et al. Efficacy and safety of
24. Rodrı́guez-Baño J, Gutierrez-Gutierrez B, Machuca I, et al. Treatment of ceftazidime/avibactam: a systematic review and meta-analysis. J Antimicrob
&& infections caused by extended-spectrum-beta-lactamase-, AmpC-, and car- Chemother 2018; 73:2021-9.
bapenemase-producing enterobacteriaceae. Clin Microbiol Rev 2018; 42. Maraolo AE, Cascella M, Corcione S, et al. Management of multidrug-
31:e00079-17. resistant Pseudomonas aeruginosa in the intensive care unit: state of the
A complete and updated guide for Enterobacteriaceae-related infections in the era art. Expert Rev Anti Infect Ther 2017; 15:861–871.
of multidrug resistance infection in which regimens must be individualized accord- 43. Paul M, Leibovici L. Editorial commentary: combination therapy for Pseudo-
ing to the susceptibility profile, type, and severity of infection and the features of the monas aeruginosa bacteremia: where do we stand? Clin Infect Dis 2013;
patient. 57:217–220.
25. Tamma PD, Rodrı́guez-Baño J. The use of noncarbapenem b-lactams for the 44. Morata L, Cobos-Trigueros N, Martinez JA, et al. Influence of multidrug
treatment of extended-spectrum b-lactamase infections. Clin Infect Dis resistance and appropriate empirical therapy on the 30-day mortality rate
2017; 64:972–980. of Pseudomonas aeruginosa bacteremia. Antimicrob Agents Chemother
26. Kim SA, Altshuler J, Paris D, et al. Cefepime versus carbapenems for the 2012; 56:4833–4837.
treatment of urinary tract infections caused by extended-spectrum b-lacta- 45. El Amari EB, Chamot E, Auckenthaler R, et al. Influence of previous
mase-producing enterobacteriaceae. Int J Antimicrob Agents 2018; exposure to antibiotic therapy on the susceptibility pattern of Pseudomo-
51:155–158. nas aeruginosa bacteremic isolates. Clin Infect Dis 2001;
27. Jiménez-Guerra G, Heras-Cañas V, Béjar Molina LD, et al. Extended-spec- 33:1859 –1864.
trum beta-lactamase-producing Escherichia coli and Klebsiella pneumoniae 46. Hu Y, Li L, Li W, et al. Combination antibiotic therapy versus monotherapy for
from urinary tract infections: evolution of antimicrobial resistance and treat- & Pseudomonas aeruginosa bacteraemia: a meta-analysis of retrospective and
ment options. Med Clin (Barc) 2018; 150:262–265. prospective studies. Int J Antimicrob Agents 2013; 42:492–496.
28. Vardakas KZ, Tansarli GS, Rafailidis PI, Falagas ME. Carbapenems versus The aim of this review was to compare both types of therapy to determine which
alternative antibiotics for the treatment of bacteraemia due to Enterobacter- delivers the best outcome for P. aeruginosa bacteremia.
iaceae producing extended-spectrum b-lactamases: a systematic review and 47. Liu Q, Li L, Li W, et al. Influence of carbapenem resistance on mortality of
meta-analysis. J Antimicrob Chemother 2012; 67:2793–2803. patients with Pseudomonas aeruginosa infection: a meta-analysis. Sci Rep
29. Son SK, Lee NR, Ko JH, et al. Clinical effectiveness of carbapenems versus 2015; 5:11715.
alternative antibiotics for treating ESBL-producing Enterobacteriaceae bac- 48. Zusman O, Avni T, Leibovici L, et al. Systematic review and meta-analysis of in
teraemia: a systematic review and meta-analysis. J Antimicrob Chemother vitro synergy of polymyxins and carbapenems. Antimicrob Agents Chemother
2018; 73:2631–2642. 2013; 57:5104–5111.
30. Harris PN, Tambyah PA, Paterson DL. b-lactam and b-lactamase inhibitor 49. Tängdén T, Karvanen M, Friberg LE, et al. Assessment of early combination
& combinations in the treatment of extended-spectrum b-lactamase producing effects of colistin and meropenem against Pseudomonas aeruginosa and
Enterobacteriaceae: time for a reappraisal in the era of few antibiotic Acinetobacter baumannii in dynamic time-kill experiments. Infect Dis 2017;
options? Lancet Infect Dis 2015; 15:475–485. 49:521–527.
Examination of the why BLBLIs are perceived as inferior in the treatment of 50. Aoki N, Tateda K, Kikuchi K, et al. Efficacy of colistin combination therapy in a
infection with ESBL producers, and discuss data that suggest these concerns mouse model of pneumonia caused by multidrug-resistant Pseudomonas
might not be strongly supported by clinical evidence. aeruginosa. J Antimicrob Chemother 2009; 63:534–542.
31. Sfeir MM, Askin G, Christos P, et al. Beta-lactam/beta-lactamase inhibitors 51. Yadav R, Bulitta JB, Schneider EK, et al. Aminoglycoside concentrations
versus carbapenem for bloodstream infections due to extended-spectrum required for synergy with carbapenems against Pseudomonas aeruginosa
beta-lactamase-producing Enterobacteriaceae: systematic review and meta- determined via mechanistic studies and modeling. Antimicrob Agents Che-
analysis. Int J Antimicrob Agents 2018; 52:554–570. mother 2017; 61:e00722-17.
32. Harris PN, Tambyah PA, Lye DC, et al. Effect of piperacillin-tazobactam vs 52. Mensa J, Barberan J, Soriano A, et al. Antibiotic selection in the treatment
&& meropenem on 30-day mortality for patients with E. coli or Klebsiella of acute invasive infections by Pseudomonas aeruginosa: guidelines by
pneumoniae bloodstream infection and ceftriaxone resistance: a randomized the Spanish Society of Chemotherapy. Rev Esp Quimioter 2018;
clinical trial. JAMA 2018; 320:984–994. 31:78–100.
Before MERINO, ‘Old’ BLBLIs (especially upon PTZ) in observational studies 53. Wright H, Bonomo RA, Paterson DL, et al. New agents for the treatment of
definitely did not show their inferiority when compared with carbapenems against infections with Gram-negative bacteria: restoring the miracle or false dawn?
ESBL-E infections in terms of mortality. Clin Microbiol Infect 2017; 23:704–712.
33. Bonten M, An endless one-sided confidence in Pip-tazo? 21/09/2018 54. Bassetti M, Castaldo N, Cattelan A, et al. Ceftolozane/tazobactam for the
https://reflectionsipc.com/2018/09/21/an-endless-one-sided-confidence- treatment of serious P. aeruginosa infections: a multicenter nationwide
in-pip-tazo/ clinical experience. Int J Antimicrob Agents 2018; 53:408–415.
34. Yang H, Zhang C, Zhou Q, et al. Clinical outcomes with alternative dosing 55. Gallagher JC, Satlin MJ, Elabor A, et al. Ceftolozane-tazobactam for the
strategies for piperacillin/tazobactam: a systematic review and meta-analysis. treatment of multidrug-resistant Pseudomonas aeruginosa infections: a
PLoS One 2015; 10:e0116769. multicenter study. Open Forum Infect Dis 2018; 5:ofy280.

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com 9

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Gram-negative infections

56. Montero M, VanScoy BD, LópezCausapé C, et al. Evaluation of ceftolozane- 78. Roberts JA, Kirkpatrick CM, Roberts MS, et al. Meropenem dosing in critically
tazobactam in combination with meropenem against Pseudomonas aerugi- ill patients with sepsis and without renal dysfunction: intermittent bolus
nosa sequence type 175 in a hollow-fiber infection model. Antimicrob Agents versus continuous administration? Monte Carlo dosing simulations and
Chemother 2018; 62:e00026-18. subcutaneous tissue distribution. J Antimicrob Chemother 2009; 64:
57. Alfouzan W, Dhar R, Nicolau DP. In vitro activity of newer and conventional 142–150; 69.
antimicrobial agents, including fosfomycin and colistin, against selected 79. Del Bono V, Giacobbe DR, Marchese V, et al. Meropenem for treating KPC-
Gram-negative bacilli in Kuwait. Pathogens 2018; 7:75. producing Klebsiella pneumoniae bloodstream infections: should we get to
58. Lemos EV, Hennon SW, Feldman MF, et al. Carbapenem resistance and the PK/PD root of the paradox? Virulence 2017; 8:66–73.
mortality in patients with Acinetobacter baumannii infection: systematic 80. Giannella M, Trecarichi EM, Giacobbe DR, et al. Italian Study Group on
review and meta-analysis. Clin Microbiol Infect 2014; 20:416–423. Resistant Infections of the Societa‘ Italiana Terapia Antinfettiva (ISGRISITA).
59. Tal-Jasper R, Katz DE, Amrami N, et al. Clinical and epidemiological sig- Effect of combination therapy containing a high-dose carbapenem on mor-
nificance of carbapenem resistance in Acinetobacter baumannii infections. tality in patients with carbapenem-resistant Klebsiella pneumoniae blood-
Antimicrob Agents Chemother 2016; 60:3127–3131. stream infection. Int J Antimicrob Agents 2018; 51:244–248.
60. Lim CL, Chua AQ, Teo JQ, et al. Importance of control groups when 81. DeRyke CA, Banevicius MA, Fan HW, et al. Bactericidal activities of
delineating antibiotic use as a risk factor for carbapenem resistance, ex- meropenem and ertapenem against extended-spectrum-beta-lactamase-pro-
treme-drug resistance, and pan-drug resistance in Acinetobacter baumannii ducing Escherichia coli and Klebsiella pneumoniae in a neutropenic mouse
and Pseudomonas aeruginosa: a systematic review and meta-analysis. Int J thigh model. Antimicrob Agents Chemother 2007; 51:14811486.
Infect Dis 2018; 76:48–57. 82. Bulik CC, Nicolau DP. Double-carbapenem therapy for carbapenemase-
61. El Chakhtoura NG, Saade E, Iovleva, et al. Therapies for multidrug resistant producing Klebsiella pneumoniae. Antimicrob Agents Chemother 2011;
and extensively drug-resistant nonfermenting Gram-negative bacteria caus- 55:3002–3004.
ing nosocomial infections: a perilous journey toward ’molecularly targeted’ 83. Ceccarelli G, Falcone M, Giordano A, et al. Successful ertapenem-doripe-
therapy. Expert Rev Anti Infect Ther 2018; 16:89–110. nem combination treatment of bacteremic ventilator-associated pneumonia
62. Falagas ME, Vardakas KZ, Kapaskelis A, et al. Tetracyclines for multidrug- due to colistin resistant KPC-producing Klebsiella pneumoniae. Antimicrob
resistant Acinetobacter baumannii infections. Int J Antimicrob Agents 2015; Agents Chemother 2013; 57:2900–2901.
45:455–460. 84. Oliva A, D’Abramo A, D’Agostino C, et al. Synergistic activity and effective-
63. U.S. Federal Drug Administration (FDA) [Internet]. Drug Safety Communica- ness of a double-carbapenem regimen in pandrug-resistant Klebsiella pneu-
tion: FDA warns of increased risk of death with IV antibacterial Tygacil moniae bloodstream infections. J Antimicrob Chemother 2014;
(tigecycline) and approves new Boxed Warning. Washington (DC): FDA; 69:1718–1720.
2016 [Last access: December 2018]. Available from: https://www.fda.gov/ 85. Chua NG, Zhou YP, Tan TT, et al. Polymyxin B with dual carbapenem
Drugs/DrugSafety/ucm369580.htm combination therapy against carbapenemase-producing Klebsiella pneumo-
64. Liu X, Zhao M, Chen Y, et al. Synergistic killing by meropenem and colistin niae. J Infect 2015; 70:309–311.
combination of carbapenem-resistant Acinetobacter baumannii isolates from 86. Oliva A, Scorzolini L, Castaldi D, et al. Double-carbapenem regimen, alone
Chinese patients in an in vitro pharmacokinetic/pharmacodynamic model. Int or in combination with colistin, in the treatment of infections caused
J Antimicrob Agents 2016; 48:559–563. by carbapenem-resistant Klebsiella pneumoniae (CR-Kp). J Infect 2017;
65. Durante Mangoni E, Signoriello G, Andini R, et al. Colistin and rifampicin 74:103–106.
compared with colistin alone for the treatment of serious infections due to 87. Thomson KS. Double-carbapenem therapy not proven to be more active than
extensively drug-resistant Acinetobacter baumannii: a multicenter, rando- carbapenem monotherapy against KPC positive Klebsiella pneumoniae.
mized clinical trial. Clin Infect Dis 2013; 57:349–358. Antimicrob Agents Chemother 2012; 56:4037.
66. Lopez-Cortes LE, Cisneros JM, Fernandez-Cuenca F, et al. Monotherapy 88. Tumbarello M, Losito A, Giamarellou H. Optimizing therapy in carbapenem-
versus combination therapy for sepsis due to multidrug-resistant Acineto- resistant Enterobacteriaceae infections. Curr Opin Infect Dis 2018;
bacter baumannii: analysis of a multicentre prospective cohort. J Antimicrob 31:566–577.
Chemother 2014; 69:3119–3126. 89. Woerther PL, Raphaël L, Charles B, et al. Carbapenems and alternative b-
67. Jung SY, Lee SH, Lee SY, et al. Antimicrobials for the treatment of drug- lactams for the treatment of infections due to extended-spectrum b-lacta-
resistant Acinetobacter baumannii pneumonia in critically ill patients: a mase-producing Enterobacteriaceae: what impact on intestinal colonisation
systemic review and Bayesian network meta-analysis. Crit Care 2017; resistance? Int J Antimicrob Agents 2018; 52:762–770.
21:319. 90. Temkin E, Torre-Cisneros J, Beovic B, et al. Ceftazidime-avibactam as
68. Kengkla K, Kongpakwattana K, Saokaew S, et al. Comparative efficacy and salvage therapy for infections caused by carbapenem-resistant organisms.
safety of treatment options for MDR and XDR Acinetobacter baumannii Antimicrob Agents Chemother 2017; 61:e01964-16.
infections: a systematic review and network meta-analysis. J Antimicrob 91. Tumbarello M, Trecarichi EM, Corona A et al., Efficacy of Ceftazidime-
Chemother 2018; 73:22–32. Avibactam Salvage Therapy in Patients With Infections Caused by Klebsiella
69. Leelasupasri S, Santimaleeworagun W, Jitwasinkul T, et al. Antimicrobial pneumoniae Carbapenemase–producing K. pneumoniae, CID 2019:68
Susceptibility among Colistin, Sulbactam, and Fosfomycin and a Synergism 92. Shields RK, Nguyen MH, Chen L, et al. Ceftazidime-avibactam is superior
Study of Colistin in Combination with Sulbactam or Fosfomycin against to other treatment regimens against carbapenem-resistant Klebsiella
clinical isolates of carbapenem-resistant Acinetobacter baumannii. J Pathog pneumoniae bacteremia. Antimicrob Agents Chemother 2017; 61:
2018; 2018:3893492. e00883-17.
70. Paul M, Daikos GL, Durante-Mangoni E, et al. Colistin alone versus colistin 93. Zhanel GG, Lawrence CK, Adam H, et al. Imipenem-relebactam and mer-
plus meropenem for treatment of severe infections caused by carbapenem- openem-vaborbactam: two novel carbapenem-b-lactamase inhibitor combi-
resistant Gram-negative bacteria: an open-label, randomised controlled trial. nations. Drugs 2018; 78:65–98. doi: 10.1007/s40265-017-0851-9.
Lancet Infect Dis 2018; 18:391–400. Review.
71. Dickstein Y, Lellouche J, Dalak Amar MB, et al. Treatment outcomes of 94. Bidair M, Zervos M, Sagan OS et al. Clinical outcomes in adults with
colistin and carbapenem-resistant Acinetobacter baumannii infections: an complicated urinary tract infections (cUTI), including acute pyelonephritis
exploratory subgroup analysis of a randomized clinical trial. Clin Infect Dis (AP) in TANGO 1, a phase 3 randomized, double-blind, double-dummy trial
2018. doi: 10.1093/cid/ciy988. comparing meropenem–vaborbactam (M-V) with piperacillin-tazobactam (P-
72. Kuti JL, Dandekar PK, Nightingale CH, et al. Use of Monte Carlo simulation to T) [abstract no. P1289 plus poster]. In: 27th European Congress of Clinical
design an optimized pharmacodynamic dosing strategy for meropenem. J Microbiology and Infectious Diseases; 2017; Vienna
Clin Pharmacol 2003; 43:1116–1123; 63. 95. Kaye KS, Vazquez J, Mathers A et al. Clinical outcomes of serious infections
73. Krueger WA, Bulitta J, Kinzig-Schippers M, et al. Evaluation by Monte Carlo due to carbapenem-resistant enterobacteriaceae (CRE) in TANGO II a phase
simulation of the pharmacokinetics of two doses of meropenem administered 3 randomized multinational open-label trial of meropenem–vaborbactam (M-
intermittently or as a continuous infusion in healthy volunteers. Antimicrob V) versus best available therapy (BAT). [abstract no. 1862]. In: IDWeek;
Agents Chemother 2005; 49:1881–1889; 64. 2017; San Diego
74. Frei CR, Wiederhold NP, Burgess DS. Antimicrobial breakpoints for Gram- 96. A Study of Meropenem-Vaborbactam Versus Piperacillin/Tazobactam in
negative aerobic bacteria based on pharmacokinetic-pharmacodynamic Participants With Hospital-Acquired and Ventilator-Associated Bacterial
models with Monte Carlo simulation. J Antimicrob Chemother 2008; Pneumonia (TANGOIII), Clinicaltrials.gov, NCT02687906
61:621–628; 65. 97. Lucasti C, Vasile L, Sandesc D, et al. Phase 2, dose-ranging study
75. DeRyke CA, Kuti JL, Nicolau DP. Reevaluation of current susceptibility of relebactam with imipenem-cilastatin in subjects with complicated
breakpoints for Gram-negative rods based on pharmacodynamic assess- intra-abdominal infection. Antimicrob Agents Chemother 2016; 60:
ment. Diagn Microbiol Infect Dis 2007; 58:337–344; 66. 6234–6243.
76. Kuti JL, Florea NR, Nightingale CH, et al. Pharmacodynamics of meropenem 98. Motsch J, de Oliveira C, Stus V et al. RESTORE-IMI 1: a multicenter,
and imipenem against Enterobacteriaceae, Acinetobacter baumannii, and randomized, double-blind, comparator-controlled trial comparing the efficacy
Pseudomonas aeruginosa. Pharmacotherapy 2004; 24:8–15; 67. and safety of imipenem/relebactam versus colistin plus imipenem in patients
77. Crandon JL, Ariano RE, Zelenitsky SA, et al. Optimization of meropenem with imipenem-nonsusceptible bacterial infections. In: Presented in 28th
dosage in the critically ill population based on renal function. Intensive Care European Congress of Clinical Microbiology and Infectious Diseases
Med 2011; 37:632–638; 68. (ECCMID). Madrid: (2018)

10 www.co-infectiousdiseases.com Volume 32  Number 00  Month 2019

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Namrta; QCO/320601; Total nos of Pages: 11;
QCO 320601

Carbapenems sparing strategy Corcione et al.

99. Imipenem/Relebactam/Cilastatin Versus Piperacillin/Tazobactam for Treat- 106. Chotiprasitsakul D, Han JH, Cosgrove SE, et al. Comparing the outcomes of
ment of Participants With Bacterial Pneumonia (MK-7655A-014) (RE- adults with enterobacteriaceae bacteremia receiving short-course versus
STORE-IMI 2), Clinicaltrial.gov, NCT02493764 prolonged-course antibiotic therapy in a multicenter, propensity score-
100. Giacobbe DR, Mikulska M, Viscoli C. Recent advances in the pharmacolo- matched cohort. Clin Infect Dis 2018; 66:172–177.
gical management of infections due to multidrug resistant gram-negative 107. Giannella M, Pascale R, Toschi A, et al. Treatment duration for Escherichia
bacteria. Expert Rev Clin Pharmacol 2018; 11:1219–1236. coli bloodstream infection and outcomes: retrospective single-centre study.
101. Okujava R, Garcia-Alcalde F, Haldimann A, et al. 1359. Activity of merope- Clin Microbiol Infect 2018; 24:1077–1083.
nem/nacubactam combination against Gram-negative clinical isolates: ROS- 108. Lew KY, Ng TM, Tan M, et al. Safety and clinical outcomes of carbapenem de-
CO Global Surveillance 2017. Open Forum Infect Dis 2018; 5(Suppl escalation as part of an antimicrobial stewardship programme in an ESBL-
1):S416; Published 2018 Nov 26. endemic setting. J Antimicrob Chemother 2015; 70:1219–1225.
102. Seah VF, Ong RY, Lim AS, et al. Impact of a carbapenem antimicrobial 109. Garcı́a-Rodrı́guez JF, Bardán-Garcı́a B, Peña-Rodrı́guez MF, et al. Merope-
stewardship program on patient outcomes. Antimicrob Agents Chemother nem antimicrobial stewardship program: clinical, economic, and antibiotic
2017; 61:e00736-17. resistance impact. Eur J Clin Microbiol Infect Dis 2018; 38:161–170.
103. Righi E, Peri AM, Harris PN, et al. Global prevalence of carbapenem 110. Canouı̈ E, Gauzit R, Bruneau A, et al. Assessment of appropriate carbape-
resistance in neutropenic patients and association with mortality and carba- nem prescribing in stewardship programmes. J Hosp Infect 2018; 100:
penem use: systematic review and meta-analysis. J Antimicrob Chemother 277–279.
2017; 72:668–677. 111. Corcione S, Angilletta R, Raviolo S, et al. Epidemiology and risk
104. Eljaaly K, Enani MA, Al-Tawfiq JA, et al. Impact of carbapenem versus non- factors for mortality in bloodstream infection by CP-Kp, ESBL-E, Candida
carbapenem treatment on the rates of superinfection: a meta-analysis of and CDI: a single center retrospective study. Eur J Intern Med 2018;
randomized controlled trials. J Infect Chemother 2018; 24:915–920. 48:44–49.
105. Tedeschi S, Trapani F, Giannella M, et al. An antimicrobial stewardship 112. Papadimitriou-Olivgeris M, Marangos M, et al. Risk factors for KPC-produ-
program based on systematic infectious disease consultation in a rehabilita- cing Klebsiella pneumoniae enteric colonization upon ICU admission.
tion facility. Infect Control Hosp Epidemiol 2017; 38:76–82. J Antimicrob Chemother 2012; 67:2976–2981.

0951-7375 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-infectiousdiseases.com 11

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

You might also like