New Drugs MDR
New Drugs MDR
https://doi.org/10.1007/s40265-024-02102-8
REVIEW ARTICLE
Abstract
Gram-negative multidrug-resistant (MDR) bacteria, including Enterobacterales, Acinetobacter baumannii, and Pseudomonas
aeruginosa, pose a significant challenge in clinical practice. Infections caused by metallo-β-lactamase (MBL)–producing
Gram-negative organisms, in particular, require careful consideration due to their complexity and varied prevalence, given
that the microbiological diagnosis of these pathogens is intricate and compounded by challenges in assessing the efficacy
of anti-MBL antimicrobials. We discuss both established and new approaches in the treatment of MBL–producing Gram-
negative infections, focusing on 3 strategies: colistin; the recently approved combination of aztreonam with avibactam (or
with ceftazidime/avibactam); and cefiderocol. Despite its significant activity against various Gram-negative pathogens, the
efficacy of colistin is limited by resistance mechanisms, while nephrotoxicity and acute renal injury call for careful dosing
and monitoring in clinical practice. Aztreonam combined with avibactam (or with avibactam/ceftazidime if aztreonam
plus avibactam is not available) exhibits potent activity against MBL–producing Gram-negative pathogens. Cefiderocol in
monotherapy is effective against a wide range of multidrug-resistant organisms, including MBL producers, and favorable
clinical outcomes have been observed in various clinical trials and case series. After examining scientific evidence in the
management of infections caused by MBL–producing Gram-negative bacteria, we have developed a comprehensive clinical
algorithm to guide therapeutic decision making. We recommend reserving colistin as a last-resort option for MDR Gram-
negative infections. Cefiderocol and aztreonam/avibactam represent favorable options against MBL–producing pathogens.
In the case of P. aeruginosa with MBL–producing enzymes and with difficult-to-treat resistance, cefiderocol is the preferred
option. Further research is needed to optimize treatment strategies and minimize resistance.
Vol.:(0123456789)
1520 C. Hidalgo‑Tenorio et al.
Table 1 Classification of β-lactamases of major clinical relevance (Bush and Jacoby 2010 [164], Bush and Bradford 2020 [10])
Molecular Type Functional Relevant/distinc- Inhibition by Inhibition by Inhibition Examples (families or representatives)
class group tive substrates clavulanic acid avibactam by EDTA
A SBL 2a P + + − PC1
2b P, Cp + + − TEM-1, SHV-1
2be P, Cp, E, M + + − TEM-ESBL, SHV-ESBL, CTX-M
2br P − + − Inhibitor-resistant TEM, SHV-10
2c P ± + − CARB-1
2e P, Cp, E + + − CepA
2f P, Cp, E, M, Cp ± + − KPC, SME
B MBL 3a (B1) P, Cp, E, Cb − − + IMP, VIM, NMD
3a (B3) L1
3b (B2) Cb − − + CphA
C SBL 1 Cp − + − Chromosomal AmpC
Plasmid-mediated AmpC
1e Cp, E − + − GC1
D SBL 2d P ± + − OXA-1
2de P, E, M ± + − OXA-11
2df P, Cb − + − OXA-48, OXA-23
1966 Essential role of zinc on the activity of the Bacillus cereus “cepha- The enzyme was inhibited by ethylenediaminetet- [154]
losporinase” raacetic acid (EDTA)
1980 Definition of structural (molecular) classes of β-lactamases Class A (serine β-lactamases, or SBL); Class B: MBLs [155]
1985 Introduction of terms MBLs and metalloenzymes MBLs hydrolyze carbapenems (carbapenemases) [156]
1981 Class C and class D SBL defined [157, 158]
1988
1988 First functional classification of β-lactamases Classification based on substrate and inhibitor (clavu- [159]
lanic acid vs EDTA) profiles
1991 The first acquired MBL (IMP-1) identified (in Pseudomonas aer- [160]
uginosa, in Japan)
1995 Updated functional classification of β-lactamases Functional groups 1, 2 and 3 defined [161]
1999 VIM-1 discovered (in P. aeruginosa, in Italy) [162]
2008 NDM-1 discovered (in Klebsiella pneumoniae and Escherichia coli, [163]
in a traveler from India to Sweden)
2010 Updated functional classification of β-lactamases Additional β-lactamase subgroups defined [164]
Phenotypic tests for MBL detection are based on the (usually) meropenem is incubated into a bacterial suspen-
hydrolytic activity of these enzymes against carbapenems sion; the disk is then placed on the surface of a plate,
(and other β-lactams). For MBL producers, minimum which has been previously streaked with a carbapenem-
inhibitory concentrations (MICs) of carbapenems will be susceptible E. coli marker strain; after incubation, a reduc-
reduced in the presence of an efficient MBL inhibitor (i.e., tion in the size of the inhibition zone indicates that the
Ethylenediaminetetraacetic acid [EDTA], dipicolinic acid, investigated bacteria produce a carbapenemase. When con-
sodium mercaptoacetate, 1,10-O-phenanthroline), but this sidering P. aeruginosa and Acinetobacter baumannii, both
approach is not commonly used in the clinical laboratory. sensitivity and specificity improve when bacterial cells are
In the carbapenem inactivation method (CIM), a disk of extracted with 0.5 M TrisHCl (CIMTris). Another variant,
1522 C. Hidalgo‑Tenorio et al.
EDTA-mCIM (eCIM), can differentiate MBLs and SBL In vitro activity of cefiderocol can be alternatively determined
among carbapenemase producers [16–19]. by disk diffusion; EUCAST has defined areas of technical
The Carba NP assay is a rapid (less than 3 h) turna- uncertainty for this assay (ATU, available at EUCAST [36])
round test. It detects acidification of a solution containing for both Enterobacterales and P. aeruginosa, and when they
a carbapenem after the compound has been degraded by a occur, EUCAST has suggested several possible solutions
carbapenemase [20, 21]. (available at EUCAST [41]).
Peaks of hydrolyzed carbapenems can be detected by Results from susceptibility testing assays for MBL–pro-
matrix-assisted laser desorption/ionization time-of-flight ducing organisms should be presented as clinical catego-
(MALDI-Tof) [22, 23] (and by liquid chromatography- ries alone or with detailed MIC values (of great value
coupled tandem mass spectrometry (LC-MS/MS) [24]. from a clinical perspective when pharmacokinetics/phar-
Metallo-β-lactamases and other common carbapen- macodynamics [PK/PD] issues are considered in concrete
emases can be detected by multiplex lateral flow immu- patients), as defined by committees such as EUCAST or
noassays, which are technically simple, provide results Clinical and Laboratory Standards Institute (CLSI). Break-
within 15 min, and have good sensitivity and specificity. points for colistin, ATM/AVI and cefiderocol are presented
Unfortunately, these tests show negative results for some in Tables 3 (MIC values) and 4 (inhibition zones).
carbapenemase alleles [25].
Molecular tests are faster than methods based on bac-
terial growth. They can detect one or more genes coding 4 Epidemiology
for MBLs but require the genes to be previously known.
There are multiple commercial molecular assays based on The global prevalence and geographic distribution of
PCR, loop-mediated isothermal amplification (LAMP) MBLs are not comprehensively known due to scant data
and DNA microarrays that detect several MBLs with high and limited updated information, particularly in certain
sensitivity and specificity and can be applied directly to regions such as Africa.
clinical samples or positive blood culture bottles [26–29]. Verona integron-encoded MBL enzymes have been
More recently, whole-genome sequencing (WGS) has been reported worldwide from multiple species, particularly P.
increasingly used for the detection of resistance-related aeruginosa and Enterobacterales (mainly K. pneumoniae,
genes, including those coding for MBLs [30–33]. E coli and E. cloacae complex). These enzymes are widely
In addition, various testing methods are available for drugs distributed in Europe, and several outbreaks have been
targeting microorganisms producing MBLs, including colistin, reported in Greece, Italy, Spain, and other countries [42].
aztreonam plus avibactam (ATM/AVI) (or ATM plus ceftazi- New Delhi MBL variants are widespread in the Indian
dime [CAZ]/AVI) and cefiderocol. subcontinent. India, Pakistan and Bangladesh are consid-
For colistin, the reference method is broth microdilution. ered reservoirs for these enzymes, as is the Balkan region
Agar dilution, disk diffusion or gradient test, are not recom- in Europe. New Delhi MBL enzymes are the second most
mended, and semi-automated methods can produce false sus- frequent enzymes (after OXA-48) in the Middle East
ceptibility results. Aztreonam plus avibactam may be tested by [43, 44] and predominate in some regions of Russia [45].
standard methods in clinical laboratories. For ATM combined Outbreaks with NDM–producing organisms have been
with CAZ/AVI, in vitro synergy tests are not standardized and reported in several European countries including Italy,
the results from different methods for synergism may offer Greece, Romania, Poland, and Denmark [46]. An increas-
variable results [34–39]. ing number of NDM-1- and NDM-1/OXA-48–producing
For testing cefiderocol, MICs of cefiderocol in cation- Klebsiella pneumoniae were detected in Germany in 2022,
adjusted Mueller-Hinton broth are not reproducible and do associated with patients with a documented history of pre-
not represent the actual in vivo activity of this compound, vious presence in Ukraine [47].
which is highly influenced by iron content. Minimum inhibi- The IMPs are currently endemic in Japan and are more
tory concentrations of cefiderocol should be determined in an common in Asian countries [48, 49], with IMP-4 being
iron-depleted medium (≤ 0.03 mg/L of iron), which is not particularly important in China and Australia [50]. A large
usually available in routine clinical laboratories and is not cur- number of acquired minor MBLs limited to specific geo-
rently used by commercial panels of semi-automated methods graphical areas are also known; for a review, see reference
[40]. Several commercial tests based on broth microdilution [10].
are available as an easy approach to determining cefiderocol The EuSCAPE project analyzed carbapenemase pro-
MICs in clinical laboratories; the European Committee on duction in Enterobacteriaceae isolated from 38 European
Antimicrobial Susceptibility Testing (EUCAST) evaluation countries in 2015. The most common enzymes were KPC
of these assays is ongoing, but results are not yet available. and OXA-48 enzymes, but VIM was most common in
Metallo-β-Lactamase Treatment 1523
ATUarea of technical uncertainty, CLSI Clinical and Laboratory Standards Institute, EUCAST European Committee for Antimicrobial Susceptibility Testing, IE insufficient evidence, MIC mini-
CLSI
≥16
≥16
≥16
≥16
were rare in most countries [51].
Among Enterobacterales non-susceptible to meropenem
evaluated in the ATLAS global surveillance program iso-
EUCAST
Resistant
lated in 2018–2019, MBLs were more frequent (36.7%
of isolates) than KPCs (25.5%) or OXA-48-like (24.1%)
>2
>2
IE
IE
enzymes, and were particularly more frequent in Africa,
Middle East and the Asia/Pacific (APAC) region. Kleb-
Intermediate
CLSI breakpoints for Enterobacterales do not apply to Salmonella and Shigella spp.
≤2
≤2
≤2
Table 4 Inhibition zone (mm) breakpoints for colistin, aztreonam-avibactam (30–20 µg disc) and cefiderocol (30 µg disc) against different
groups of microorganisms
Aztreonam/avibactam Cefiderocol
Susceptible ATU Resistant Susceptible ATU Intermediate Resistant
EUCAST EUCAST EUCAST CLSI EUCAST CLSI EUCAST CLSI
significantly increased in extensively drug-resistant or mero- mutations in lipid A biosynthesis genes lpxA, lpxC, or lpxD,
penem-resistant isolates [59]. leading to complete loss of LPS production [64]. Several
mobile colistin resistance (mcr) genes carried by transfer-
able plasmids have been reported, after they were discovered
5 Drugs for Treating Infections Caused (2016) in an E. coli strain isolated in China [65].
by MBL–Producing Organisms An extensive global surveillance program from 39 coun-
tries has noted an association between the presence of a
From a clinical perspective, it is standard practice for clini- carbapenemase and increased resistance to colistin among
cians to directly receive microbiological laboratory results surveillance isolates of Enterobacteriaceae. Colistin suscep-
in a report. These reports are interpreted by microbiologists tibility was established at 98.4% overall, while it decreased
based on in vitro findings. to 88% among carbapenemase producers. Colistin suscep-
Furthermore, the selection of treatment must carefully tibility was 92.6% among MBL-positive isolates [66]. In a
consider various additional factors. These include the sever- large surveillance study (SIDERO-CR), which included 457
ity of the infection, the patient’s comorbidities, the antibi- isolated carbapenem-resistant Enterobacteriaceae, colistin
otic penetration into the site of infection, and any potential susceptibility was 93.5% for VIM producers and 78.4% for
adverse effects. Each of these factors is critical in deter- NDM producers [67]. In an observational prospective study
mining the appropriateness and efficacy of the treatment performed in two Italian hospitals, which focused on MBL,
regimen. NMD-producing strains had a colistin susceptibility rate of
90.2%, while VIM-producing strains had a rate of 80% [68].
5.1 Colistin Although colistin and polymyxin B were initially aban-
doned due to toxicity, they regained relevance given the lack
Colistin (polymyxin E) shows significant activity against of effective antimicrobials against MDR Gram-negative
most Enterobacterales, A. baumannii, P. aeruginosa and pathogens [69, 70].
S. maltophilia. However, some Gram-negative species are There are other issues that clearly limit their use in clini-
naturally resistant to colistin, including Proteus spp., Provi- cal practice. First, PK/PD is critical for optimizing dosing.
dencia spp., Morganella morganii, or S. marcescens. Colistin administration as an inactive prodrug affects the
The most common colistin resistance mechanism is the time needed to achieve desirable plasma concentrations
modification of lipopolysaccharides (LPS) via the addition [71]. A target plasma colistin concentration of 2 mg/L seems
of cationic molecules, such as L-aminoarabinose and phos- appropriate, but the risk of nephrotoxicity increases above
phoethanolamine [60]. This is frequently linked to muta- 2.5 mg/L. Achieving target concentrations in plasma may not
tions in 2-component regulatory systems [61, 62]. Mutations be sufficient for adequately treating lower respiratory tract
in mgrB, coding for a negative PhoPQ regulator, are fre- infections due to reduced lung penetration [71–74].
quently associated with colistin resistance in K. pneumoniae Preclinical lung infection models suggest poor in vivo
[63]. Colistin resistance in A. baumannii may emerge by response to the polymyxins when administered parenterally
Metallo-β-Lactamase Treatment 1525
[74]. The PK and concentration of colistin in bronchoalveo- Enterobacterales tested were inhibited at a concentration
lar lavage (BAL) were evaluated in 13 adult patients who threshold of 8 mg/L [84].
developed ventilator-associated pneumonia (VAP) caused Resistance to ATM/AVI in MBL producing Enterobac-
by A. baumannii and P. aeruginosa during their ICU stay. terales has been reported to be lower than 1% globally [85].
The administration of 2 million International Units (IU) of However, resistance to ATM/AVI in 15% NDM producing E.
colistin every 8 h results in apparently suboptimal plasma coli has been specifically linked to the production of CMY-
concentrations of colistin, which was undetectable in BAL 42 and/or PBP3 modifications [86]. Production of double
[75]. Epithelial lining fluid penetration ratios appear reduced carbapenemases may also compromise the activity of ATM/
in critically ill patients; nebulized administration of antibiot- AVI in Enterobacterales [87]. The picture is different for P.
ics has been advocated to achieve target concentrations at the aeruginosa. First, the co-production of MBLs and ESBL
infection site with minimal systemic toxicity [76]. The data or other carbapenemases is infrequent in this species. Sec-
indicate that higher doses of nebulized colistin may achieve ond, while the overexpression of AmpC may compromise
adequate concentrations in lung compartments; however, the activity of ATM to some extent, the main chromosomal
in order to optimize drug delivery both the nebulizers and resistance mechanism to ATM is related to the expression
nebulization technique are also important [77]. In a recent of the efflux pump MexAB-OprM [88]. Thus, the added
metanalysis, nebulized colistin was associated with better value of AVI in P. aeruginosa is lower than for Enterobac-
microbiological outcomes but did not result in any remark- terales, and synergy between ATM and AVI cannot be taken
able changes in the prognosis of patients with VAP [78]. for granted. Likewise, the activity of ATM/AVI activity is
Second, acute kidney injury (AKI) is a concern. This much lower in MBL producing P. aeruginosa as compared
typically occurs 5–7 days after exposure, often as a result with MBL producing Enterobacterales. For example, in one
of acute tubular necrosis [79]. Lodise et al found an AKI study, the susceptibility rate of ATM against MBL–produc-
incidence of 25.1% (RIFLE criteria). Acute kidney injury ing P. aeruginosa strains (53.8%) was only slightly increased
was associated with a higher incidence of mortality and with the addition of AVI (69.2%). On the other hand, ATM,
increased costs [80]. alone or combined with AVI, shows no significant activity
Clinical experience for colistin efficacy against infections against A. baumannii [89].
caused by MBLs is limited. A multicenter study analyzed In vitro study data found that the addition of AVI could
102 cases of bacteremia, 82 of which were caused by NDM- reduce or close the mutant selection window (MSW) of
producing K. pneumoniae or E. coli, and 20 by MIV-produc- ATM in Enterobacterales harboring MBLs. For ATM/AVI
ing strains (70% of which were K. pneumoniae). The over- dosing regimens evaluated in clinical trials, the free drug
all mortality rate in the group treated with the combination concentration was above the mutant prevention concentra-
ATM plus CAZ/AVI was 19%, while the mortality rate in tion values of >90% and >80%, whereas the fraction of time
the group treated with colistin was 59.3% [68]. within the 24 h that the free drug concentration was within
In fact, in a recent report on 343 patients with MBL–pro- the mutant selection window measures were <10% and
ducing Enterobacterales infections, the 30-day mortality rate <20% in plasma and epithelial lining fluid, respectively [90].
was 29.7%. Sensitivity analysis showed that ATM plus CAZ/ An open-label, multicenter Phase 2 trial of ATM/AVI in
AVI, compared with colistin, was independently associated complicated intra-abdominal infections (cIAI) compared 3
with a reduced 30-day mortality rate [81]. dosing regimens for PK/PD target achievement. The regimen
that prevailed, with no safety concerns, was the 500-mg/167-
5.2 Aztreonam/Avibactam (ATM/AVI) mg loading dose followed by 1500-mg/500-mg four times
daily in a 3-h infusion [91].
Aztreonam/avibactam, recently approved in April 2024 by The results of the ASSEMBLE (NCT03580044) study
European Medicines Agency (EMA), combines the inher- showed that 5/12 (41.7%) patients with confirmed MBL–pro-
ent stability of ATM against MBLs with classes A, C and ducing Enterobacterales infections, who received ATM/AVI
some D (OXA-48-like) β-lactamase inhibition offered by ± metronidazole (MTZ), were cured at test of cure (TOC)
AVI. Many carbapenem-resistant Enterobacterales (CRE) versus 0/3 (0%) of those on best available therapy [92].
strains co-produce MBL and serine enzymes that can inac- Aztreonam plus avibactam is indicated for the treatment
tivate ATM. In fact, β-lactamase inhibitors, such as AVI, of adult patients for cIAI, hospital-acquired pneumonia,
zidebactam or nacubactam, protect ATM from hydrolysis including ventilator-associated pneumonia, and complicated
and enhance its anti-MBL activity against Enterobacterales urinary tract infection (cUTI), including pyelonephritis.
and, to a lesser extent, P. aeruginosa [82, 83]. Furthermore, Aztreonam plus avibactam is also indicated for the treat-
a large in vitro broth microdilution study recently con- ment of infections due to aerobic Gram-negative organisms
firmed the potent activity of ATM/AVI against Enterobac- in adult patients with limited treatment options [93]. This
terales isolates. In particular, all carbapenemase-producing resolution also highlights the microbiology data that indicate
1526 C. Hidalgo‑Tenorio et al.
that ATM in combination with AVI may also be of particular reported a nosocomial outbreak by cefiderocol-resistant
utility in infections caused by MBL–producing Enterobac- NDM-1–producing Klebsiella pneumoniae occurring in a
terales and that this combination could therefore address large tertiary care hospital in Florence. Retrospective analy-
an unmet medical need. The most common side effects are sis of cases observed in January 2021–June 2022 revealed
serum transaminase elevation, and diarrhea [91]. Dose selec- that 21/52 were cefiderocol resistant due to the inactivation
tion for ATM/AVI, including adjustments for renal impair- of the cirA siderophore receptor gene. Cefiderocol resist-
ment based on a series of iterative population PK modeling ance in K. pneumoniae has also been associated with the
and probability of target attainment (PTA) analyses, has production of NDM-5 [107]. In another recent report by
recently suggested optimized dosing regimens [94]. Given Lasarte-Rubio et al. [108], cefiderocol resistance in an epi-
the microbiologically active molecules now approved, the demic E. cloacae complex strain was associated with the
use of this combination would be reasonable and could production of the MBL VIM-1, the ESBL SHV-12 and the
replace ATM plus CAZ/AVI. inactivation of a FcuA-like siderophore receptor. Likewise,
high-level cefiderocol resistance in a ST167 E. coli strain
5.3 Aztreonam plus Ceftazidime/Avibactam was associated with the production of NDM-35, showing an
approximate 10-fold increase in hydrolytic activity toward
Aztreonam plus CAZ/AVI has been used as an alternative cefiderocol compared to NDM-1 [109]. Additionally, the
to ATM/AVI in the past due to lack of other therapeutic isolate co-produced a CMY-type β-lactamase, exhibited a
options. In the absence of ATM/AVI in clinical practice, four amino-acid insertion in PBP3, and possessed a trun-
the combination of CAZ/AVI plus ATM can be considered. cated CirA. Moreover, the acquisition of a transferable
However, it is essential that the microbiology laboratory extrachromosomal fec operon has been associated with a
verifies the synergy between AVI (from the CAZ/AVI com- cefiderocol MIC increase in Enterobacterales [110]. The
bination) and ATM, since such synergy does not always number of reports of cefiderocol resistance in P. aeruginosa
occur, and in its absence, the use of this bi-therapy would is lower than for Enterobacterales. Iron transport systems
not be feasible. Aztreonam plus CAZ/AVI has been used as are also typically involved, but in addition to the expression
an alternative to ATM/AVI due to limited options, although of ESBLs or carbapenemases, a major source of resistance
clinical experience is mainly from case reports [95–98]. Fal- development, are the mutations within the catalytic center
cone et al. [99] noted better outcomes with ATM plus CAZ/ of AmpC [111, 112]. Recent in vitro evolution experiments
AVI for NDM- or VIM-producing Enterobacterales bacte- in P. aeruginosa, including XDR high-risk clones, revealed
remia compared to colistin. A retrospective study showed that the most frequent mutations associated with cefidero-
clinical cure rates of 91.6%, 66.7%, and 85.7% for ATM col resistance were those on piuC, fptA and pirR, related to
plus CAZ/AVI alone, with polymyxin, and with fosfomycin, iron uptake [113]. Additionally, an L320P AmpC mutation
respectively, in NDM and OXA-48 infections [100]. Timsit was selected in multiple lineages, and cloning confirmed its
et al reported clinical cure in 5 of 7 NDM-producing Entero- major impact on cefiderocol (but not ceftolazane/tazobactam
bacterales cases with this combination [101]. or CAZ/AVI) resistance [113]. Mutations in CpxS and PBP3
A systematic review by Mauri et al found a 19% 30-day were also documented. Cefiderocol resistance in A. bauman-
mortality rate for ATM plus CAZ/AVI, lower than the 44 nii has been shown to be multifactorial and dependent on
% in the control group (odds ratio 0.33; 95% CI 0.13–0.66) the interplay of the expression of β-lactamases, siderophore
[82] although effectiveness was low against MBL–producing receptors and PBP3 mutations [114].
P. aeruginosa. A meta-analysis showed ATM plus CAZ/AVI Hypotheses suggest that undetected hetero-resistance of
had a lower 30-day mortality risk compared to polymyxins Acinetobacter to cefiderocol – defined as the presence of
(risk ratio 0.51; 95% CI, 0.34–0.76) [102]. resistant subpopulations within a majority population sus-
ceptible to this agent – might explain the higher all-cause
5.4 Cefiderocol mortality rates observed in the CREDIBLE-CR study among
patients with infections caused by this organism [115, 116].
Cefiderocol is a novel siderophore cephalosporin with activ- However, a subsequent study focused exclusively on car-
ity against a large range of multi-resistant organisms, includ- bapenem-resistant isolates of the Acinetobacter calcoaceti-
ing MBL producers. cus-baumannii complex, which were cultured from patients
Resistance to cefiderocol has been documented in several treated with cefiderocol in the CREDIBLE-CR study. This
bacterial species and is generally mediated by the interplay study employed the methodology Population Analysis Pro-
of the expression of β-lactamases with potent cephalospori- filing (PAP) and surprisingly, for infections caused by PAP-
nase activity, such as several extended spectrum beta lacta- hetero-resistant isolates, the clinical cure rate was higher and
mases (ESBLs) or carbapenemases, with mutations in iron mortality lower than for infections due to PAP-susceptible
uptake systems [103–105]. Coppi et al. [106] have recently or PAP-resistant isolates [117]. The underlying causes of
Metallo-β-Lactamase Treatment 1527
cefiderocol hetero-resistant phenotypes remain poorly under- differences were likely due to deaths occurring within the
stood. In other studies involving K. pneumoniae, hetero- first 72 h and after 29 days, as well as the fact that patients
resistance has been linked to mutations in cirA (which codes in the cefiderocol arm were more severely ill, with higher
for a siderophore receptor) [118] and to blaSHV-12 ampli- rates of ICU admission, septic shock, and mechanical ven-
fication [119]. Cefiderocol hetero-resistance has also been tilation. All these factors are known independent predictors
hypothesized to be related to bacterial recurrence in a patient of mortality [128].
with bacteremia caused by caused NDM-5–producing K. The efficacy of combination treatment compared to cefi-
pneumoniae [120]. Importantly, the standard methodology derocol monotherapy remains unresolved. In carbapenem-
for defining hetero-resistance (population analysis profiling) resistant A. baumannii (CRAB) infections, a meta-analysis
lacks clinical validation as a predictor of patient outcomes, by Onorato et al. evaluated cefiderocol monotherapy and
both clinically and microbiologically. At present, correlating the combination therapy among seven studies. They found
hetero-resistance to cefiderocol with clinical outcomes in a significantly lower mortality rate among patients receiv-
patients treated with this cephalosporin remains challenging. ing cefiderocol in monotherapy as compared to treated
Specific tissue penetration data are available for the res- with combination regimens. However, these findings were
piratory system: in healthy volunteers after a single 2-g not confirmed in the sub-analysis including only patients
intravenous (IV) dose, the drug penetrates into epithelial with bloodstream infections nor in the analysis including
lining fluid (ELF) with geometric mean concentration ratios, patients with pneumonia [129]. A recent review also found
over 6 h, ranging from 0.0927 to 0.116 mg/L for ELF and no significant difference in terms of mortality, microbiologi-
total plasma. In patients with VAP, the geometric mean cal eradication and clinical cure between monotherapy and
ELF concentration of cefiderocol was 7.63 mg/L at the end combination therapy [130, 131].
of infusion, and 10.40 mg/L 2 h later. The ELF/unbound The most frequently reported treatment-emergent adverse
plasma concentration ratio was 0.212 (21.2 %) at the end of events were diarrhea, pyrexia, and vomiting. Liver enzyme
infusion and 0.547 after 2 h, suggesting delayed lung distri- increases or clotting abnormalities occurred more frequently
bution, with concentrations sufficient to treat Gram-negative in the cefiderocol group [125]. Treatment-emergent adverse
bacteria [121, 122]. events reported in at least 5% of patients in either treatment
Although few reports have evaluated the efficiency of group were generally balanced across treatment groups.
newly developed antibiotics in relation to the inoculum Four of 148 (3%) patients in the cefiderocol group and four
effect, this phenomenon could be responsible for clinical of 150 (3%) patients in the meropenem group developed
failure and the selection of resistant bacteria. A recent study Clostridium difficile infection or colitis [126].
investigated the impact of inoculum size on the MICs of In randomized clinical trials conducted with cefiderocol
cefiderocol and new β-lactams/β-lactamase inhibitors. When in patients with nosocomial pneumonia, bacteremia, and
the inoculum increased to 1 07 colony-forming units (CFU)/ sepsis or complicated urinary tract infections (cUTI), cefi-
mL, all molecules were affected, particularly cefiderocol derocol demonstrated numerically higher rates of clinical
and imipenem-relebactam. In contrast, CAZ/AVI remained cure and microbiological eradication compared to compara-
only mildly affected [123]. Another recent paper demon- tors against MBL–producing pathogens, including entero-
strated that NDM producers exhibited lower susceptibil- bacteria and non-fermenters [132]. The benefit in outcomes
ity to cefiderocol and ATM/AVI compared to VIM or IMP was observed across different genera or species and cefi-
producers. Inoculum effects on cefiderocol and ATM/AVI derocol MIC levels (up to 4 μg/mL), demonstrating effective
were observed in over 90% of susceptible carbapenemase- eradication of highly carbapenem-resistant bacteria. A simi-
producing Enterobacteriaceae isolates [124]. lar observation can be made for NDM-type carbapenemases,
Pivotal clinical trials [125, 126] demonstrated the efficacy as the clinical cure rate (56.2% [9/16]) for NDM-producing
of cefiderocol monotherapy against infections caused by infections was lower than for non-NDM enzymes (100%
MBL–producing Gram-negative bacteria. Overall, the rates [8/8]) [101, 127].
of clinical cure (70.8% [17/24]), microbiological eradication A recent narrative case series by a French group docu-
(58.3% [14/24]), and all-cause mortality at 28 days (12.5% mented the experience with a cefiderocol regimen in treating
[3/24]) were favorable compared to the comparator arms in a cohort of 16 critically ill patients with difficult-to-treat
the trials. These included the option of prescriber-judged nosocomial infections caused by MDR Gram-negative bacte-
best available therapy and high-dose meropenem, with rates ria [133]. Within this cohort, four patients with five isolates
of 40.0% (4/10), 30.0% (3/10), and 50% (5/10), respectively of MBL–producing Gram-negative bacteria (all character-
[127]. ized as VIM) were highlighted. All these patients received
A higher mortality rate was observed in patients with Aci- cefiderocol treatment, primarily as monotherapy, except 1
netobacter infections, which was 50% compared to 18% in patient with A. baumannii-induced ventilator-associated
those who received the best available therapy [125]. These pneumonia in whom it was combined with tigecycline.
1528 C. Hidalgo‑Tenorio et al.
Importantly, none of these patients treated with cefiderocol shock) and epidemiological characteristics (i.e., age, immu-
for infections caused by MBL–producing Gram-negative nosuppression, recent admissions, living in a nursing home,
bacteria died during their ICU stay. In another case series, and administration of antibiotics in the last 3 months); local
among 18 evaluable patients with infections caused by cefi- epidemiology considering susceptibility rates and resistance
derocol-susceptible MBL–producing CRE (clinical cure was mechanisms of the main microorganisms isolated from the
72.2 % [13, 18], eradication at end of treatment was 77.8 % hospital center; and the type and source of the infection
[14, 18], and 28-day mortality was 22.2 % [4 of 18 patients: (high inoculum, such as pneumonia, CNS, and bacteremia;
4 of 16 in NDM-producing CRE vs 0 of 2 in VIM-producing or low inoculum, such as urinary tract infection or skin and
CRE]) [134]. soft tissue infection) [136].
6 Guideline Recommendations
7 New Approaches
Clinical practice guidelines from various scientific societies
have addressed this issue. In 2022, the European Society Several new β-lactamase inhibitors combined with different
for Clinical Microbiology and Infectious Diseases (ESC- β-lactams have recently been introduced in clinical prac-
MID) recommended the use of cefiderocol for the treat- tice. The already commented successful combination of AVI
ment of severe infections caused by MBL–producing CRE with ATM for treating MBL–producing organisms is not
or those resistant to all other antibiotics, including CAZ/ similar to the situation with other inhibitors such as vabor-
AVI and meropenem/vaborbactam (low-level evidence) bactam (combined with meropenem) or relebactam (com-
[135]. According to ESCMID, for patients with non-severe bined with imipenem), as they are not active against class B
CRE infections, it is recommended to use an antibiotic that β-lactamases, but only class A and C enzymes.
has been shown to be effective in vitro, and the choice of
antibiotic should be based on the individual case and the 7.1 Cefepime/Zidebactam (WCK 5222)
source of the infection. In case of urinary tract infections,
aminoglycosides, such as plazomicin, are recommended over Cefepime/Zidebactam (WCK 5222) is a singular combi-
tigecycline (conditional recommendation and low evidence). nation of cefepime with a bicyclo-acylhydrazide zidebac-
Tigecycline is not recommended for bacteremia or nosoco- tam that acts as a β-lactam enhancer mediating multiple
mial or ventilator-associated pneumonia. Finally, guidelines penicillin-binding proteins (PBP) binding. Zidebactam not
suggested that if tigecycline was deemed necessary for treat- only protects cefepime from hydrolysis by certain serine
ing pneumonia, it should be administered at high doses (low β-lactamases but also has stand-alone antibacterial activity
certainty of evidence) [135]. due to its potent PBP2 binding in all Gram-negative bacte-
According to IDSA guidelines, the preferred antibiotics ria (Enterobacterales and P. aeruginosa) [137, 138]. Several
for infections outside the urinary tract caused by NDM-pro- studies have demonstrated the potent activity of cefepime/
ducing CRE are CAZ/AVI in combination with ATM, or zidebactam against a range of carbapenem-resistant patho-
cefiderocol as monotherapy. In cases of P. aeruginosa-diffi- gens, including VIM/NDM-expressing P. aeruginosa iso-
cult to treat resistance (PA-DTR) isolates producing MBL, lates, which co-amplify efflux and impermeability [139,
the recommended treatment is cefiderocol monotherapy. 140].
Cefiderocol should be reserved for CRAB infections unre-
sponsive to other antibiotics or when other agents cannot be 7.2 Taniborbactam
used due to intolerance or resistance. When used for CRAB
infections, cefiderocol should be included in a combination Taniborbactam (formerly VNRX-5133) is a cyclic boronate
regimen (https://www.idsociety.org/practiceguideline/amr- with a broad spectrum of activity, including KPC, OXA-48
guidance/) [131]. and MBLs (VIM and most NDM, but not IMP) and is the
Given this situation, hospital antimicrobial stewardship first inhibitor against all classes of β-lactamases. It uses a
programs play a fundamental role in the recommendation different mechanism to inhibit both SBLs and MBLs. For
and appropriateness of empirical or targeted antibiotic MBLs, it acts as a reversible competitive inhibitor with low
treatment with cefiderocol in patients with suspected or inhibition constants (Ki) and rapid dissociation. Tanibor-
confirmed infection with MBL–producing Gram-negative bactam is being developed in combination with cefepime or
bacilli. Recommendations take into account the following meropenem to treat severe infections caused by MDR patho-
premises: patients’ clinical situation (febrile, septic or in gens (CRE and carbapenem-resistant P. aeruginosa) [141].
Metallo-β-Lactamase Treatment 1529
Fig. 1 Flowchart of treatment options for infections caused by a lack of standardized synergy testing methods. c Avibactam does not
metallo-β-lactamase-producing Gram-negative bacteria. CRAB: significantly contribute to increased aztreonam activity in aztreonam-
carbapenem-resistant Acinetobacter baumannii; CRE: carbapenem- resistant P. aeruginosa and is not usually an adequate option for
resistant Enterobacterales; HD: high doses; refers to the need to use PA-DTR. The CHMP positive opinion, highlights its utility in infec-
loading doses of certain drugs and/or doses higher than those usually tions caused by MBL–producing Enterobacterales. d Combined: The
recommended by the technical sheet; MBL: metallo-β-lactamases. a combination could depend on the source of infection; recommended
Ideally the choice of treatment should be guided by appropriate sus- in hospital-acquired pneumonia (HAP) or ventilator-associated pneu-
ceptibility testing results. According to current trends of resistance to monia (VAP), bloodstream infections, cIAI and cUTI. The combined
the novel β-lactams, this is particularly relevant in the case of ATM/ treatment option for colistin includes the possibility of its inhalation
AVI for NDM-producing Gram-negative bacilli and for all MBL–pro- route through aerosol therapy in pneumonia, along with other sys-
ducing P. aeruginosa. b Aztreonam/avibactam, approved in April temic antibiotics. Combinations that increase nephrotoxicity or other
2024, is not yet available for clinical use. Ceftazidime/avibactam adverse effects should be avoided." e Eravacycline is not yet (April
plus aztreonam could be used as an alternative, although some limit- 2024) available. f Combined with aminoglycosides in cUTI. g ATM/
ing factors should be considered: (i) there is no evidence of clinical AVI is approved for the treatment of MBL–producing Enterobacte-
equivalence with aztreonam-avibactam, (ii) there is a risk of emer- rales and for the treatment of infections caused by aerobic Gram-neg-
gence of resistant mutants if dosing is not optimal, and (iii) there is ative organisms in adult patients with limited treatment options.
higher activity and reported efficacy against MBL–produc- second agent for A. baumannii (see Fig. 1). All decisions
ing P. aeruginosa [151, 152] compared to Enterobacterales must be comprehensive and taken under the guidance of the
(see Fig. 1). In short, when an infection is identified as stewardship team.
being caused by MBL–producing Gram-negative bacteria Combining antibiotics might be a useful strategy to
(e.g., NDM, VIM or IMP), which typically cause cross- improve the success rate and efficacy of these drugs and may
resistance phenomena between β-lactams and co-resistance prevent the emergence of resistance to new agents, although
to other antibiotic families, the therapeutic options currently some authors believe that it may lead to increased toler-
include cefiderocol monotherapy or AZT/AVI. Actually, in ance to antibiotics and favor the development of resistance
the absence of comparative studies between cefiderocol and [153]. Cefiderocol could also be an option as an empirical
AVI/ATM in infections caused by MBL–producing Entero- choice for early active treatment of well selected patients
bacterales, it is difficult to establish the order of priority with severe infections and resistance risk factors in areas
in the choice between the two drugs, so the decision will endemic for more than one MBL–producing Gram-negative
depend on local epidemiology, microorganism type, and organism (Enterobacterales, P. aeruginosa or A. bauman-
the MBL expressed. Considering the in vitro and clinical nii). In such a situation, rapid de-escalation to another agent
data discussed above, cefiderocol could be a first option for is crucial whenever possible, based on rapid and detailed
MBL–producing P. aeruginosa and in combination with a microbiological characterization of the causative pathogen,
Metallo-β-Lactamase Treatment 1531
and following recommendations by stewardship team. from Shionogi, Gilead Sciences, Janssen, Merck Sharp & Dohme, and
Finally, therapeutic alternatives to antibiotics are emerging, ViiV Healthcare. LM-M has been a consultant for MSD, Shionogi and
Advanz, has served as a speaker for MSD, Pfizer and Shionogi and
such as bacteriophage therapy. Nevertheless, the clinical use has received research grants from Pfizer, MSD and Shionogi. MR-A
of phages (usually as personalized treatment) is still limited has conducted consulting work for Shionogi and Viatris. MR-A has
by their excessive specificity and by technical preparation served as a speaker for Pfizer, Gilead Sciences and Shionogi. AO has
issues. In summary, cefiderocol in monotherapy or combina- received research grants and speakers fees from Shionogi, Pfizer and
MSD. GB has been a consultant for Pfizer, Astellas, Roche, MSD,
tion, and AZT/AVI are a suitable option for MBL–producing Shionogi and Advanz, has served as a speaker for MSD, Pfizer, Roche,
Gram-negative infection. Further innovation and research Advanz and Shionogi and has received research grants from Pfizer,
with new antimicrobial molecules in monotherapy or com- Advanz and MSD. MSL has received remuneration for lectures and
bination therapy will be decisive in addressing this contro- advisory boards (Advanz Pharma, Angelini Pharma, Janssen, Menar-
ini, MSD, Pfizer, Shionogi, Viatris) and educational grants (Gilead,
versial matter. Tedec-Meiji).
iotics-10-01283/article_deploy/antibiotics-10-01283.pdf?versi (CIM), a simple and low-cost alternative for the Carba NP test
on=1634894799. https://doi.org/10.3390/antibiotics10111283 to assess phenotypic carbapenemase activity in gram-negative
5. Paño Pardo JR, Serrano Villar S, Ramos Ramos JC, Pintado rods. PLoS One. 2015;10:e0123690. Available from: https://
V. Infections caused by carbapenemase-producing enterobac- www.ncbi.nlm.nih.gov/pubmed/25798828. https://doi.org/10.
teriaceae: risk factors, clinical features and prognosis. Enferm 1371/journal.pone.0123690
Infecc Microbiol Clin. 2014;32 Suppl 4:41-8. Available from: 17. Uechi K, Tada T, Shimada K, Kuwahara-Arai K, Arakaki M,
https://www.sciencedirect.com/science/article/abs/pii/S0213 Tome T, et al. A modified carbapenem inactivation method,
005X14701739?via%3Dihub. https://doi.org/10.1016/s0213- CIMTris, for carbapenemase production in acinetobacter and
005x(14)70173-9 pseudomonas species. J Clin Microbiol. 2017;55:3405-10.
6. Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Machuca I, Pascual Available from: https://www.ncbi.nlm.nih.gov/pubmed/28954
A. Treatment of infections caused by extended-spectrum-beta- 898. https://doi.org/10.1128/JCM.00893-17
lactamase-, AmpC-, and carbapenemase-producing enterobac- 18. Sfeir MM, Hayden JA, Fauntleroy KA, Mazur C, Johnson
teriaceae. Clin Microbiol Rev. 2018;31. Available from: https:// JK, Simner PJ, et al. EDTA-modified carbapenem inactiva-
doi.org/10.1128/cmr.00079-17?download=true. https://doi.org/ tion method: a phenotypic method for detecting metallo-beta-
10.1128/cmr.00079-17 lactamase-producing enterobacteriaceae. J Clin Microbiol.
7. Karaiskos I, Galani I, Papoutsaki V, Galani L, Giamarellou H. 2019;57:e01757-18. Available from: https://www.ncbi.nlm.nih.
Carbapenemase producing Klebsiella pneumoniae: implication gov/pubmed/30867235. https://doi.org/10.1128/JCM.01757-18
on future therapeutic strategies. Expert Rev Anti Infect Ther. 19. Lasko MJ, Gill CM, Asempa TE, Nicolau DP. EDTA-modified
2022;20:53-69. Available from: https://www.ncbi.nlm.nih.gov/ carbapenem inactivation method (eCIM) for detecting IMP
pubmed/34033499. https://doi.org/10.1080/14787210.2021. Metallo-beta-lactamase-producing Pseudomonas aeruginosa: an
1935237 assessment of increasing EDTA concentrations. BMC Microbiol.
8. Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, 2020;20:220. Available from: https://w ww.n cbi.n lm.n ih.g ov/p ub-
Monnet DL, et al. Discovery, research, and development of new med/32690021. https://doi.org/10.1186/s12866-020-01902-8
antibiotics: the WHO priority list of antibiotic-resistant bacteria 20. Nordmann P, Poirel L, Dortet L. Rapid detection of carbap-
and tuberculosis. Lancet Infect Dis. 2018;18:318–27. https://doi. enemase-producing Enterobacteriaceae. Emerg Infect Dis.
org/10.1016/s1473-3099(17)30753-3. 2012;18:1503–7. https://doi.org/10.3201/eid1809.120355.
9. WHO Bacterial Priority Pathogens List, 2024: bacterial patho- 21. Dortet L, Poirel L, Nordmann P. Rapid detection of carbap-
gens of public health. importance to guide research, development enemase-producing Pseudomonas spp. J Clin Microbiol.
and strategies to prevent and control antimicrobial resistance. 2012;50:3773–6. https://doi.org/10.1128/jcm.01597-12.
Geneva: World Health Organization; 2024. Licence: CC BY-NC- 22. Burckhardt I, Zimmermann S. Using matrix-assisted laser des-
SA 3.0 IGO Available at: https://iris.who.int/bitstream/handle/ orption ionization-time of flight mass spectrometry to detect
10665/376776/9789240093461-eng.pdf?sequence=1 carbapenem resistance within 1 to 2.5 hours. J Clin Microbiol.
10. Bush K, Bradford PA. Epidemiology of beta-lactamase-produc- 2011;49:3321-4. https://doi.org/10.1128/jcm.00287-11
ing pathogens. Clin Microbiol Rev. 2020;33. Available from: 23. Hrabák J, Chudácková E, Walková R. Matrix-assisted laser des-
https://www.ncbi.nlm.nih.gov/pubmed/32102899; https://doi. orption ionization-time of flight (maldi-tof) mass spectrometry
org/10.1128/cmr.00047-19?download=true. https://doi.org/10. for detection of antibiotic resistance mechanisms: from research
1128/CMR.00047-19 to routine diagnosis. Clin Microbiol Rev. 2013;26:103–14.
11. Galleni M, Lamotte-Brasseur J, Rossolini GM, Spencer J, Dide- https://doi.org/10.1128/cmr.00058-12.
berg O, Frere JM, et al. Standard numbering scheme for class B 24. Li G, Ye Z, Zhang W, Chen N, Ye Y, Wang Y, et al. Rapid
beta-lactamases. Antimicrob Agents Chemother. 2001;45:660- LC-MS/MS detection of different carbapenemase types in
3. Available from: https://www.ncbi.nlm.nih.gov/pubmed/11181 carbapenemase-producing Enterobacterales. Eur J Clin Micro-
339. https://doi.org/10.1128/AAC.45.3.660-663.2001 biol Infect Dis. 2022;41:815-25. Available from: https://www.
12. Mojica MF, Bonomo RA, Fast W. B1-Metallo-beta-lactamases: ncbi.nlm.nih.gov/pubmed/35396654. https://doi.org/10.1007/
where do we stand? Curr Drug Targets. 2016;17:1029-50. Avail- s10096-022-04440-5
able from: https://www.ncbi.nlm.nih.gov/pubmed/26424398. 25. Boutal H, Vogel A, Bernabeu S, Devilliers K, Creton E, Cotel-
https://doi.org/10.2174/1389450116666151001105622 lon G, et al. A multiplex lateral flow immunoassay for the rapid
13. Boyd SE, Livermore DM, Hooper DC, Hope WW. Metallo-beta- identification of NDM-, KPC-, IMP- and VIM-type and OXA-
lactamases: structure, function, epidemiology, treatment options, 48-like carbapenemase-producing Enterobacteriaceae. J Antimi-
and the development pipeline. Antimicrob Agents Chemother. crob Chemother. 2018;73:909-15. Available from: https://www.
2020;64. Available from: https://w ww.n cbi.n lm.n ih.g ov/p ubmed/ ncbi.n lm.n ih.g ov/p ubmed/2 93650 94. https://d oi.o rg/1 0.1 093/j ac/
32690645; https://www.ncbi.nlm.nih.gov/pmc/articles/PMC75 dkx521
08574/pdf/AAC.00397-20.pdf. https://doi.org/10.1128/AAC. 26. Bisiklis A, Papageorgiou F, Frantzidou F, Alexiou-Daniel S.
00397-20 Specific detection of blaVIM and blaIMP metallo-beta-lacta-
14. Zakhour J, El Ayoubi LW, Kanj SS. Metallo-beta-lactamases: mase genes in a single real-time PCR. Clin Microbiol Infect.
mechanisms, treatment challenges, and future prospects. Expert 2007;13:1201-3. Available from: https://www.ncbi.nlm.nih.gov/
Rev Anti Infect Ther. 2024;22:189–201. https://doi.org/10.1080/ pubmed/17956573. https://doi.org/10.1111/j.1469-0691.2007.
14787210.2024.2311213. 01832.x
15. Ract P, Compain F, Robin F, Decre D, Gallah S, Podglajen I. 27. Cheng C, Zheng F, Rui Y. Rapid detection of bla NDM, bla
Synergistic in vitro activity between ATM and amoxicillin- KPC, bla IMP, and bla VIM carbapenemase genes in bacteria
clavulanate against enterobacteriaceae-producing class B and/ by loop-mediated isothermal amplification. Microb Drug Resist.
or class D carbapenemases with or without extended-spectrum 2014;20:533–8.
beta-lactamases. J Med Microbiol. 2019;68:1292-8. Available 28. Moore NM, Canton R, Carretto E, Peterson LR, Sautter RL, Trac-
from: https://www.ncbi.nlm.nih.gov/pubmed/31361213. https:// zewski MM, et al. Rapid Identification of Five Classes of Car-
doi.org/10.1099/jmm.0.001052 bapenem Resistance Genes Directly from Rectal Swabs by Use
16. van der Zwaluw K, de Haan A, Pluister GN, Bootsma HJ, de of the Xpert Carba-R Assay. J Clin Microbiol. 2017;55:2268-75.
Neeling AJ, Schouls LM. The carbapenem inactivation method
Metallo-β-Lactamase Treatment 1533
Available from: https://www.ncbi.nlm.nih.gov/pubmed/28515 41. EUCAST. Area of Technical Uncertainty (ATU) in antimicro-
213. https://doi.org/10.1128/JCM.00137-17 bial susceptibility testing (8 February 2024) Available from:
29. Girlich D, Oueslati S, Bernabeu S, Langlois I, Begasse C, Aran- https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_
gia N, et al. Evaluation of the BD MAX Check-Points CPO files/Guidance_documents/Area_of_Technical_Uncer tainty_-_
Assay for the Detection of Carbapenemase Producers Directly guidance_v4_2024.pdf 2024 [
from Rectal Swabs. J Mol Diagn. 2020;22:294-300. Available 42. Giani T, Marchese A, Coppo E, Kroumova V, Rossolini GM.
from: https://www.ncbi.nlm.nih.gov/pubmed/31751674. https:// VIM-1-producing Pseudomonas mosselii isolates in Italy, pre-
doi.org/10.1016/j.jmoldx.2019.10.004 dating known VIM-producing index strains. Antimicrob Agents
30. Ellington MJ, Ekelund O, Aarestrup FM, Canton R, Doumith Chemother. 2012;56:2216-7. Available from: https://www.ncbi.
M, Giske C, et al. The role of whole genome sequencing in nlm.nih.gov/pubmed/22290983. https://doi.org/10.1128/AAC.
antimicrobial susceptibility testing of bacteria: report from the 06005-11
EUCAST Subcommittee. Clin Microbiol Infect. 2017;23:2-22. 43. Zowawi Hosam M, Sartor Anna L, Balkhy Hanan H, Walsh Tim-
Available from: https://www.ncbi.nlm.nih.gov/pubmed/27890 othy R, Al Johani Sameera M, AlJindan Reem Y, et al. Molecular
457. https://doi.org/10.1016/j.cmi.2016.11.012 characterization of carbapenemase-producing Escherichia coli
31. Hendriksen RS, Bortolaia V, Tate H, Tyson GH, Aarestrup FM, and Klebsiella pneumoniae in the countries of the gulf coop-
McDermott PF. Using Genomics to Track Global Antimicrobial eration council: dominance of OXA-48 and NDM Producers.
Resistance. Front Public Health. 2019;7:242. Available from: Antimicrob Agents Chemother. 2014;58:3085-90. https://d oi.o rg/
https://w
ww.n cbi.n lm.n ih.g ov/p ubmed/3 15522 11. https://d oi.o rg/ 10.1128/aac.02050-13
10.3389/fpubh.2019.00242 44. Al-Agamy MH, Aljallal A, Radwan HH, Shibl AM. Charac-
32. Su M, Satola SW, Read TD. Genome-Based Prediction of Bacte- terization of carbapenemases, ESBLs, and plasmid-mediated
rial Antibiotic Resistance. J Clin Microbiol. 2019;57:e01405-18. quinolone determinants in carbapenem-insensitive Escherichia
Available from: https://www.ncbi.nlm.nih.gov/pubmed/30381 coli and Klebsiella pneumoniae in Riyadh hospitals. Journal
421. https://doi.org/10.1128/JCM.01405-18 of Infection and Public Health. 2018;11:64-8. Available from:
33. Doyle RM, O'Sullivan DM, Aller SD, Bruchmann S, Clark T, https://www.sciencedirect.com/science/article/pii/S187603411
Coello Pelegrin A, et al. Discordant bioinformatic predictions 7301028. https://doi.org/10.1016/j.jiph.2017.03.010
of antimicrobial resistance from whole-genome sequencing data 45. Barantsevich EP, Churkina IV, Barantsevich NE, Pelkonen J,
of bacterial isolates: an inter-laboratory study. Microb Genom. Schlyakhto EV, Woodford N. Emergence of Klebsiella pneu-
2020;6:e000335. Available from: https://www.ncbi.nlm.nih.gov/ moniae producing NDM-1 carbapenemase in Saint Petersburg,
pubmed/32048983. https://doi.org/10.1099/mgen.0.000335 Russia. J Antimicrob Chemother. 2013;68:1204–6. https://doi.
34. EUCAST. Rationale for the EUCAST clinical breakpoints, ver- org/10.1093/jac/dks503.
sion 1.0 2013. Available from: https://w ww.e ucast.o rg/fi
lead min/ 46. Baraniak A, Izdebski R, Fiett J, Gawryszewska I, Bojarska K,
src/media/PDFs/EUCAST_files/Rationale_documents/Fosfo Herda M, et al. NDM-producing Enterobacteriaceae in Poland,
mycin_rationale_1.0_20130203.pdf. 2012-14: inter-regional outbreak of Klebsiella pneumoniae ST11
35. EUCAST. Antimicrobial susceptibility testing of colistin - prob- and sporadic cases. J Antimicrob Chemother. 2016;71:85-91.
lems detected with several commercially available products. Available from: https://www.ncbi.nlm.nih.gov/pubmed/26386
2019 [Available from: https://www.eucast.org/fileadmin/src/ 745. https://doi.org/10.1093/jac/dkv282
media/PDFs/EUCAST_files/Warnings/Warnings_docs/Warni 47. Sandfort M, Hans JB, Fischer MA, Reichert F, Cremanns M, Eis-
ng_-_colistin_AST.pdf feld J, et al. Increase in NDM-1 and NDM-1/OXA-48-producing
36. EUCAST. The European Committee on Antimicrobial Suscepti- Klebsiella pneumoniae in Germany associated with the war in
bility Testing. Breakpoint tables for interpretation of MICs and Ukraine, 2022. Euro Surveill. 2022;27:2200926. Available from:
zone diameters.Version 14.0, 2024. http://w ww.e ucast.o rg. Avail- https://w
ww.n cbi.n lm.n ih.g ov/p ubmed/3 66954 68. https://d oi.o rg/
able from: https://www.eucast.org/fileadmin/src/media/PDFs/ 10.2807/1560-7917.ES.2022.27.50.2200926
EUCAST_files/Breakpoint_tables/v_14.0_Breakpoint_Tables. 48. Zhao WH, Hu ZQ. Epidemiology and genetics of VIM-type met-
pdf 2024 allo-beta-lactamases in Gram-negative bacilli. Future Microbiol.
37. EUCAST. The European Committee on Antimicrobial Suscep- 2011;6:317-33. Available from: https://www.ncbi.nlm.nih.gov/
tibility Testing. Aztreonam-avibactam and cefepime-enmeta- pubmed/21449842. https://doi.org/10.2217/fmb.11.13
zobactam now available. Available at: https://www.eucast.org/ 49. Matsumura Y, Peirano G, Motyl MR, Adams MD, Chen L,
eucast_news/news_singleview?tx_ttnews%5Btt_news%5D= Kreiswirth B, et al. Global molecular epidemiology of IMP-
585&cHash=b88acafdf84e6e15d8f8df308ef2a514 2024 producing enterobacteriaceae. Antimicrob Agents Chemother.
38. Wenzler E, Deraedt MF, Harrington AT, Danizger LH. Syner- 2017;61:e02729-16. Available from: https://www.ncbi.nlm.nih.
gistic activity of ceftazidime-avibactam and aztreonam against gov/pubmed/28167555. https://doi.org/10.1128/AAC.02729-16
serine and metallo-β-lactamase-producing gram-negative patho- 50. Kizny Gordon A, Phan HTT, Lipworth SI, Cheong E, Gottlieb
gens. Diagn Microbiol Infect Dis. 2017;88:352–4. https://d oi.o rg/ T, George S, et al. Genomic dynamics of species and mobile
10.1016/j.diagmicrobio.2017.05.009. genetic elements in a prolonged blaIMP-4-associated carbapen-
39. Collar GDS, Moreira NK, Becker J, Barth AL, Caierão J. Deter- emase outbreak in an Australian hospital. J Antimicrob Chem-
mination of aztreonam/ceftazidime-avibactam synergism and other. 2020;75:873-82. Available from: https://www.ncbi.nlm.
proposal of a new methodology for the evaluation of suscepti- nih.gov/pubmed/31960024. https://doi.org/10.1093/jac/dkz526
bility in vitro. Diagn Microbiol Infect Dis. 2024;109: 116236. 51. Grundmann H, Glasner C, Albiger B, Aanensen DM, Tomlinson
https://doi.org/10.1016/j.diagmicrobio.2024.116236. CT, Andrasevic AT, et al. Occurrence of carbapenemase-pro-
40. Simner PJ, Palavecino EL, Satlin MJ, Mathers AJ, Weinstein ducing Klebsiella pneumoniae and Escherichia coli in the Euro-
MP, Lewis JS, 2nd, et al. Potential of inaccurate cefiderocol sus- pean survey of carbapenemase-producing Enterobacteriaceae
ceptibility results: a CLSI AST subcommittee advisory. J Clin (EuSCAPE): a prospective, multinational study. Lancet Infect
Microbiol. 2023;61:e0160022. Available from: https://www. Dis. 2017;17:153-63. Available from: https://www.ncbi.nlm.nih.
ncbi.nlm.nih.gov/pubmed/36946754. https://doi.org/10.1128/ gov/p ubmed/2 78669 44. https://d oi.o rg/1 0.1 016/S
1473-3 099(16)
jcm.01600-22 30257-2
1534 C. Hidalgo‑Tenorio et al.
52. Estabrook M, Muyldermans A, Sahm D, Pierard D, Stone G, Utt 62. Jayol A, Nordmann P, Brink A, Poirel L. Heteroresistance to
E. Epidemiology of resistance determinants identified in merope- colistin in Klebsiella pneumoniae associated with alterations in
nem-nonsusceptible enterobacterales collected as part of a global the PhoPQ regulatory system. Antimicrob Agents Chemother.
surveillance study, 2018 to 2019. Antimicrob Agents Chemother. 2015;59:2780-4. Available from: https://www.ncbi.nlm.nih.gov/
2023;67: e0140622. https://doi.org/10.1128/aac.01406-22. pubmed/25733503; https://www.ncbi.nlm.nih.gov/pmc/articles/
53. Estabrook M, Muyldermans A, Sahm D, Pierard D, Stone G, Utt PMC4394806/pdf/zac2780.pdf. https://doi.org/10.1128/AAC.
E. Erratum for Estabrook et al., "Epidemiology of Resistance 05055-14
Determinants Identified in Meropenem-Nonsusceptible Entero- 63. Cannatelli A, Giani T, D'Andrea MM, Di Pilato V, Arena F,
bacterales Collected as Part of a Global Surveillance Study, 2018 Conte V, et al. MgrB inactivation is a common mechanism of
to 2019". Antimicrob Agents Chemother. 2024;68:e0164023. colistin resistance in KPC-producing Klebsiella pneumoniae of
https://doi.org/10.1128/aac.01640-23 clinical origin. Antimicrob Agents Chemother. 2014;58:5696-
54. Sader HS, Carvalhaes CG, Kimbrough JH, Mendes RE, Castan- 703. Available from: https://www.ncbi.nlm.nih.gov/pubmed/
heira M. Activity of aztreonam-avibactam against Enterobac- 25022583; https://www.ncbi.nlm.nih.gov/pmc/articles/PMC41
terales resistant to recently approved beta-lactamase inhibitor 87966/pdf/zac5696.pdf. https://doi.org/10.1128/AAC.03110-14
combinations collected in Europe, Latin America, and the Asia- 64. Moffatt JH, Harper M, Adler B, Nation RL, Li J, Boyce JD. Inser-
Pacific Region (2020–2022). International Journal of Antimicro- tion sequence ISAba11 is involved in colistin resistance and loss
bial Agents. 2024;63:107113. Available from: https://w ww.s cien of lipopolysaccharide in Acinetobacter baumannii. Antimicrob
cedirect.com/science/article/pii/S0924857924000311. https:// Agents Chemother. 2011;55:3022-4. Available from: https://
doi.org/10.1016/j.ijantimicag.2024.107113 www.ncbi.nlm.nih.gov/pubmed/21402838; https://www.ncbi.
55. Karlowsky JA, Lob SH, Chen WT, DeRyke CA, Siddiqui F, nlm.n ih.g ov/p mc/a rticl es/P
MC310 1452/p df/z ac302 2.p df. https://
Young K, et al. In vitro activity of imipenem/relebactam against doi.org/10.1128/AAC.01732-10
non-Morganellaceae Enterobacterales and Pseudomonas aer- 65. Liu YY, Wang Y, Walsh TR, Yi LX, Zhang R, Spencer J, et al.
uginosa in the Asia-Pacific region: SMART 2017–2020. Int J Emergence of plasmid-mediated colistin resistance mechanism
Antimicrob Agents. 2023;62: 106900. https://doi.org/10.1016/j. MCR-1 in animals and human beings in China: a microbiological
ijantimicag.2023.106900. and molecular biological study. Lancet Infect Dis. 2016;16:161-
56. Karlowsky JA, Lob SH, Siddiqui F, Pavia J, DeRyke CA, Young 8. Available from: https://www.ncbi.nlm.nih.gov/pubmed/26603
K, et al. In vitro activity of imipenem/relebactam against non- 172. https://doi.org/10.1016/S1473-3099(15)00424-7
Morganellaceae Enterobacterales and Pseudomonas aerugi- 66. Bradford PA, Kazmierczak KM, Biedenbach DJ, Wise MG,
nosa in Latin America: SMART 2018–2020. Braz J Infect Dis. Hackel M, Sahm DF. Correlation of β-Lactamase Production
2023;27: 102775. https://doi.org/10.1016/j.bjid.2023.102775. and Colistin Resistance among Enterobacteriaceae Isolates from
57. Canada-Garcia JE, Moure Z, Sola-Campoy PJ, Delgado-Val- a Global Surveillance Program. Antimicrob Agents Chemother.
verde M, Cano ME, Gijon D, et al. CARB-ES-19 Multicenter 2015;60:1385-92. Available from: https://w ww.n cbi.n lm.n ih.g ov/
Study of Carbapenemase-Producing Klebsiella pneumoniae pmc/articles/PMC4775915/pdf/zac1385.pdf. https://doi.org/10.
and Escherichia coli From All Spanish Provinces Reveals Inter- 1128/aac.01870-15
regional Spread of High-Risk Clones Such as ST307/OXA-48 67. Longshaw C, Manissero D, Tsuji M, Echols R, Yamano Y.
and ST512/KPC-3. Front Microbiol. 2022;13:918362. Available In vitro activity of the siderophore cephalosporin, cefiderocol,
from: https://www.ncbi.nlm.nih.gov/pubmed/35847090. https:// against molecularly characterized, carbapenem-non-susceptible
doi.org/10.3389/fmicb.2022.918362 Gram-negative bacteria from Europe. JAC Antimicrob Resist.
58. Gracia-Ahufinger I, Lopez-Gonzalez L, Vasallo FJ, Galar A, 2020;2:dlaa060. Available from: https://www.ncbi.nlm.nih.gov/
Siller M, Pitart C, et al. The CARBA-MAP study: national map- pubmed/34223017; https://www.ncbi.nlm.nih.gov/pmc/articles/
ping of carbapenemases in Spain (2014-2018). Front Microbiol. PMC8210120/pdf/dlaa060.pdf. https://doi.org/10.1093/jacamr/
2023;14:1247804. Available from: https://www.ncbi.nlm.nih. dlaa060
gov/pubmed/37744921. https://doi.org/10.3389/fmicb.2023. 68. Falcone M, Daikos GL, Tiseo G, Bassoulis D, Giordano C,
1247804 Galfo V, et al. Efficacy of ceftazidime-avibactam plus aztre-
59. Sastre-Femenia MA, Fernandez-Munoz A, Gomis-Font MA, Tal- onam in patients with bloodstream infections caused by
tavull B, Lopez-Causape C, Arca-Suarez J, et al. Pseudomonas metallo-β-lactamase–producing enterobacterales. Clin Infect
aeruginosa antibiotic susceptibility profiles, genomic epidemi- Dis. 2020;72:1871–8. https://doi.org/10.1093/cid/ciaa586.
ology and resistance mechanisms: a nation-wide five-year time 69. Michalopoulos A, Falagas ME. Colistin and polymyxin B in
lapse analysis. Lancet Reg Health Eur. 2023;34:100736. Avail- critical care. Crit Care Clin. 2008;24:377–91. https://doi.org/
able from: https://www.ncbi.nlm.nih.gov/pubmed/37753216. 10.1016/j.ccc.2007.12.003.
https://doi.org/10.1016/j.lanepe.2023.100736 70. Michalopoulos AS, Karatza DC. Multidrug-resistant Gram-
60. Aslan AT, Akova M. The Role of Colistin in the Era of New negative infections: the use of colistin. Expert Rev Anti-Infect
beta-Lactam/beta-Lactamase Inhibitor Combinations. Antibiot- Ther. 2010;8:1009–17. https://doi.org/10.1586/eri.10.88.
ics (Basel). 2022;11. Available from: https://www.ncbi.nlm.nih. 71. Giacobbe DR, Karaiskos I, Bassetti M. How do we optimize
gov/p ubmed/3 52038 79; https://m dpi-r es.c om/d_a ttach ment/a ntib the prescribing of intravenous polymyxins to increase their
iotics/a ntibi otics-1 1-0 0277/a rticl e_d eploy/a ntibi otics-1 1-0 0277- longevity and efficacy in critically ill patients? Expert Opin
v2.pdf?version=1645509261. https://doi.org/10.3390/antibiotic Pharmacother. 2021;23:5–8. https://doi.org/10.1080/14656566.
s11020277 2021.1961743.
61. Jayol A, Poirel L, Brink A, Villegas MV, Yilmaz M, Nordmann P. 72. Cheah S-E, Wang J, Nguyen V, Turnidge J, Li J, Nation RL.
Resistance to colistin associated with a single amino acid change New pharmacokinetic/pharmacodynamic studies of systemically
in protein PmrB among Klebsiella pneumoniae isolates of world- administered colistin against Pseudomonas aeruginosa and Aci-
wide origin. Antimicrob Agents Chemother. 2014;58:4762-6. netobacter baumannii in mouse thigh and lung infection mod-
Available from: https://www.ncbi.nlm.nih.gov/pubmed/24914 els: smaller response in lung infection. J Antimicrob Chemother.
122; https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4136042/ 2015;70(12):3291–7.
pdf/zac4762.pdf. https://doi.org/10.1128/AAC.00084-14
Metallo-β-Lactamase Treatment 1535
93. EMBLAVEO. Emblaveo 1.5 g/0.5 g powder for concentrate for iotics-11-00723/article_deploy/antibiotics-11-00723-v2.pdf?
solution for infusion. SUMMARY OF PRODUCT CHARAC- version=1654076988. https://doi.org/10.3390/antibiotics1106
TERISTICS. Technical data sheet. Available at: https://www. 0723
ema.europa.eu/en/documents/product-infor mation/emblaveo- 104. Nordmann P, Shields RK, Doi Y, Takemura M, Echols R, Matsu-
epar-product-information_en.pdf naga Y, et al. Mechanisms of Reduced Susceptibility to Cefidero-
94. Das S, Riccobene T, Carrothers TJ, Wright JG, MacPherson M, col Among Isolates from the CREDIBLE-CR and APEKS-NP
Cristinacce A, et al. Dose selection for aztreonam-avibactam, Clinical Trials. Microb Drug Resist. 2022;28:398-407. Available
including adjustments for renal impairment, for Phase IIa and from: https://www.ncbi.nlm.nih.gov/pubmed/35076335; https://
Phase III evaluation. Eur J Clin Pharmacol. 2024;80:529–43. doi.o rg/1 0.1 089/m
dr.2 021.0 180?d ownlo ad=t rue. https://d oi.o rg/
https://doi.org/10.1007/s00228-023-03609-x. 10.1089/mdr.2021.0180
95. Alghoribi MF, Alqurashi M, Okdah L, Alalwan B, AlHebai- 105. Kohira N, Hackel MA, Ishioka Y, Kuroiwa M, Sahm DF, Sato T,
shi YS, Almalki A, et al. Successful treatment of infective et al. Reduced susceptibility mechanism to cefiderocol, a sidero-
endocarditis due to pandrug-resistant Klebsiella pneumoniae phore cephalosporin, among clinical isolates from a global sur-
with ceftazidime-avibactam and aztreonam. Scientific Reports. veillance programme (SIDERO-WT-2014). J Glob Antimicrob
2021;11. Available from: https://w ww.s copus.c om/i nward/ Resist. 2020;22:738-41. Available from: https://www.ncbi.nlm.
record.uri?eid=2-s2.0-85105431162&doi=10.1038/s41598- nih.g ov/p ubmed/3 27023 96; https://w ww.s cienc edire ct.c om/s cien
021-89255-8&partnerID=40&md5=f4b419a5dabfb193b026 ce/article/pii/S2213716520301843?via%3Dihub. https://doi.org/
e06fc0282899. https://doi.org/10.1038/s41598-021-89255-8 10.1016/j.jgar.2020.07.009
96. Cairns KA, Hall V, Martin GE, Griffin DWJ, Stewart JD, Khan 106. Coppi M, Antonelli A, Niccolai C, Bartolini A, Bartolini L,
SF, et al. Treatment of invasive IMP-4 Enterobacter cloacae Grazzini M, et al. Nosocomial outbreak by NDM-1-producing
infection in transplant recipients using ceftazidime/avibactam Klebsiella pneumoniae highly resistant to cefiderocol, Florence,
with aztreonam: A case series and literature review. Transpl Italy, August 2021 to June 2022. Euro Surveill. 2022;27. Avail-
Infect Dis. 2021;23:e13510. Available from: https://www.ncbi. able from: https://www.ncbi.nlm.nih.gov/pubmed/36305334;
nlm.nih.gov/pubmed/33217119. https://doi.org/10.1111/tid. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9615416/pdf/
13510 eurosurv-27-43-1.pdf. https://doi.org/10.2807/1560-7917.ES.
97. Mularoni A, Mezzatesta ML, Pilato M, Medaglia AA, Cervo 2022.27.43.2200795
A, Bongiorno D, et al. Combination of aztreonam, ceftazi- 107. Zhou Y, Wu X, Wu C, Zhou P, Yang Y, Wang B, et al. Emergence
dime-avibactam and amikacin in the treatment of VIM-1 of KPC-2 and NDM-5-coproducing hypervirulent carbapenem-
Pseudomonas aeruginosa ST235 osteomyelitis. Int J Infect resistant Klebsiella pneumoniae with high-risk sequence types
Dis. 2021;108:510-2. Available from: https://www.ncbi.nlm. ST11 and ST15. mSphere. 2024;9: e0061223. https://doi.org/10.
nih.gov/pubmed/34091004. https://doi.org/10.1016/j.ijid.2021. 1128/msphere.00612-23.
05.085 108. Lasarte-Monterrubio C, Guijarro-Sanchez P, Vazquez-Ucha
98. Pelaez Bejarano A, Sanchez-Del Moral R, Montero-Perez O, JC, Alonso-Garcia I, Alvarez-Fraga L, Outeda M, et al. Anti-
Martinez-Marcos FJ. Successful treatment of Verona integron- microbial Activity of Cefiderocol against the Carbapenemase-
encoded metallo-beta-lactamase-producing Klebsiella pneumo- Producing Enterobacter cloacae Complex and Characterization
niae infection using the combination of ceftazidime/avibactam of Reduced Susceptibility Associated with Metallo-beta-Lacta-
and aztreonam. Eur J Hosp Pharm. 2022;29:113-5. Available mase VIM-1. Antimicrob Agents Chemother. 2023;67:e0150522.
from: https://www.ncbi.nlm.nih.gov/pubmed/34716170. https:// Available from: https://www.ncbi.nlm.nih.gov/pubmed/37195
doi.org/10.1136/ejhpharm-2021-002772 077. https://doi.org/10.1128/aac.01505-22
99. Falcone M, Daikos GL, Tiseo G, Bassoulis D, Giordano C, 109. Poirel L, Ortiz de la Rosa JM, Sakaoglu Z, Kusaksizoglu A,
Galfo V, et al. Efficacy of ceftazidime-avibactam plus aztre- Sadek M, Nordmann P. NDM-35-Producing ST167 Escherichia
onam in patients with bloodstream infections caused by coli Highly Resistant to beta-Lactams Including Cefiderocol.
metallo-β-lactamase-producing enterobacterales. Clin Infect Dis. Antimicrob Agents Chemother. 2022;66:e0031122. Available
2021;72:1871–8. https://doi.org/10.1093/cid/ciaa586. from: https://www.ncbi.nlm.nih.gov/pubmed/35867524; https://
100. Nagvekar V, Shah A, Unadkat VP, Chavan A, Kohli R, Hodgar S, www.n cbi.n lm.n ih.g ov/p mc/a rticl es/P MC938 0521/p df/a ac.
et al. Clinical outcome of patients on ceftazidime-avibactam and 00311-22.pdf. https://doi.org/10.1128/aac.00311-22
combination therapy in carbapenem-resistant enterobacteriaceae. 110. Kocer K, Boutin S, Heeg K, Nurjadi D. The acquisition of
Indian J Crit Care Med. 2021;25:780–4. https://doi.org/10.5005/ transferable extrachromosomal fec operon is associated with
jp-journals-10071-23863. a cefiderocol MIC increase in Enterobacterales. J Antimicrob
101. Timsit J-F, Wicky P-H, de Montmollin E. Treatment of Severe Chemother. 2022;77:3487-95. Available from: https://w ww.n cbi.
Infections Due to Metallo-Betalactamases Enterobacterales nlm.nih.gov/pubmed/36245258; https://academic.oup.com/jac/
in Critically Ill Patients. Antibiotics (Basel, Switzerland). article-abstract/77/12/3487/6761816?redirectedFrom=fulltext.
2022;11:144. Available from: https://pubmed.ncbi.nlm.nih.gov/ https://doi.org/10.1093/jac/dkac347
35203747; https://www.ncbi.nlm.nih.gov/pmc/articles/PMC88 111. Lopez-Causape C, Maruri-Aransolo A, Gomis-Font MA, Penev
68391/. https://doi.org/10.3390/antibiotics11020144 I, Castillo MG, Mulet X, et al. Cefiderocol resistance genomics in
102. Gupta N, Boodman C, Prayag P, Manesh A, Kumar TP. Ceftazi- sequential chronic Pseudomonas aeruginosa isolates from cystic
dime-avibactam and aztreonam combination for Carbapenem- fibrosis patients. Clin Microbiol Infect. 2023;29:538 e7- e13.
resistant Enterobacterales bloodstream infections with presumed Available from: https://www.ncbi.nlm.nih.gov/pubmed/36435
Metallo-β-lactamase production: a systematic review and meta- 424. https://doi.org/10.1016/j.cmi.2022.11.014
analysis. Expert Rev Anti Infect Ther. 2024. https://doi.org/10. 112. Lasarte-Monterrubio C, Fraile-Ribot PA, Vazquez-Ucha JC,
1080/14787210.2024.2307912. Cabot G, Guijarro-Sanchez P, Alonso-Garcia I, et al. Activity of
103. Karakonstantis S, Rousaki M, Kritsotakis EI. Cefiderocol: Sys- cefiderocol, imipenem/relebactam, cefepime/taniborbactam and
tematic Review of Mechanisms of Resistance, Heteroresistance cefepime/zidebactam against ceftolozane/tazobactam- and cef-
and In Vivo Emergence of Resistance. Antibiotics (Basel). tazidime/avibactam-resistant Pseudomonas aeruginosa. J Antimi-
2022;11. Available from: https://w ww.n cbi.n lm.n ih.g ov/p ubmed/ crob Chemother. 2022;77:2809-15. Available from: https://w ww.
35740130; https://mdpi-res.com/d_attachment/antibiotics/antib ncbi.nlm.nih.gov/pubmed/35904000; https://academic.oup.com/
Metallo-β-Lactamase Treatment 1537
Microbiol Infect. 2022;28:521-47. Available from: https://www. with limited treatment options: a narrative review. Antibiotics
ncbi.nlm.nih.gov/pubmed/34923128. https://doi.org/10.1016/j. (Basel). 2022;11:579. Available from: https://w ww.n cbi.n lm.n ih.
cmi.2021.11.025 gov/pubmed/35625223. https://doi.org/10.3390/antibiotics1105
136. Cercenado E, Rodriguez-Bano J, Alfonso JL, Calbo E, Escosa L, 0579
Fernandez-Polo A, et al. Antimicrobial stewardship in hospitals: 146. Viale P, Sandrock CE, Ramirez P, Rossolini GM, Lodise TP.
Expert recommendation guidance document for activities in spe- Treatment of critically ill patients with cefiderocol for infections
cific populations, syndromes and other aspects (PROA-2) from caused by multidrug-resistant pathogens: review of the evidence.
SEIMC, SEFH, SEMPSPGS, SEMICYUC and SEIP. Enferm Ann Intensive Care. 2023;13:52. Available from: https://www.
Infecc Microbiol Clin (Engl Ed). 2023;41:238-42. Available ncbi.nlm.nih.gov/pubmed/37322293. https://doi.org/10.1186/
from: https://www.ncbi.nlm.nih.gov/pubmed/36610836. https:// s13613-023-01146-5
doi.org/10.1016/j.eimce.2022.05.013 147. Gijon Cordero D, Castillo-Polo JA, Ruiz-Garbajosa P, Canton
137. Moya B, Barcelo IM, Bhagwat S, Patel M, Bou G, Papp-Wallace R. Antibacterial spectrum of cefiderocol. Rev Esp Quimioter.
KM, et al. WCK 5107 (Zidebactam) and WCK 5153 Are Novel 2022;35 Suppl 2:20-7. Available from: https://www.ncbi.nlm.
Inhibitors of PBP2 Showing Potent "beta-Lactam Enhancer" nih.gov/pubmed/36193981. https://doi.org/10.37201/req/s02.03.
Activity against Pseudomonas aeruginosa, Including Multidrug- 2022
Resistant Metallo-beta-Lactamase-Producing High-Risk Clones. 148. Tamma PD, Aitken SL, Bonomo RA, Mathers AJ, van Duin D,
Antimicrob Agents Chemother. 2017;61:e02529-16. Available Clancy CJ. Infectious Diseases Society of America Guidance
from: https://www.ncbi.nlm.nih.gov/pubmed/28289035. https:// on the Treatment of AmpC β-lactamase-producing enterobac-
doi.org/10.1128/AAC.02529-16 terales, carbapenem-resistant Acinetobacter baumannii, and
138. Yahav D, Giske CG, Gramatniece A, Abodakpi H, Tam VH, Stenotrophomonas maltophilia infections. Clin Infect Dis.
Leibovici L. New beta-Lactam-beta-Lactamase Inhibitor Combi- 2022;74:2089-114. Available from: https://w aterm
ark.s ilver chair.
nations. Clin Microbiol Rev. 2020;34:e00115-20. Available from: com/ciab1013.pdf?token=AQECAHi208BE49Ooan9kkhW_
https://w
ww.n cbi.n lm.n ih.g ov/p ubmed/3 31771 85. https://d oi.o rg/ Ercy7Dm3ZL_9Cf3qfKAc485ysgAAAsIwggK-BgkqhkiG9w
10.1128/CMR.00115-20 0BBwa g ggKv M IICq w IBAD C CAqQ G CSqG S Ib3D Q EHAT
139. Moya B, Bhagwat S, Cabot G, Bou G, Patel M, Oliver A. Effec- AeBgl g hkgB Z QMEA S 4wEQ Q MeHg t xubF K 6ivW s MzAg
tive inhibition of PBPs by cefepime and zidebactam in the pres- EQgII C dUu_ n 7HrVO- I JMJE Y 9Pnk N 9oAz k AVQX E Ow1n
ence of VIM-1 drives potent bactericidal activity against MBL- F6cNKTFSSQAnXBfTe7Fr9WPFLEccdwl_F22t2j3eWqcbqVz
expressing Pseudomonas aeruginosa. J Antimicrob Chemother. UbPNbaktoxR9Ni6M1AaP4JDMVyFPOe4uGB7zyQqDb6kxa
2020;75:1474-8. Available from: https://www.ncbi.nlm.nih.gov/ CGxz7YgrvBfGrCuHMhEI9QZ8W3hf3ZlBsvoXN5spe9J8f
pubmed/32083659. https://doi.org/10.1093/jac/dkaa036 66nWiYXqFiZGK6WdF202L1G9P3vXd98s2M10TGdgMti4_
140. Mullane EM, Avery LM, Nicolau DP. Comparative evaluation PkX3lbDCI58HwaX7BFjr_aYSIdyhL2GXEYuQn6aIFzKYPQ
of the in vitro activities of WCK 5222 (Cefepime-Zidebactam) eQOiwZ2mU-D8flLuft7DXG731liDp27woMFrVLbNCz0Rx
and combination antibiotic therapies against carbapenem-resist- qprmD2 fFvuQ uj3s5 8jkXM GlIw1-B IB4IL w-W
LTl5e c2_Y qmHg
ant Pseudomonas aeruginosa. Antimicrob Agents Chemother. IfaY8m 9BV39 Rf1-P JIF32 aekkW gp6cy LL8Zc NLyFz 4Rdsy Fqyj
2020;64:e01669-19. Available from: https://www.ncbi.nlm.nih. 9_-5mm4evLS07BjI72gdM9ahMjideJUey0HZDhCZKvu58Bza
gov/pubmed/31844009. https://doi.org/10.1128/AAC.01669-19 7M6NZF eTtIb 6ziSV 14WVr tw2hM-d Aiib_h_6 0jPk7 ipukb gg6W
141. Hamrick JC, Docquier JD, Uehara T, Myers CL, Six DA, gIcbp9 bIYcu Na9Kb QthPd LLG3z zBWI-v 47VPc v1QaF v6sI
Chatwin CL, et al. VNRX-5133 (Taniborbactam), a broad-spec- GpSt2 l OrX9 L 03SR b ENA5 Q dAoq 0 lWuZ z zlQC r zejTv 8f8e
trum inhibitor of serine- and metallo-beta-lactamases, restores e0TWEZeVTHoXZ8hAX4J3JCR9DYOHCNCCZOmQDAI1
activity of cefepime in enterobacterales and Pseudomonas aer- Yhov- H WKJn T Mljn 6 xpzl p pgAV k RlHJ F cahl Aw7SI 5 G23P
uginosa. Antimicrob Agents Chemother. 2020;64:e01963-19. zN23NoNll7z-vnQlykBrJqyWnRw_B_ASNPx9AvOsMEYiP
Available from: https://www.ncbi.nlm.nih.gov/pubmed/31871 mtrd4zgk_6Ew1ayOnt4R6Yr0VvPg3oPMwuXYtsNMbwy7
094. https://doi.org/10.1128/AAC.01963-19 NBHYSOxEEo8Bo2hd8YUvxUN_tlbVaLz1BJFOjRmfMkBD
142. Vazquez-Ucha JC, Arca-Suarez J, Bou G, Beceiro A. New Car- 0cPdUE w4jjD XFJsI sUNWY MQ4p. https://d oi.o rg/1 0.1 093/c id/
bapenemase inhibitors: clearing the way for the beta-lactams. Int ciab1013
J Mol Sci. 2020;21:9308. Available from: https://w ww.n cbi.n lm. 149. Vidal-Cortes P, Martin-Loeches I, Rodriguez A, Bou G, Canton
nih.gov/pubmed/33291334. https://doi.org/10.3390/ijms212393 R, Diaz E, et al. Current positioning against severe infections
08 due to klebsiella pneumoniae in hospitalized adults. Antibiotics
143. Giacobbe DR, Ciacco E, Girmenia C, Pea F, Rossolini GM, Sot- (Basel). 2022;11:1160. Available from: https://www.ncbi.nlm.
giu G, et al. Evaluating cefiderocol in the treatment of multidrug- nih.gov/pubmed/36139940. https://doi.org/10.3390/antibiotic
resistant gram-negative bacilli: a review of the emerging data. s11091160
Infect Drug Resist. 2020;13:4697-711. Available from: https:// 150. Chaïbi K, Jaureguy F, Do Rego H, Ruiz P, Mory C, El Helali N,
pubmed.ncbi.nlm.nih.gov/33402840; https://www.ncbi.nlm.nih. et al. What to do with the new antibiotics? Antibiotics (Basel,
gov/pmc/articles/PMC7778378/. https://doi.org/10.2147/IDR. Switzerland). 2023;12:654. Available from: https://p ubmed.n cbi.
S205309 nlm.nih.gov/37107016; https://www.ncbi.nlm.nih.gov/pmc/artic
144. Cortegiani A, Ingoglia G, Ippolito M, Girardis M, Falcone M, les/P
MC101 35159/. https://d oi.o rg/1 0.3 390/a ntibi otics 12040 654
Pea F, et al. Empiric treatment of patients with sepsis and septic 151. Karlowsky JA, Kazmierczak KM, de Jonge BLM, Hackel MA,
shock and place in therapy of cefiderocol: a systematic review Sahm DF, Bradford PA. In vitro activity of aztreonam-avibactam
and expert opinion statement. Journal of Anesthesia, Analgesia against enterobacteriaceae and Pseudomonas aeruginosa isolated
and Critical Care (Online). 2022;2:34-. Available from: https:// by clinical laboratories in 40 countries from 2012 to 2015. Anti-
pubmed.ncbi.nlm.nih.gov/37386663; https://www.ncbi.nlm. microb Agents Chemother. 2017;61:e00472-17. Available from:
nih.gov/pmc/articles/PMC9361889/. https://doi.org/10.1186/ https://w ww.n cbi.n lm.n ih.g ov/p ubmed/2 86301 92. https://d oi.o rg/
s44158-022-00062-7 10.1128/AAC.00472-17
145. Losito AR, Raffaelli F, Del Giacomo P, Tumbarello M. New 152. Canton R, Doi Y, Simner PJ. Treatment of carbapenem-resistant
drugs for the treatment of Pseudomonas aeruginosa infections Pseudomonas aeruginosa infections: a case for cefiderocol.
Metallo-β-Lactamase Treatment 1539
Expert Rev Anti-infect Ther. 2022;20:1077-94. Available from: from: https://www.ncbi.nlm.nih.gov/pubmed/2658781. https://
https://d oi.o rg/1 0.1 080/1 47872 10.2 022.2 07170 1. https://d oi.o rg/ doi.org/10.1128/AAC.33.3.271
10.1080/14787210.2022.2071701 160. Watanabe M, Iyobe S, Inoue M, Mitsuhashi S. Transferable
153. Liu J, Gefen O, Ronin I, Bar-Meir M, Balaban NQ. Effect of imipenem resistance in Pseudomonas aeruginosa. Antimicrob
tolerance on the evolution of antibiotic resistance under drug Agents Chemother. 1991;35:147-51. Available from: https://
combinations. Science. 2020;367:200–4. https://d oi.o rg/1 0.1 126/ www.ncbi.nlm.nih.gov/pubmed/1901695. https://doi.org/10.
science.aay3041. 1128/AAC.35.1.147
154. Sabath LD, Abraham EP. Zinc as a cofactor for cephalospori- 161. Bush K, Jacoby GA, Medeiros AA. A functional classification
nase from Bacillus cereus 569. Biochem J. 1966;98:11C-3C. scheme for beta-lactamases and its correlation with molecular
Available from: https://www.ncbi.nlm.nih.gov/pubmed/49571 structure. Antimicrob Agents Chemother. 1995;39(6):1211-33.
74. https://doi.org/10.1042/bj0980011c https://doi.org/10.1128/AAC.39.6.1211.
155. Ambler RP. The structure of beta-lactamases. Philos Trans R Soc 162. Lauretti L, Riccio ML, Mazzariol A, Cornaglia G, Amicosante
Lond B Biol Sci. 1980;289:321-31. Available from: https://w ww. G, Fontana R, et al. Cloning and characterization of blaVIM, a
ncbi.n lm.n ih.g ov/p ubmed/6 10932 7. https://d oi.o rg/1 0.1 098/r stb. new integron-borne metallo-beta-lactamase gene from a Pseu-
1980.0049 domonas aeruginosa clinical isolate. Antimicrob Agents Chem-
156. Bicknell R, Waley SG. Cryoenzymology of Bacillus cereus beta- other. 1999;43:1584-90. Available from: https://www.ncbi.nlm.
lactamase II. Biochemistry. 1985;24:6876-87. Available from: nih.gov/pubmed/10390207. https://doi.org/10.1128/AAC.43.7.
https://www.ncbi.nlm.nih.gov/pubmed/3935166. https://doi.org/ 1584
10.1021/bi00345a021 163. Yong D, Toleman MA, Giske CG, Cho HS, Sundman K, Lee K,
157. Jaurin B, Grundström T. ampC cephalosporinase of Escheri- et al. Characterization of a new metallo-beta-lactamase gene,
chia coli K-12 has a different evolutionary origin from that of bla(NDM-1), and a novel erythromycin esterase gene carried on a
beta-lactamases of the penicillinase type. Proc Natl Acad Sci. unique genetic structure in Klebsiella pneumoniae sequence type
1981;78:4897-901. Available from: https://d oi.o rg/1 0.1 073/p nas. 14 from India. Antimicrob Agents Chemother. 2009;53:5046-54.
78.8.4897. https://doi.org/10.1073/pnas.78.8.4897 Available from: https://www.ncbi.nlm.nih.gov/pubmed/19770
158. Huovinen P, Huovinen S, Jacoby GA. Sequence of PSE-2 275. https://doi.org/10.1128/AAC.00774-09
beta-lactamase. Antimicrobial Agents and Chemotherapy. 164. Bush K, Jacoby GA. Updated functional classification of beta-
1988;32:134-6. Available from: https://d oi.o rg/1 0.1 128/a ac.3 2.1. lactamases. Antimicrob Agents Chemother. 2010;54:969-76.
134. https://doi.org/10.1128/aac.32.1.134 Available from: https://www.ncbi.nlm.nih.gov/pubmed/19995
159. Bush K. Classification of beta-lactamases: groups 2c, 2d, 2e, 3, 920. https://doi.org/10.1128/AAC.01009-09
and 4. Antimicrob Agents Chemother. 1989;33:271-6. Available
5
* Carmen Hidalgo‑Tenorio Infectious Diseases Unit, Hospital Universitario y Politécnico
[email protected] La Fe, Valencia, Spain
6
Luis Martínez‑Martínez Microbiology Unit, Hospital Universitario Reina Sofía,
[email protected] Córdoba, Spain
7
1 Department of Agricultural Chemistry, Soil Sciences
Hospital Universitario Virgen de las Nieves de Granada,
and Microbiology, Universidad de Córdoba, Córdoba, Spain
Instituto de Investigación Biosanitario de Granada (IBS-
8
Granada), Granada, Spain Instituto Maimónides de Investigación Biomédica de
2 Córdoba (IMIBIC), Córdoba, Spain
Servicio de Microbiología, Complejo Hospitalario
9
Universitario A Coruña, A Coruña, Spain Centro de Investigación Biomédica en Red de Enfermedades
3 Infecciosas (CIBERINFEC), Instituto de Salud Carlos III,
Servicio de Microbiología y Unidad de Investigación,
Madrid, Spain
Hospital Son Espases, IdISBa, Palma de Mallorca, Spain
10
4 Departamento de Medicina, Universidad de Granada,
Critical Care Department. Hospital, Universitario La
Granada, Spain
Paz. (IdiPaz), Madrid, Spain