1 s2.0 S0924857922001455 Main
1 s2.0 S0924857922001455 Main
1 s2.0 S0924857922001455 Main
a r t i c l e i n f o a b s t r a c t
Article history: Multidrug-resistant (MDR) Gram-negative bacteria (GNB) pose a critical threat to global healthcare,
Received 18 January 2022 worsening outcomes and increasing mortality among infected patients. Carbapenemase- and extended-
Accepted 26 June 2022
spectrum β -lactamase-producing Enterobacterales, as well as carbapenemase-producing Pseudomonas and
Acinetobacter spp., are common MDR pathogens. New antibiotics and combinations have been developed
Editor: Stefania Stefani to address this threat. Clinical trial findings support several combinations, notably ceftazidime–avibactam
(CZA, a cephalosporin-β -lactamase inhibitor combination), which is effective in treating complicated uri-
Keywords:
Antibiotic resistance nary tract infections (cUTI), complicated intra-abdominal infections and hospital-acquired and ventilator-
β -lactam-β -lactamase inhibitor associated pneumonia caused by GNBs. Other clinically effective combinations include meropenem–
Clinical trial vaborbactam (MVB), ceftolozane–tazobactam (C/T) and imipenem–relebactam (I–R). Cefiderocol is a re-
Gram-negative bacteria cent siderophore β -lactam antibiotic that is useful against cUTIs caused by carbapenem-resistant Enter-
obacterales (CRE) and is stable against many β -lactamases. Carbapenem-resistant Enterobacterales are a
genetically heterogeneous group that vary in different world regions and are a substantial cause of in-
fections, among which Klebsiella pneumoniae are the most common. Susceptible CRE infections can be
treated with fluoroquinolones, aminoglycosides or fosfomycin, but alternatives include CZA, MVB, I–R,
cefiderocol, tigecycline and eravacycline. Multidrug-resistant Acinetobacter baumannii and Pseudomonas
aeruginosa are increasingly common pathogens producing a range of different carbapenemases, and in-
fections are challenging to treat, often requiring novel antibiotics or combinations. Currently, no single
agent can treat all MDR-GNB infections, but new β -lactam–β -lactamase inhibitor combinations are of-
ten effective for different infection sites and, when used appropriately, have the potential to improve
outcomes. This article reviews clinical studies investigating novel β -lactam approaches for treatment of
MDR-GNB infections.
© 2022 The Authors. Published by Elsevier Ltd.
This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
https://doi.org/10.1016/j.ijantimicag.2022.106633
0924-8579/© 2022 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
Fig. 1. Priority list of antibiotic-resistant bacteria, other than MDR Mycobacterium tuberculosis, for the research and development of new antibiotics.
Priority list classified by the WHO. Republished with permission of Elsevier Science & Technology journals, from Tacconelli et al. Discovery, research, and development of
new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 2018;18:318–27 [1]; permission conveyed through Copyright Clearance
Center, Inc.
Important initiatives such as the Combating Antibiotic-Resistant with minor activity against class D β -lactamases also observed
Bacteria Biopharmaceutical Accelerator (CARB-X) global non-profit [9] (Fig. 2). In February 2015, avibactam (AVI) (in combination with
partnership, the 10 x ’20 initiative of the Infectious Diseases So- ceftazidime [CAZ]) was the first DBO to be approved by the FDA
ciety of America (IDSA) and others have partly prompted many [11]. Although activity is partner dependent (e.g. with CAZ, ceftaro-
small- and medium-sized enterprises and several pharmaceuti- line, aztreonam, cefepime or imipenem [IPM]), β -lactam-avibactam
cal companies to dedicate resources to the discovery of novel combinations are potentially highly effective against many MDR-
agents with activity against MDR-GNB [5–7]. This has resulted GNB pathogens, including Enterobacterales and P. aeruginosa, pro-
in US Food and Drug Administration (FDA) approval of several ducing class A, C, and some class D enzymes [9,12]. Switching
novel antibacterial agents. This review focuses on combinations the β -lactam partner of a clinically available β -lactamase inhibitor
of old β -lactam antibiotics with novel β -lactamase inhibitors is another approach to treat infections caused by strains that
such as ceftazidime–avibactam (CZA), meropenem–vaborbactam carry multiple classes of β -lactamases, for example the pairing of
(MVB), imipenem–relebactam (I–R), a combination of an old β - tazobactam with the novel cephalosporin, ceftolozane [9,13]. The
lactamase inhibitor with a novel anti-pseudomonal cephalosporin C/T partnership has a spectrum of activity that includes class A,
(ceftolozane–tazobactam [C/T]) and a cephalosporin with a novel- C, and some class D β -lactamases, but not carbapenemases [14].
entry mechanism of action (cefiderocol). Another DBO that has been developed is relebactam (REL), and al-
though it has a similar spectrum of activity to AVI (activity against
2. New β-lactamase inhibitors and a new β-lactam MBLs and most OXAs is lacking), greater activity against OXA-48
has been observed for AVI [15,16] (Fig. 2). Additionally, vaborbac-
The original β -lactam–β -lactamase inhibitor (BL–BLI) com- tam (VAB) (approved for use in combination with meropenem) is
binations (i.e. amoxicillin–clavulanic acid, ampicillin–sulbactam, the first FDA-approved β -lactamase inhibitor containing a cyclic
cefoperazone–sulbactam, piperacillin–tazobactam and ticarcillin– boronate pharmacophore; this forms a covalent adduct with the
clavulanic acid) were highly active against class A serine β - catalytic serine side chain of serine β -lactamases, and can inhibit
lactamases when they were introduced to the market [8,9]. Re- various class A and C β -lactamases [14,17,18].
sistance against these developed with the appearance of four Finally, cefiderocol, the first in its class, is an injectable
structurally and functionally distinct groups of β -lactamases: the siderophore cephalosporin that combines a catechol-type
metallo-β -lactamases (MBLs) of class B, AmpC serine β -lactamases siderophore and cephalosporin core with side chains like cefepime
belonging to class C, oxacillinases (OXA) – serine β -lactamases of and CAZ [19]. This structure and mechanism confer enhanced
class D and a new class A-like Klebsiella pneumoniae (K. pneumo- stability against hydrolysis by many β -lactamases, including ESBLs
niae) carbapenemase (KPC) [8,9]. As a result, a single BL–BLI part- such as CTX-M and carbapenemases such as KPC, NDM, Verona
nership with activity against all clinically important β -lactamases integron-encoded MBL (VIM), imipenem-hydrolysing MBL (IMP),
(e.g. KPC-2, OXA-23, OXA-24/40, AmpC, and New Delhi MBL-1 OXA-23, OXA-48-like, OXA-51-like and OXA-58 (Fig. 2) [19].
[NDM-1]) has been lacking, but there are some new combina-
tions such as cefepime–taniborbactam and cefepime–zidebactam
currently in development that cover a wide spectrum of these 3. Five new β-lactam approaches for various Gram-negative
enzymes and may address this need [9,10]. Diazabicyclooctanes infections
(DBO) are synthetic non-β -lactam-based β -lactamase inhibitors;
there has been an exponential expansion of this class of inhibitors, Key recent clinical studies of novel β -lactam antibiotics and
with most modifications occurring at the C2 side chain [9]. Most combinations in the treatment of MDR-GNB are summarised in
studies indicate that DBOs inhibit class A and class C β -lactamases, Table 1 .
2
Table 1
Design and findings of key clinical studies of novel β -lactam antibiotics in combination with β -lactam inhibitors and other antibiotics in the treatment of multidrug-resistant Gram-negative bacteria.
REPROVE Torres et al. NP, including VAP CZA vs. MEM Randomised, CZA 2 g/0.5 g q8h vs. Clinical cure (at TOC Clinical cure rate, cMITT
n =726 (cMITT), 2018 [24] double-blind, MEM 1 g q8h. visit) CZA: 68.8% MEM:73.0%
n = 527 (CE) Phase III Duration: 7–14 days Clinical cure rate, CE
non-inferiority CZA: 2 h-infusion, MEM: CZA: 77.4% MEM:78.1%
30-min infusion (CZA non-inferior in the treatment of NP)
ASPECT-NP Kollef et al. Ventilated NP C/T vs. MEM Randomised, C/T 3 g q8h vs. MEM 1 g 28-day all-cause C/T: 24.0%
n = 726 (ITT) 2019 [44] controlled, q8h. mortality in ITT MEM: 25.3%
double-blind, Duration: 8–14 days, population (C/T non-inferior for 28-day mortality)
Phase III both given by 1-h
non-inferiority infusion
ASPECT-cIAI Solomkin cIAI C/T + MZL vs. Randomised, C/T 1.5 g + MZL 500 mg Clinical cure at TOC in Clinical cure at TOC in microbiological ITT
n = 806 et al. 2015 MEM prospective, q8h vs. MEM q8h. microbiological ITT population
(microbiological [45] double-blind, Duration: 4–14 days population C/T + MZL: 83%
ITT) non-inferiority MEM: 87.3%
Clinical cure at TOC in microbiologically
evaluable (secondary) population
C/T + MZL:
94.2%
MEM: 94.7%
(prespecified non-inferiority margin met)
ASPECT cUTI Complicated lower C/T vs. LVX Randomised, C/T 1.5 g q8h vs. LVX 750 Composite of C/T: 76.9%
n = 800 (mMITT) Wagenlehner UTI or pyelonephritis double-blind, mg OD. microbiological LVX: 68.4%
et al. 2015 double-dummy, Duration: 7 days eradication and clinical (C/T non-inferior to LVX for composite cure)
[46] non-inferiority cure 5–9 days post
treatment in mMITT
population
TANGO I Kaye et al. cUTI, including acute MVB vs. PPN–TAZ Randomised, MVB (2 g/2 g over 3 h) FDA: Overall success FDA endpoint, overall success
3
n = 374 2018 [116] pyelonephritis multicentre, vs. PPN–TAZ (4 g/0.5 g (clinical cure or MVB: 98.4%
(microbiological double-blind, over 30 min) q8h for 15 improvement and PPN–TAZ: 94.0%
mITT population) double-dummy, doses. microbial eradication EMA endpoint (microbiological mITT):
n = 347 active-control, Mean duration, IV and composite) at end of IV MVB: 66.7%
(microbiological Phase III oral stepdown therapy: treatment in PPN–TAZ: 57.7%
evaluable 10 days microbiological mITT EMA endpoint (microbiological evaluable):
population) population MVB: 66.3%
EMA: microbial PPN–TAZ: 60.4%
eradication at TOC visit (non-inferiority criterion met)
Trial (n) Reference Infection Investigational Design Treatment groups and Primary endpoint Results (% of patients)
drugs dose
APEKS-NP Wunderink Hospital-acquired CDL vs. MEM Randomised, 3 h IV infusion CDL 2 g All-cause mortality at CDL: 12.4%
n = 292 (mITT) 2021 et al. ventilator-associated double-blind, q8h vs. MEM 2 g q8h. Day 14 in mITT MEM: 11.6%
[52] or parallel-group, Duration 7–14 days population (prespecified non-inferiority margin met)
healthcare-associated Phase III,
Gram-negative non-inferiority
pneumonia
CREDIBLE-CR Bassetti Life-threatening CDL vs. BAT Randomised, 3 h IV infusion CDL 2 g For NP, BSI and sepsis, NP, clinical cure
n = 118 (CR-MITT) et al. 2021 carbapenem-resistant open-label, q8h vs. BAT. clinical cure at TOC in CDL: 50%
[53] Gram-negative multicentre, Duration 7–14 days the CR-MITT. For cUTI, BAT: 53%
infections parallel-group, microbiological BSI/sepsis, clinical cure
pathogen- eradication at TOC in the CDL: 43%
focused, CR-MITT BAT: 43%
descriptive, cUTI, microbiological eradication
Phase III CDL: 53%
BAT: 20%
(CDL similar efficacy to BAT)
RESTORE-IMI 1 Motsch et al. Hospitalised patients I–R vs. IPM + CST Multicentre, I–R (500 mg/250 mg, Favourable overall Favourable overall response
n = 31 (mMITT) 2020 [117] with randomised, q6h) vs. IPM (500 mg response (defined by I–R: 71%
HAP/VAP, cIAI or cUTI controlled, q6h) + CST (loading dose relevant endpoints for IPM + CST: 70%
caused by double-blind, 300 mg, then each infection) in mMITT Day 28 all-cause mortality
IPM-resistant Phase III maintenance doses up to population I–R: 10%
pathogens 150 mg, q12h) IPM + CST: 30%
Duration: 5–21 days
RESTORE-IMI 2 Titov et al. HABP or VABP I–R vs. PPN–TAZ Multicentre, I–R (500 mg/500 mg/250 Day 28 all-cause Day 28 all-cause mortality
4
n = 531 2020 [38] randomised, mg) vs. PPN–TAZ (4 mortality in mITT I–R: 15.9%
(mITT) controlled, g/500 mg) q6h. Duration: population PPN–TAZ: 21.3%
double-blind, 7–14 days Favourable clinical response at early
Phase III follow-up
I–R: 61.0%
PPN–TAZ: 55.8%
Dose-ranging study Lucasti et al. cIAI REL (2 doses) vs. Randomised, REL (125 mg), REL (250 Proportion of ME pts Favourable clinical response at DCIV
n = 255 2016 [39] PBO (all + IPM) multicentre, mg), PBO, IV (all + IPM with a favourable clinical REL 250 mg: 96.3%
(ME at DCIV) double-blind, 500 mg) q6h. response at DCIV REL 125 mg: 98.8%
Fig. 2. A) Activity of avibactam, tazobactam, vaborbactam and relebactam against important β -lactamases within different classes; and B) stability of cefiderocol against
these enzymes [16,19,50,143,144].
ESBL, extended-spectrum β -lactamase; KPC, Klebsiella pneumoniae carbapenemase; MBL, metallo-β -lactamase; OXA, oxacillinase.
∗
Enhanced stability to β -lactamases does not always correlate with clinical efficacy.
The combination CZA is an intravenously administered for- Intravenous MVB was the first carbapenem–β -lactamase
mulation of the extended-spectrum cephalosporin CAZ and the inhibitor combination approved in the US for use in pa-
novel, non-β -lactam β -lactamase inhibitor AVI [20]. Ceftazidime– tients with cUTI, including pyelonephritis [18,29]. One compo-
avibactam has potent activity against Enterobacterales carrying nent, meropenem, is active against MDR bacteria, except for
blaKPC and blaOXA-48 and the activity of this combination also ex- carbapenemase-producing strains [18]. The other component, VAB,
tends to MDR P. aeruginosa [16,21]. It is approved in many coun- is a potent inhibitor of class A serine carbapenemases and restores
tries for the treatment of adults and paediatric patients (≥ 3 meropenem activity against class A and class C carbapenemases,
months) with cUTI, including pyelonephritis [22,23], complicated but not against class B or class D carbapenemases [30]. It also
intra-abdominal infections (cIAI) [22,23], hospital-acquired pneu- restores the activity of meropenem against β -lactamase-producing
monia (HAP) including ventilator-associated pneumonia (VAP) [24], Enterobacterales, including KPC- and ESBL-producers [29,31]. It
and other infections caused by aerobic MDR-GNB in patients with may be an alternative for KPC-producing Enterobacterales but
limited treatment options [22,25,26]. The efficacy and safety of CZA has no effect on other MDR bacteria such as A. baumannii [30].
has been studied in five Phase III clinical trials. In the RECAPTURE Meropenem has good activity against most P. aeruginosa strains
study (CZA versus Doripenem for the Treatment of cUTI, Including but not those that have porin mutations or overproduce efflux
Acute Pyelonephritis), CZA was non-inferior to doripenem and both pumps, and VAB does not restore this activity [32–34]. Of note,
treatments showed similar efficacy against CAZ-non-susceptible some CZA-resistant KPC-3 variants (e.g. V240G and D179Y) have
pathogens [26]. The REPRISE study was a pathogen-directed, in- increased susceptibility to meropenem [35], and thus susceptibility
ternational study of 333 patients with cUTI or cIAI caused by to MVB would be expected.
CAZ-resistant Enterobacterales or P. aeruginosa who were treated In the Phase III, non-inferiority Targeting Antibiotic Non-
with CZA or best available treatment (BAT) [22]; clinical cure Susceptible Gram-negative Organisms (TANGO I) trial in patients
rates were similar between the two treatment arms (91% in both with cUTIs, intravenous MVB was found to be non-inferior to
groups). intravenous piperacillin–tazobactam for overall success [29]. The
In another study, CZA plus metronidazole was found to be non- TANGO II trial evaluated the safety and tolerability of MVB alone
inferior to meropenem in clinical cure at test of cure (TOC) and and MVB in combination with a polymyxin (colistin), high-dose
30-day all-cause mortality in the treatment of cIAIs. The efficacy meropenem, CZA (n = 1), or an aminoglycoside versus BAT in pa-
of this combination was similar against infections caused by CAZ- tients with serious CRE infections [36]. Meropenem–vaborbactam
susceptible and CAZ-resistant pathogens [22,24]. In the case of was associated with increased clinical cure, decreased mortality
HAP and VAP, CZA was found to be non-inferior to meropenem and reduced nephrotoxicity compared with BAT [36]. The use of
in terms of clinical cure rates and 30-day all-cause mortality MVB for treating HAP, including VAP, has been approved by the
[24]. European Medicines Agency (EMA) [37].
Several studies have assessed the spectrum of activity and the
efficacy of CZA in the treatment of infections due to MDR-GNB 3.3. Imipenem–relebactam (I–R)
[14]. A meta-analysis showed in 12 studies that this combination
had a pooled clinical success rate of 73% (95% CI 67.7–78.4%) in Imipenem–relebactam, a novel BL–BLI combination, was re-
treating CRE and MDR P. aeruginosa infections (mainly pneumo- cently approved for the treatment of cUTI, cIAI, HAP and VAP [38].
nia) [14]. Based on this evidence, the UK Clinical Pharmacy As- Relebactam is a β -lactamase inhibitor with the ability to inhibit
sociation recommends CZA for the treatment of KPC- and OXA- a broad spectrum of β -lactamases such as class A and class C β -
48-producing Enterobacterales [27]. In addition, a recent observa- lactamases, including carbapenemases [39]. The addition of REL to
tional study showed a therapeutic advantage for CZA in combina- IPM restores IPM activity against several IPM-resistant bacteria, in-
tion with aztreonam compared with other active antibiotics in pa- cluding MDR P. aeruginosa and Enterobacterales such as CRE KPC
tients with bloodstream infections (BSI) due to MBL-producing En- producers [39,40]. Imipenem–relebactam has also shown clinical
terobacterales [28]. activity against several other aerobic (Escherichia coli, Enterobac-
5
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
6
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
Table 2
Multidrug-resistant Gram-negative pathogens – key drugs and characteristics.
7
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
to 2017, most of which were community acquired, although where > 500 patients had CDC-defined CRKP, 83% were carbapen-
healthcare-associated infections (HAI) are also a major source of emase producers and 94% of these expressed KPC [70].
these infections [60]. The ESBL-producing Enterobacterales are a The site of infection and knowledge of carbapenemase pro-
serious threat due to their resistance to many extended-spectrum duction are important, as these factors will guide treatment de-
penicillins and other agents, including cephalosporins and aztre- cisions [54]. Similar to infections with other MDR pathogens, poor
onam. Furthermore, they often have additional genes conferring re- source control and failure to implement early rapid diagnostics can
sistance to a wider range of antibiotics, including quinolones. ESBLs delay proper management(manuscript submitted for publication).
are produced across the Enterobacterales, and high rates of these The lack of availability of new drugs such as the newer BL–BLI
enzymes have been reported in E. coli, K. pneumoniae, K. oxytoca agents, plazomycin, cefiderocol and eravacycline, in some countries
and Proteus mirabilis [61,62]. The CTX-M ESBL inactivates a wide may hinder adequate treatment and force clinicians to use alterna-
range of antibiotics, including cefotaxime and ceftriaxone, and over tive drugs with lower efficacy and higher toxicity.
time has become more active against CAZ [63]. Bacteria producing Whenever bacterial pathogens are susceptible, preferable treat-
this enzyme are rapidly spreading worldwide and this is the most ments for CRE infections such as cystitis are ciprofloxacin,
frequently reported ESBL in the US [61]. This type of ESBL is dis- levofloxacin, trimethoprim–sulfamethoxazole, nitrofurantoin, fos-
tinct from β -lactamases such as TEM and SHV, which have a nar- fomycin or a single dose of an aminoglycoside [54]. Alternatively,
rower range of action [64,65]. CZA, MVB or cefiderocol can be used. For pyelonephritis or cUTI,
The MERINO trial did not support the use of piperacillin– preferred treatments are CZA, MVB, I–R, or cefiderocol, or, alterna-
tazobactam vs. meropenem in K. pneumoniae or E. coli BSIs with tively, aminoglycosides [54].
ceftriaxone resistance [66]. Following these results, carbapenems For non-UTI infections or where KPC has been identified, pre-
were considered the standard of care for the treatment of bac- ferred treatment options are CZA, MVB and I–R. Cefiderocol (for
teraemia with ESBL-producing Enterobacterales infections. How- indications other than BSI) and eravacycline or tigecycline (for non-
ever, this is controversial as there is a counter argument that car- bacteraemic intra-abdominal infection [IAI] only or in combination)
bapenems are not necessary for all patients with infections caused are alternative options, depending on the resistance profile of the
by ceftriaxone-resistant Enterobacterales, and that BL–BLIs can be pathogen (notably resistance to ertapenem or meropenem) [54]. In
used in appropriate settings [67]. This challenge is based on a re- cases in which an MBL is detected, the preferred treatment op-
port that some of the MICs in the MERINO trial may have been tions are CZA plus aztreonam or cefiderocol [54]. In cases where an
incorrectly determined by local laboratories who used MIC strip OXA-48-like carbapenemase has been detected, the preferred treat-
tests, and the observation that piperacillin–tazobactam was not ad- ment is CZA, or alternatively cefiderocol (for indications other than
ministered as an extended infusion [67,68]. Isolates from patients BSI), tigecycline or eravacycline (for cIAI only). Combination thera-
with worse outcomes were frequently found to be non-susceptible pies such as a β -lactam with an aminoglycoside, fluoroquinolone
to piperacillin–tazobactam when MICs were determined using ref- or polymyxin are not recommended for treating CRE infections if
erence methods [67]. Alternative new drugs and combinations with the pathogen is susceptible to a β -lactam [54].
in vitro activity against ESBL-producing Enterobacterales include: For cystitis caused by CRE that are resistant to ertapenem but
CZA, C/T, I–R, MVB, cefiderocol and plazomicin [50,59]. susceptible to meropenem, without available carbapenemase test-
ing, a standard infusion of meropenem may work due to high
achievable concentrations in the urine [54]. In cases of cystitis
4.2. Carbapenem-resistant Enterobacterales caused by CRE that are resistant to meropenem and other agents
such as aminoglycosides, including amikacin and plazomicin, al-
The CREs are a more serious threat than ESBLs; they account ternative options include CZA, MVB, I–R and cefiderocol [54]. For
for > 13 0 0 0 nosocomial infections and > 10 0 0 deaths per year isolates producing MBLs (NDM, IMP or VIM), CZA plus aztreonam,
in the US [54]. Carbapenemase-producing isolates are the cause of aztreonam–avibactam or cefiderocol are preferred [53,79]. Colistin
ca. 50% of all CRE infections in the US [69,70]. There is hetero- (not polymyxin B), notwithstanding the side effects, is also an al-
geneity within the CRE, with differing sets of carbapenemases be- ternative, although not preferred [54,80].
ing produced, and consequently there is resistance to various car- Complicated UTI and pyelonephritis caused by CRKP that is re-
bapenem antibiotics. The most common of the Ambler classes A, sistant to both ertapenem or meropenem are best treated with
B and D enzymes are KPCs of the class A type, and these can be CZA, MVB, I–R or cefiderocol [54]. Oral fosfomycin should be
produced by any of the species belonging to the Enterobacterales. avoided [81,82]. For infections outside the urinary tract caused
In the US, the most prominent type of carbapenemases are KPCs; by K. pneumoniae that are resistant to both ertapenem and
other types such as class B MBLs (e.g. NDM, VIM, IMP) and class meropenem, it is important to identify the specific carbapenemase
D oxacillinase (e.g. OXA-48-like) are much less frequently reported gene to give specific treatment [75,83].
[71,72]. It should be noted that carbapenem resistance not only For KPC producers, the preferred options are CZA, MVB and I–R,
arises from carbapenemase production but also decreased porin with cefiderocol as an alternative [81]. For IAIs, depending on the
expression and increased efflux pump action [73,74]. severity of the infection, tigecycline is an alternative treatment but
Carbapenem-resistant K. pneumoniae are the most common and can be limited by gastrointestinal side effects, particularly when
clinically important of the CRE reported worldwide [75]. The geo- high doses are used; eravacycline is a newer alternative but data
graphic variation of the carbapenem resistance genes varies across are limited for this monotherapy [54,81,84]. For isolates producing
continents [75]. When comparing studies, it is important to note MBLs, such as NDM, IMP or VIM, the preferred drugs are CZA with
that phenotypic definitions of carbapenem-resistant K. pneumoniae aztreonam or cefiderocol alone [54]. For Enterobacterales produc-
(CRKP) may differ. In South-East Asia, NDM and other MBLs (e.g. ing OXA-48-like carbapenemases, CZA is preferred [54].
IMP, VIM) and OXA-48–type are predominant carbapenemases in
CRE [76]. In the European Survey on carbapenemase-producing 4.3. Carbapenem-resistant Acinetobacter baumannii
Enterobacteriaceae (EuSCAPE), 37% of the tested CRKP had a car-
bapenemase gene: KPC (42%) and OXA-48 (38%) were predomi- Acinetobacter baumannii is found in many healthcare environ-
nantly encoded [77]. In the US, the CDC Gram-negative initiative ments and is a highly effective human coloniser in hospitals [85].
surveillance revealed that 47.9% of tested CRE isolates were KPC- Acinetobacter baumannii can easily survive on multiple surfaces,
producing species [78]. In another study from the US (CRACKLE-2), causing hospital infections and leading to numerous global out-
8
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
breaks [86,87]. Furthermore, there have been reports of CRAB An analysis of seven studies (total = 1613 patients) found that
strains being transmitted from countries with high antimicrobial patients with CRPA BSIs were at higher risk of death than non-
resistance rates to countries where the rates are usually low, such CRPA BSIs (pooled ORs, 3.07 and 1.46) [102] and showed that 8–
as from Spain to Norway [87]. Infections due to A. baumannii are 18.4% of deaths in this study were attributable to carbapenem re-
frequently found in intensive care units (ICUs), where they are im- sistance. Risk factors for CRPA infection include carbapenem use,
plicated as the cause of VAP, UTIs and bacteraemia [88]. The preva- other antibiotic use [103,104] such as fluoroquinolones, medical
lence of A. baumannii infection and colonisation is higher in ICUs devices and ICU admission [105]. Additionally, a study in Japan
since patients with severe clinical conditions are often hospitalised showed a clear correlation between carbapenem consumption and
in these facilities. the prevalence of CRPA on a national scale [95].
Antibiotic resistance in A. baumannii is frequently due to low Alternative treatments to carbapenems include antipseu-
outer membrane permeability, antibiotic binding-site modifications domonal drugs (e.g. antipseudomonal cephalosporins such as
and efflux pump expression [89,90]. The acquisition of carbapene- CAZ, cefepime and piperacillin–tazobactam) and are usually part-
mases is also an important mechanism of resistance for CRAB and nered with another active agent such as an aminoglycoside (e.g.
resistance genes can also be gained through the acquisition of mo- amikacin) [103]. Carbapenems should be reserved for polymicro-
bile genetic elements [89,91]. bial infections or P. aeruginosa isolates that are resistant to other
Various antimicrobials are used against CRAB, including β -lactams [106]. Resistance to antipseudomonal drugs is increasing
polymyxins (such as polymyxins B or E [colistin]), tetracyclines and MDR P. aeruginosa showing resistance to three or more drug
(such as tigecycline and minocycline), β -lactams in combination classes has emerged and spread across different regions worldwide
with β -lactamase inhibitors (such as sulbactam) and other com- [107].
binations such as trimethoprim–sulfamethoxazole. However, many The C/T combination is one of the preferred treatment op-
A. baumannii strains show high-level resistance to these agents tions for MDR/XDR Pseudomonas infections [108]. In an analysis of
and there are pharmacokinetic limitations with polymyxins and 11 clinical studies (number of strains ranged from 38 to 3229),
tetracyclines: high and potentially toxic doses are necessary to the susceptibility of MDR/XDR P. aeruginosa to C/T ranged from
achieve good tissue levels [92]. However, the newer tetracycline, 55–96.6% [108]. When compared with other agents, C/T had a
eravacycline, shows efficacy against CRAB infections with low MIC lower probability of co-resistance in β -lactam-resistant P. aerugi-
values. This drug was shown to be non-inferior to ertapenem nosa [48]; however, dosing and infusion need to be optimised ac-
and meropenem against CRAB in two randomised clinical trials cording to indication. Other notable effective approaches to treat-
but is only approved for use for cIAI. However, in two further ing CRPA infections include cefiderocol, CZA and I–R [81].
Phase III clinical trials, eravacycline was inferior to levofloxacin
and ertapenem in the treatment of cUTI [92,93]. Cefiderocol has 5. Real-world evidence
shown efficacy in CRAB infections and non-inferiority to IPM in the
treatment of cUTIs has been demonstrated [19,92]. Although it is Various observational studies have added to the body of clin-
now approved for the treatment of cUTIs, the CREDIBLE-CR study ical trial evidence showing the viability of using monotherapies
(where patients with pneumonia, sepsis and UTI participated, and and antibiotic combinations to treat MDR-GNB infections in reg-
where CRAB isolates were identified in 46% of patients) all-cause ular clinical practice. A recent study involving 22 clinical centres
mortality at the end of the study was higher when using cefidero- from Italy (n = 577) showed that CZA is an effective option for
col (50%) compared with BAT (18%) in patients with Acinetobacter treating KPC infections (e.g. BSIs and UTIs) when used alone [109].
spp. positive cultures [53]. More investigations are needed to support its use in lower respi-
The OXA (class D) are the most prevalent types of carbapene- ratory tract infections and the potential benefits of longer infusion
mases among A. baumannii isolates worldwide. Among these are times [109]. A retrospective cohort study (n = 203) found that CZA
six subclasses: OXA-51-like, OXA-23-like, OXA-24/40-like, OXA-58- did not impact clinical failure in the overall population, or high-
like, OXA-143-like and OXA-235-like [94]. To address such resistant risk subgroups or 30-day mortality among those with CRE or Pseu-
infections, new approaches are being developed for treating re- domonas spp. infections [110]. However, receipt of CZA within 48
sistant A. baumannii infections, including new β -lactam inhibitors hours of infection onset was associated with improved clinical out-
in combination with existing β -lactams and non-β -lactams, new comes [110]. It was concluded that CZA can be an effective ther-
polymyxins, a new aminoglycoside (apramycin), monoclonal anti- apy for CRE and MDR Pseudomonas, but that there is a need for
body treatments and bacteriophages [93]. advances in the treatment of vulnerable patients with pneumonia
and severe renal impairment.
Real-world studies and observational studies have shown the
4.4. Carbapenem-resistant Pseudomonas aeruginosa efficacy and safety of C/T in a range of different MDR/XDR P. aerug-
inosa infections, including UTI, IAI, HAP, VAP and skin/soft tissue
Pseudomonas aeruginosa is increasingly associated with nosoco- infections [42,108,111–113]. The C/T combination has also shown
mial infections, particularly in immunocompromised patients and lower toxicity than BATs such as polymyxin or aminoglycosides in
those with indwelling devices [95]. Carbapenem-resistant P. aerug- the treatment of P. aeruginosa infections [114].
inosa is associated with a high risk of mortality. Worldwide, MBLs Although the body of real-world evidence is growing, more of
are the most prevalent type of carbapenemase produced by clin- these studies are needed to determine the value of new antibi-
ical isolates of CRPA; VIM MBLs are the most widely dissemi- otic combinations in everyday use in critically ill patients, and to
nated, followed by imipenemases [96]. In many territories, car- evaluate their usefulness compared with older treatment options
bapenem resistance is due to MBLs that diminish the usefulness such as polymyxins, tigecycline, fosfomycin and aminoglycosides
of β -lactamase inhibitors in the treatment of CRPA infections [97]. [20,115].
In the US, 10–20% of P. aeruginosa isolates are resistant to at least
one carbapenem, making this resistance a serious problem [98,99]. 6. Conclusions and final recommendation
Compared with worldwide literature, India reports increasing MBLs
(NDM and VIM) in addition to other resistance mechanisms, which Antimicrobial resistance in GNB is a global concern and signifi-
further compromise the efficacy of the newer BL–BLIs [100], whilst cantly affects outcomes in patients with limited options for treat-
KPC-producing P. aeruginosa is becoming a concern in China [101]. ment. To date, there is no single agent BL–BLI with a spectrum
9
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
that covers all MDR-GNB in empiric or targeted therapy. Whilst ce- [6] Talbot GH, Jezek A, Murray BE, Jones RN, Ebright RH, Nau GJ, et al. The In-
fiderocol provides broad coverage, further analyses are warranted fectious Diseases Society of America’s 10 x ’20 Initiative (10 New Systemic
Antibacterial Agents US Food and Drug Administration Approved by 2020): Is
to fully understand its effectiveness against certain MDR Gram- 20 x ’20 a Possibility? Clin Infect Dis 2019;69:1–11.
negative pathogens such as Acinetobacter spp. and difficult-to-treat [7] Theuretzbacher U, Outterson K, Engel A, Karlen A. The global preclinical an-
resistance Pseudomonas aeruginosa. The availability of new antibi- tibacterial pipeline. Nat Rev Microbiol 2020;18:275–85.
[8] Drawz SM, Bonomo RA. Three decades of beta-lactamase inhibitors. Clin Mi-
otics and the use of novel combinations, such as the new BL–BLI crobiol Rev 2010;23:160–201.
combinations, provide hope for the successful management of var- [9] Papp-Wallace KM, Bonomo RA. New beta-Lactamase Inhibitors in the Clinic.
ious pathogens at different sites of infection. Effective and well- Infect Dis Clin North Am 2016;30:441–64.
[10] Yahav D, Giske CG, Gramatniece A, Abodakpi H, Tam VH, Leibovici L.
managed antimicrobial stewardship policies are essential to ensure
New beta-Lactam-beta-Lactamase Inhibitor Combinations. Clin Microbiol Rev
that these agents are properly used, and thus maintain their po- 2020;34:e00115–20.
tency and help limit or slow resistance development. [11] US Food and Drug Administration. Drug Trials Snapshot: AVYCAZ (cIAI),
2015. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-
snapshot- avycaz- ciai [accessed 06 August 2021].
Acknowledgements [12] Nichols WW, Newell P, Critchley IA, Riccobene T, Das S. Avibactam Pharma-
cokinetic/Pharmacodynamic Targets. Antimicrob Agents Chemother 2018;62
e02446-17.
The authors would like to thank Clóvis Arns da Cunha for his [13] Zhanel GG, Chung P, Adam H, Zelenitsky S, Denisuik A, Schweizer F, et al.
contribution to the conception and design of this review. The au- Ceftolozane/tazobactam: a novel cephalosporin/beta-lactamase inhibitor com-
thors would also like to thank Lucid Group Communications Ltd, bination with activity against multidrug-resistant gram-negative bacilli. Drugs
2014;74:31–51.
Marlow, Buckinghamshire, UK, for providing editorial and medical
[14] Wilson GM, Fitzpatrick M, Walding K, Gonzalez B, Schweizer ML, Suda KJ,
writing support (which was funded by Pfizer, who were able to re- et al. Meta-analysis of Clinical Outcomes Using Ceftazidime/Avibactam,
view and provide feedback on medical accuracy and fair balance). Ceftolozane/Tazobactam, and Meropenem/Vaborbactam for the Treatment
of Multidrug-Resistant Gram-Negative Infections. Open Forum Infect Dis
2021;8:ofaa651.
Declarations [15] de Sousa Coelho F, Mainardi JL. The multiple benefits of second-generation
beta-lactamase inhibitors in treatment of multidrug-resistant bacteria. Infect
Dis Now 2021 S2666-9919(20)0 0 017-2.
Funding: Editorial and medical writing support was funded by
[16] Vazquez-Ucha JC, Arca-Suarez J, Bou G, Beceiro A. New Carbapenemase
Pfizer. Inhibitors: Clearing the Way for the beta-Lactams. Int J Mol Sci 2020;21:9308.
Competing Interests: The authors declare the following finan- [17] Hecker SJ, Reddy KR, Totrov M, Hirst GC, Lomovskaya O, Griffith DC, et al.
Discovery of a Cyclic Boronic Acid beta-Lactamase Inhibitor (RPX7009)
cial interests/personal relationships that may be considered as po-
with Utility vs Class A Serine Carbapenemases. J Med Chem
tential competing interests. SSK has received honoraria for ad- 2015;58:3682–92.
visory boards, conferences from Pfizer, Merck, Gilead, Novartis [18] Lee YR, Baker NT. Meropenem-vaborbactam: a carbapenem and beta-lacta-
and Menarini and honoraria for author contributions to UpTo- mase inhibitor with activity against carbapenem-resistant Enterobacteriaceae.
Eur J Clin Microbiol Infect Dis 2018;37:1411–19.
Date. MB has received research grants and/or has been an advi- [19] Wu JY, Srinivas P, Pogue JM. Cefiderocol: A Novel Agent for the Man-
sor/consultant and/or speaker/chairman for the following: Angelini, agement of Multidrug-Resistant Gram-Negative Organisms. Infect Dis Ther
Bayer, bioMérieux, Cidara, Cipla, Gilead, Menarini, MSD, Pfizer, Sh- 2020;9:17–40.
[20] Karaiskos I, Lagou S, Pontikis K, Rapti V, Poulakou G. The "Old" and the "New"
ionogi. MVV has received honoraria and research grants from MSD, Antibiotics for MDR Gram-Negative Pathogens: For Whom, When, and How.
Pfizer, bioMérieux, West and GPC pharma. CR has received advisory Front Public Health 2019;7:151.
board and speaker honoraria from Pfizer, Sanofi and bioMérieux, [21] Yang Y, Guo Y, Yin D, Zheng Y, Wu S, Zhu D, et al. In Vitro Activ-
ity of Cefepime-Zidebactam, Ceftazidime-Avibactam, and Other Compara-
and speaker honoraria from B. Braun, Becton Dickinson, Cipla, tors against Clinical Isolates of Enterobacterales, Pseudomonas aeruginosa,
Glenmark, Novartis, and Cepheid. DvD is a consultant for Actavis, and Acinetobacter baumannii: Results from China Antimicrobial Surveil-
Tetraphase, Sanofi Pasteur, MedImmune, Astellas, Merck, Allergan, lance Network (CHINET) in 2018. Antimicrob Agents Chemother 2020;65
e01726-20.
T2Biosystems, Roche, Achaogen, Neumedicine, Shionogi, Pfizer, En-
[22] Carmeli Y, Armstrong J, Laud PJ, Newell P, Stone G, Wardman A, et al. Cef-
tasis, QPex, Wellspring, Karius, and Utility. DvD receives an editor’s tazidime-avibactam or best available therapy in patients with ceftazidime-re-
stipend from BSAC. DvD reports grants from Shionogi, Merck, NIH, sistant Enterobacteriaceae and Pseudomonas aeruginosa complicated urinary
tract infections or complicated intra-abdominal infections (REPRISE):
outside the submitted work. All authors received an honorarium
a randomised, pathogen-directed, phase 3 study. Lancet Infect Dis
from Pfizer to attend a steering committee meeting to discuss their 2016;16:661–73.
regional perspective on MDR Gram-negative infections and antimi- [23] Mazuski JE, Gasink LB, Armstrong J, Broadhurst H, Stone GG, Rank D, et al.
crobial stewardship strategies. The output of this discussion was Efficacy and Safety of Ceftazidime-Avibactam Plus Metronidazole Versus
Meropenem in the Treatment of Complicated Intra-abdominal Infection: Re-
used to create to a framework for this manuscript prior to its de- sults From a Randomized, Controlled, Double-Blind, Phase 3 Program. Clin In-
velopment. fect Dis 2016;62:1380–9.
Ethical Approval: Not required. [24] Torres A, Zhong N, Pachl J, Timsit JF, Kollef M, Chen Z, et al. Ceftazidime-av-
ibactam versus meropenem in nosocomial pneumonia, including ventila-
Sequence Information: Not applicable. tor-associated pneumonia (REPROVE): a randomised, double-blind, phase 3
non-inferiority trial. Lancet Infect Dis 2018;18:285–95.
References [25] Zaviceft a 2 g/0.5g powder for concentrate for solutionfor infusion - summary
of product characteristics, 2021. https://www.medicines.org.uk/emc/product/
[1] Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, 2465 [accessed 06 August 2021].
et al. Discovery, research, and development of new antibiotics: the WHO pri- [26] Wagenlehner FM, Sobel JD, Newell P, Armstrong J, Huang X, Stone GG, et al.
ority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis Ceftazidime-avibactam Versus Doripenem for the Treatment of Complicated
2018;18:318–27. Urinary Tract Infections, Including Acute Pyelonephritis: RECAPTURE, a Phase
[2] WHO Global priority list of antibiotics-resistant bacteria to guide research, 3 Randomized Trial Program. Clin Infect Dis 2016;63:754–62.
discovery, and development of new antibiotics. Geneva: World Health Organi- [27] Hughes S, Gilchrist M, Heard K, Hamilton R, Sneddon J. Treating infections
zation; 2017. https://www.who.int/medicines/publications/WHO- PPL- Short_ caused by carbapenemase-producing Enterobacterales (CPE): a pragmatic ap-
Summary_25Feb-ET_NM_WHO.pdf. [accessed 09 April 2021]. proach to antimicrobial stewardship on behalf of the UKCPA Pharmacy Infec-
[3] Centers for Disease Control and Prevention Facility guidance for control tion Network (PIN). JAC Antimicrob Resist 2020;2 dlaa075.
of carbapenem-resistant Enterobacteriaceae—November 2015 update. Atlanta, [28] Falcone M, Daikos GL, Tiseo G, Bassoulis D, Giordano C, Galfo V, et al. Efficacy
GA: CDC; 2015. https://www.cdc.gov/hai/pdfs/cre/CRE- guidance- 508.pdf. [ac- of Ceftazidime-avibactam Plus Aztreonam in Patients With Bloodstream In-
cessed 31 March 2021]. fections Caused by Metallo-β -lactamase-Producing Enterobacterales. Clin In-
[4] Centers for Disease Control. Antibiotic resistance threats in the United fect Dis 2021;72:1871–8.
States, 2019. https://www.cdc.gov/drugresistance/pdf/threats- report/2019- ar- [29] Dhillon S. Meropenem/Vaborbactam: A Review in Complicated Urinary Tract
threats-report-508.pdf [accessed 01 April 2021]. Infections. Drugs 2018;78:1259–70.
[5] Alm RA, Gallant K. Innovation in Antimicrobial Resistance: The CARB-X Per- [30] Vena A, Castaldo N, Bassetti M. The role of new beta-lactamase inhibitors in
spective. ACS Infect Dis 2020;6:1317–22. gram-negative infections. Curr Opin Infect Dis 2019;32:638–46.
10
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
[31] Sun D, Rubio-Aparicio D, Nelson K, Dudley MN, Lomovskaya O. Meropen- treatment of Gram-negative nosocomial pneumonia (APEKS-NP): a ran-
em-Vaborbactam Resistance Selection, Resistance Prevention, and Molecular domised, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis
Mechanisms in Mutants of KPC-Producing Klebsiella pneumoniae. Antimicrob 2021;21:213–25.
Agents Chemother 2017;61 e01694-17. [53] Bassetti M, Echols R, Matsunaga Y, Ariyasu M, Doi Y, Ferrer R, et al. Effi-
[32] Joly-Guillou ML, Kempf M, Cavallo JD, Chomarat M, Dubreuil L, Maugein J, cacy and safety of cefiderocol or best available therapy for the treatment
et al. Comparative in vitro activity of Meropenem, Imipenem and of serious infections caused by carbapenem-resistant Gram-negative bacte-
Piperacillin/tazobactam against 1071 clinical isolates using 2 different meth- ria (CREDIBLE-CR): a randomised, open-label, multicentre, pathogen-focused,
ods: a French multicentre study. BMC Infect Dis 2010;10:72. descriptive, phase 3 trial. Lancet Infect Dis 2021;21:226–40.
[33] Jorgensen SCJ, Rybak MJ. Meropenem and Vaborbactam: Stepping up the [54] Tamma PD, Aitken SL, Bonomo RA, Mathers AJ, van Duin D, Clancy CJ. In-
Battle against Carbapenem-resistant Enterobacteriaceae. Pharmacotherapy fectious Diseases Society of America Guidance on the Treatment of Extend-
2018;38:444–61. ed-Spectrum beta-lactamase Producing Enterobacterales (ESBL-E), Carbapen-
[34] Lapuebla A, Abdallah M, Olafisoye O, Cortes C, Urban C, Quale J, et al. Activity em-Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Diffi-
of Meropenem Combined with RPX7009, a Novel beta-Lactamase Inhibitor, cult-to-Treat Resistance (DTR-P. aeruginosa). Clin Infect Dis 2021;72:1109–16.
against Gram-Negative Clinical Isolates in New York City. Antimicrob Agents [55] Clinicaltrials.gov. NCT03869437. https://clinicaltrials.gov/ct2/show/
Chemother 2015;59:4856–60. NCT03869437 [accessed 22 April 2022].
[35] Shields RK, Chen L, Cheng S, Chavda KD, Press EG, Snyder A, et al. Emer- [56] Lukac PJ, Bonomo RA, Logan LK. Extended-spectrum beta-lactamase-produc-
gence of Ceftazidime-Avibactam Resistance Due to Plasmid-Borne blaKPC-3 ing Enterobacteriaceae in children: old foe, emerging threat. Clin Infect Dis
Mutations during Treatment of Carbapenem-Resistant Klebsiella pneumoniae 2015;60:1389–97.
Infections. Antimicrob Agents Chemother 2017;61 e02097-16. [57] Musa BM, Imam H, Lendel A, Abdulkadir I, Gumi HS, Aliyu MH, et al. The
[36] Wunderink RG, Giamarellos-Bourboulis EJ, Rahav G, Mathers AJ, Bassetti M, burden of extended-spectrum beta-lactamase-producing Enterobacteriaceae
Vazquez J, et al. Effect and Safety of Meropenem-Vaborbactam versus in Nigeria: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg
Best-Available Therapy in Patients with Carbapenem-Resistant Enterobacte- 2020;114:241–8.
riaceae Infections: The TANGO II Randomized Clinical Trial. Infect Dis Ther [58] Rodriguez-Bano J, Pascual A. Clinical significance of extended-spectrum be-
2018;7:439–55. ta-lactamases. Expert Rev Anti Infect Ther 2008;6:671–83.
[37] Carvalhaes CG, Shortridge D, Sader HS, Castanheira M. Activity of Meropen- [59] Pana ZD, Zaoutis T. Treatment of extended-spectrum beta-lactamase-produc-
em-Vaborbactam against Bacterial Isolates Causing Pneumonia in Patients in ing Enterobacteriaceae (ESBLs) infections: what have we learned until now?
U.S. Hospitals during 2014 to 2018. Antimicrob Agents Chemother 2020;64 F10 0 0Res 2018;7:F10 0 0.
e02177-19. [60] Jernigan JA, Hatfield KM, Wolford H, Nelson RE, Olubajo B, Reddy SC,
[38] Titov I, Wunderink RG, Roquilly A, Rodriguez Gonzalez D, David-Wang A, et al. Multidrug-Resistant Bacterial Infections in U.S. Hospitalized Patients,
Boucher HW, Randomized A, et al. Double-blind, Multicenter Trial 2012-2017. N Engl J Med 2020;382:1309–19.
Comparing Efficacy and Safety of Imipenem/Cilastatin/Relebactam Ver- [61] Doi Y, Iovleva A, Bonomo RA. The ecology of extended-spectrum beta-lacta-
sus Piperacillin/Tazobactam in Adults With Hospital-acquired or Ventila- mases (ESBLs) in the developed world. J Travel Med 2017;24:S44–51.
tor-associated Bacterial Pneumonia (RESTORE-IMI 2 Study). Clin Infect Dis [62] Tamma PD, Sharara SL, Pana ZD, Amoah J, Fisher SL, Tekle T, et al. Molec-
2020:ciaa803. ular Epidemiology of Ceftriaxone Non-Susceptible Enterobacterales Isolates
[39] Lucasti C, Vasile L, Sandesc D, Venskutonis D, McLeroth P, Lala M, et al. Phase in an Academic Medical Center in the United States. Open Forum Infect Dis
2, Dose-Ranging Study of Relebactam with Imipenem-Cilastatin in Subjects 2019;6:ofz353.
with Complicated Intra-abdominal Infection. Antimicrob Agents Chemother [63] Bonnet R. Growing group of extended-spectrum beta-lactamases: the CTX-M
2016;60:6234–43. enzymes. Antimicrob Agents Chemother 2004;48:1–14.
[40] Mansour H, Ouweini AE, Chahine EB, Karaoui LR. [64] Bush K, Bradford PA. Epidemiology of beta-Lactamase-Producing Pathogens.
Imipenem/cilastatin/relebactam: A new carbapenem beta-lactamase in- Clin Microbiol Rev 2020;33 e0 0 047-19.
hibitor combination. Am J Health Syst Pharm 2021;78:674–83. [65] Bush K, Jacoby GA. Updated functional classification of beta-lactamases. An-
[41] Kohno S, Bando H, Yoneyama F, Kikukawa H, Kawahara K, Shirakawa M, timicrob Agents Chemother 2010;54:969–76.
et al. The safety and efficacy of relebactam/imipenem/cilastatin in Japanese [66] Harris PNA, Tambyah PA, Lye DC, Mo Y, Lee TH, Yilmaz M, et al. Effect of
patients with complicated intra-abdominal infection or complicated urinary Piperacillin-Tazobactam vs Meropenem on 30-Day Mortality for Patients With
tract infection: A multicenter, open-label, noncomparative phase 3 study. J E coli or Klebsiella pneumoniae Bloodstream Infection and Ceftriaxone Resis-
Infect Chemother 2021;27:262–70. tance: A Randomized Clinical Trial. JAMA 2018;320:984–94.
[42] Gallagher JC, Satlin MJ, Elabor A, Saraiya N, McCreary EK, Molnar E, [67] Rodriguez-Bano J, Gutierrez-Gutierrez B, Pascual A. CON: Carbapenems are
et al. Ceftolozane-Tazobactam for the Treatment of Multidrug-Resistant Pseu- NOT necessary for all infections caused by ceftriaxone-resistant Enterobac-
domonas aeruginosa Infections: A Multicenter Study. Open Forum Infect Dis terales. JAC Antimicrob Resist 2021;3:dlaa112.
2018;5:ofy280. [68] Henderson A, Paterson DL, Chatfield MD, Tambyah PA, Lye DC, De PP,
[43] ZERBAXA®. Highlights of Prescribing Information. 2020. https://www.merck. et al. Association between minimum inhibitory concentration, beta-lacta-
com/product/usa/pi_circulars/z/zerbaxa/zerbaxa_pi.pdf [accessed 22 April mase genes and mortality for patients treated with piperacillin/tazobactam
2022]. or meropenem from the MERINO study. Clin Infect Dis 2020:ciaa1479.
[44] Kollef MH, Novacek M, Kivistik U, Rea-Neto A, Shime N, Martin-Loeches I, [69] Tamma PD, Goodman KE, Harris AD, Tekle T, Roberts A, Taiwo A,
et al. Ceftolozane-tazobactam versus meropenem for treatment of nosoco- et al. Comparing the Outcomes of Patients With Carbapenemase-Produc-
mial pneumonia (ASPECT-NP): a randomised, controlled, double-blind, phase ing and Non-Carbapenemase-Producing Carbapenem-Resistant Enterobacteri-
3, non-inferiority trial. Lancet Infect Dis 2019;19:1299–311. aceae Bacteremia. Clin Infect Dis 2017;64:257–64.
[45] Solomkin J, Hershberger E, Miller B, Popejoy M, Friedland I, Steenber- [70] van Duin D, Arias CA, Komarow L, Chen L, Hanson BM, Weston G, et al.
gen J, et al. Ceftolozane/Tazobactam Plus Metronidazole for Complicated In- Molecular and clinical epidemiology of carbapenem-resistant Enterobac-
tra-abdominal Infections in an Era of Multidrug Resistance: Results From terales in the USA (CRACKLE-2): a prospective cohort study. Lancet Infect Dis
a Randomized, Double-Blind, Phase 3 Trial (ASPECT-cIAI). Clin Infect Dis 2020;20:731–41.
2015;60:1462–71. [71] Logan LK, Weinstein RA. The Epidemiology of Carbapenem-Resistant Enter-
[46] Wagenlehner FM, Umeh O, Steenbergen J, Yuan G, Darouiche RO. Ceftolozane– obacteriaceae: The Impact and Evolution of a Global Menace. J Infect Dis
tazobactam compared with levofloxacin in the treatment of complicated uri- 2017;215:S28–36.
nary-tract infections. including pyelonephritis: a randomised, double-blind, [72] Senchyna F, Gaur RL, Sandlund J, Truong C, Tremintin G, Kultz D, et al. Diver-
phase 3 trial (ASPECT-cUTI. Lancet 2015;385:1949–56. sity of resistance mechanisms in carbapenem-resistant Enterobacteriaceae at
[47] Escola-Verge L, Pigrau C, Almirante B. Ceftolozane/tazobactam for the treat- a health care system in Northern California, from 2013 to 2016. Diagn Micro-
ment of complicated intra-abdominal and urinary tract infections: current biol Infect Dis 2019;93:250–7.
perspectives and place in therapy. Infect Drug Resist 2019;12:1853–67. [73] Rood IGH, Li Q. Review: Molecular detection of extended spectrum-beta-lac-
[48] Moise PA, Gonzalez M, Alekseeva I, Lopez D, Akrich B, DeRyke CA, et al. Col- tamase- and carbapenemase-producing Enterobacteriaceae in a clinical set-
lective assessment of antimicrobial susceptibility among the most common ting. Diagn Microbiol Infect Dis 2017;89:245–50.
Gram-negative respiratory pathogens driving therapy in the ICU. JAC Antimi- [74] Smith HZ, Kendall B. Carbapenem Resistant Enterobacteriaceae. Treasure Is-
crob Resist 2021;3:dlaa129. land (FL): StatPearls; 2021.
[49] Karaiskos I, Giamarellou H. Carbapenem-Sparing Strategies for ESBL Produc- [75] Codjoe FS, Donkor ES. Carbapenem Resistance: A Review. Med Sci (Basel)
ers: When and How. Antibiotics (Basel) 2020;9:61. 2017;6:1.
[50] Zhanel GG, Golden AR, Zelenitsky S, Wiebe K, Lawrence CK, Adam HJ, [76] Suwantarat N, Carroll KC. Epidemiology and molecular characterization of
et al. Cefiderocol: A Siderophore Cephalosporin with Activity Against Car- multidrug-resistant Gram-negative bacteria in Southeast Asia. Antimicrob Re-
bapenem-Resistant and Multidrug-Resistant Gram-Negative Bacilli. Drugs sist Infect Control 2016;5:15.
2019;79:271–89. [77] Grundmann H, Glasner C, Albiger B, Aanensen DM, Tomlinson CT, Andrase-
[51] Portsmouth S, van Veenhuyzen D, Echols R, Machida M, Ferreira JCA, vic AT, et al. Occurrence of carbapenemase-producing Klebsiella pneumoniae
Ariyasu M, et al. Cefiderocol versus imipenem-cilastatin for the treatment of and Escherichia coli in the European survey of carbapenemase-producing En-
complicated urinary tract infections caused by Gram-negative uropathogens: terobacteriaceae (EuSCAPE): a prospective, multinational study. Lancet Infect
a phase 2, randomised, double-blind, non-inferiority trial. Lancet Infect Dis Dis 2017;17:153–63.
2018;18:1319–28. [78] Guh AY, Bulens SN, Mu Y, Jacob JT, Reno J, Scott J, et al. Epidemiology of
[52] Wunderink RG, Matsunaga Y, Ariyasu M, Clevenbergh P, Echols R, Kaye KS, Carbapenem-Resistant Enterobacteriaceae in 7 US Communities, 2012-2013.
et al. Cefiderocol versus high-dose, extended-infusion meropenem for the JAMA 2015;314:1479–87.
11
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
[79] Wright H, Bonomo RA, Paterson DL. New agents for the treatment of infec- [109] Tumbarello M, Raffaelli F, Giannella M, Mantengoli E, Mularoni A, Venditti M,
tions with Gram-negative bacteria: restoring the miracle or false dawn? Clin et al. Ceftazidime-avibactam use for KPC-Kp infections: a retrospective obser-
Microbiol Infect 2017;23:704–12. vational multicenter study. Clin Infect Dis 2021;73:1664–76.
[80] Poirel L, Jayol A, Nordmann P. Polymyxins: Antibacterial Activity, Suscepti- [110] Jorgensen SCJ, Trinh TD, Zasowski EJ, Lagnf AM, Bhatia S, Melvin SM,
bility Testing, and Resistance Mechanisms Encoded by Plasmids or Chromo- et al. Real-World Experience With Ceftazidime-Avibactam for Multidrug-
somes. Clin Microbiol Rev 2017;30:557–96. Resistant Gram-Negative Bacterial Infections. Open Forum Infect Dis
[81] Doi Y. Treatment Options for Carbapenem-resistant Gram-negative Bacterial 2019;6:ofz522.
Infections. Clin Infect Dis 2019;69:S565–SS75. [111] Escola-Verge L, Pigrau C, Los-Arcos I, Arevalo A, Vinado B, Campany D, et al.
[82] European Medicines Agency. Recommendations to restrict use of fos- Ceftolozane/tazobactam for the treatment of XDR Pseudomonas aeruginosa in-
fomycin antibiotics EMA/670563/2019, 2020. https://www.ema.europa.eu/ fections. Infection 2018;46:461–8.
en/documents/press-release/recommendations-restrict-use-fosfomycin- [112] Munita JM, Aitken SL, Miller WR, Perez F, Rosa R, Shimose LA, et al.
antibiotics_en.pdf [accessed 24 May 2021]. Multicenter Evaluation of Ceftolozane/Tazobactam for Serious Infections
[83] Reyes J, Aguilar AC, Caicedo A. Carbapenem-Resistant Klebsiella pneumoniae: Caused by Carbapenem-Resistant Pseudomonas aeruginosa. Clin Infect Dis
Microbiology Key Points for Clinical Practice. Int J Gen Med 2019;12:437–46. 2017;65:158–61.
[84] Sheu CC, Chang YT, Lin SY, Chen YH, Hsueh PR. Infections Caused by Car- [113] Xipell M, Paredes S, Fresco L, Bodro M, Marco F, Martinez JA, et al. Clin-
bapenem-Resistant Enterobacteriaceae: An Update on Therapeutic Options. ical experience with ceftolozane/tazobactam in patients with serious in-
Front Microbiol 2019;10:80. fections due to resistant Pseudomonas aeruginosa. J Glob Antimicrob Resist
[85] Centers for Disease Control and Prevention Acinetobacter in health- 2018;13:165–70.
care settings. Atlanta, GA: CDC; 2019. https://www.cdc.gov/hai/organisms/ [114] Pogue JM, Kaye KS, Veve MP, Patel TS, Gerlach AT, Davis SL, et al.
acinetobacter.html. [accessed 06 April 2021]. Ceftolozane/Tazobactam vs Polymyxin or Aminoglycoside-based Regimens
[86] Villegas MV, Hartstein AI. Acinetobacter outbreaks, 1977-20 0 0. Infect Control for the Treatment of Drug-resistant Pseudomonas aeruginosa. Clin Infect Dis
Hosp Epidemiol 2003;24:284–95. 2020;71:304–10.
[87] Perez F, Hujer AM, Hujer KM, Decker BK, Rather PN, Bonomo RA. Global [115] Bassetti M, Peghin M, Vena A, Giacobbe DR. Treatment of Infections Due to
challenge of multidrug-resistant Acinetobacter baumannii. Antimicrob Agents MDR Gram-Negative Bacteria. Front Med (Lausanne) 2019;6:74.
Chemother 2007;51:3471–84. [116] Kaye KS, Bhowmick T, Metallidis S, Bleasdale SC, Sagan OS, Stus V, et al.
[88] Fournier PE, Richet H. The epidemiology and control of Acinetobacter bauman- Effect of Meropenem-Vaborbactam vs Piperacillin-Tazobactam on Clinical
nii in health care facilities. Clin Infect Dis 2006;42:692–9. Cure or Improvement and Microbial Eradication in Complicated Urinary
[89] Kurihara MNL, Sales RO, Silva KED, Maciel WG, Simionatto S. Multidrug-resis- Tract Infection: The TANGO I Randomized Clinical Trial. JAMA
tant Acinetobacter baumannii outbreaks: a global problem in healthcare set- 2018;319:788–99.
tings. Rev Soc Bras Med Trop 2020;53:e20200248. [117] Motsch J, Murta de Oliveira C, Stus V, Koksal I, Lyulko O, Boucher HW, et al.
[90] Peleg AY, Seifert H, Paterson DL. Acinetobacter baumannii: emergence of a suc- RESTORE-IMI 1: A Multicenter, Randomized, Double-blind Trial Comparing
cessful pathogen. Clin Microbiol Rev 2008;21:538–82. Efficacy and Safety of Imipenem/Relebactam vs Colistin Plus Imipenem in
[91] Queenan AM, Bush K. Carbapenemases: the versatile beta-lactamases. Clin Patients With Imipenem-nonsusceptible Bacterial Infections. Clin Infect Dis
Microbiol Rev 2007;20:440–58. 2020;70:1799–808.
[92] Piperaki ET, Tzouvelekis LS, Miriagou V, Daikos GL. Carbapenem-resistant [118] Sims M, Mariyanovski V, McLeroth P, Akers W, Lee YC, Brown ML, et al.
Acinetobacter baumannii: in pursuit of an effective treatment. Clin Microbiol Prospective, randomized, double-blind, Phase 2 dose-ranging study com-
Infect 2019;25:951–7. paring efficacy and safety of imipenem/cilastatin plus relebactam with
[93] Isler B, Doi Y, Bonomo RA, Paterson DL. New Treatment Options against imipenem/cilastatin alone in patients with complicated urinary tract infec-
Carbapenem-Resistant Acinetobacter baumannii Infections. Antimicrob Agents tions. J Antimicrob Chemother 2017;72:2616–26.
Chemother 2019;63:e01110–18. [119] Morris S, Cerceo E. Trends, Epidemiology, and Management of Multi-Drug Re-
[94] Djahmi N, Dunyach-Remy C, Pantel A, Dekhil M, Sotto A, Lavigne JP. Epidemi- sistant Gram-Negative Bacterial Infections in the Hospitalized Setting. Antibi-
ology of carbapenemase-producing Enterobacteriaceae and Acinetobacter bau- otics (Basel) 2020;9:196.
mannii in Mediterranean countries. Biomed Res Int 2014;2014:305784. [120] US Department of Health and Human Services Centre for Disease Control.
[95] Terahara F, Nishiura H. Carbapenem-resistant Pseudomonas aeruginosa Multidrug-resistant Pseudomonas aeruginosa AR Threats Report, 2019. https://
and carbapenem use in Japan: an ecological study. J Int Med Res www.cdc.gov/drugresistance/pdf/threats-report/pseudomonas-aeruginosa-
2019;47:4711–22. 508.pdf [accessed 30 October 2021].
[96] Yoon EJ, Jeong SH. Mobile Carbapenemase Genes in Pseudomonas aeruginosa. [121] Jurado-Martin I, Sainz-Mejias M, McClean S. Pseudomonas aeruginosa: An Au-
Front Microbiol 2021;12:614058. dacious Pathogen with an Adaptable Arsenal of Virulence Factors. Int J Mol
[97] Kazmierczak KM, Rabine S, Hackel M, McLaughlin RE, Biedenbach DJ, Bouch- Sci 2021;22:3128.
illon SK, et al. Multiyear, Multinational Survey of the Incidence and Global [122] Allocati N, Masulli M, Alexeyev MF, Di Ilio C. Escherichia coli in Europe: an
Distribution of Metallo-beta-Lactamase-Producing Enterobacteriaceae and overview. Int J Environ Res Public Health 2013;10:6235–54.
Pseudomonas aeruginosa. Antimicrob Agents Chemother 2016;60:1067–78. [123] Kaye KS, Pogue JM. Infections Caused by Resistant Gram-Negative Bacteria:
[98] Huband MD, Castanheira M, Flamm RK, Farrell DJ, Jones RN, Sader HS. In Vitro Epidemiology and Management. Pharmacotherapy 2015;35:949–62.
Activity of Ceftazidime-Avibactam against Contemporary Pseudomonas aerug- [124] Oliveira J, Reygaert WC. Gram Negative Bacteria. Treasure Island, Florida: Stat-
inosa Isolates from U.S. Medical Centers by Census Region, 2014. Antimicrob Pearls; 2021.
Agents Chemother 2016;60:2537–41. [125] Sirot J, Chanal C, Petit A, Sirot D, Labia R, Gerbaud G. Klebsiella pneumoniae
[99] Sader HS, Farrell DJ, Flamm RK, Jones RN. Antimicrobial susceptibility of and other Enterobacteriaceae producing novel plasmid-mediated beta-lacta-
Gram-negative organisms isolated from patients hospitalised with pneumonia mases markedly active against third-generation cephalosporins: epidemio-
in US and European hospitals: results from the SENTRY Antimicrobial Surveil- logic studies. Rev Infect Dis 1988;10:850–9.
lance Program, 2009-2012. Int J Antimicrob Agents 2014;43:328–34. [126] Tumbarello M, Trecarichi EM, De Rosa FG, Giannella M, Giacobbe DR, Bas-
[100] Dortet L, Poirel L, Nordmann P. Worldwide dissemination of the ND- setti M, et al. Infections caused by KPC-producing Klebsiella pneumoniae:
M-type carbapenemases in Gram-negative bacteria. Biomed Res Int differences in therapy and mortality in a multicentre study. J Antimicrob
2014;2014:249856. Chemother 2015;70:2133–43.
[101] Zhu Y, Chen J, Shen H, Chen Z, Yang QW, Zhu J, et al. Emer- [127] Davin-Regli A, Pages JM. Enterobacter aerogenes and Enterobacter cloacae; ver-
gence of Ceftazidime- and Avibactam-Resistant Klebsiella pneumoniae satile bacterial pathogens confronting antibiotic treatment. Front Microbiol
Carbapenemase-Producing Pseudomonas aeruginosa in China. mSystems 2015;6:392.
2021;6:e0078721. [128] Kotloff KL, Riddle MS, Platts-Mills JA, Pavlinac P, Zaidi AKM. Shigellosis.
[102] Zhang Y, Chen XL, Huang AW, Liu SL, Liu WJ, Zhang N, et al. Mortality Lancet 2018;391:801–12.
attributable to carbapenem-resistant Pseudomonas aeruginosa bacteremia: a [129] Puzari M, Sharma M, Chetia P. Emergence of antibiotic resistant Shigella
meta-analysis of cohort studies. Emerg Microbes Infect 2016;5:e27. species: A matter of concern. J Infect Public Health 2018;11:451–4.
[103] Bassetti M, Vena A, Croxatto A, Righi E, Guery B. How to manage Pseudomonas [130] US Department of Health and Human Services Centers for Desease Control
aeruginosa infections. Drugs Context 2018;7:212527. and Prevention. Drug-resistant Salmonella serotype typhi, 2019. https://www.
[104] Driscoll JA, Brody SL, Kollef MH. The epidemiology, pathogenesis and treat- cdc.gov/drugresistance/pdf/threats-report/salmonella-typhi-508.pdf [accessed
ment of Pseudomonas aeruginosa infections. Drugs 2007;67:351–68. 01 November 2021].
[105] Khadem T, Stevens V, Holt K, Hoffmann C, Dumyati G, Brown J. Risk factors [131] US Department of Health and Human Services Centers for Disease Control
for carbapenem-nonsusceptible Pseudomonas aeruginosa: Case-control study. and Prevention. Drug-resistant nontyphoidal Salmonella, 2019. https://www.
Diagn Microbiol Infect Dis 2017;89:146–50. cdc.gov/drugresistance/pdf/threats-report/nt-salmonella-508.pdf [accessed 01
[106] Zhanel GG, Wiebe R, Dilay L, Thomson K, Rubinstein E, Hoban DJ, et al. Com- November 2021].
parative review of the carbapenems. Drugs 2007;67:1027–52. [132] Karkey A, Thwaites GE, Baker S. The evolution of antimicrobial resistance in
[107] Zavascki AP, Carvalhaes CG, Picao RC, Gales AC. Multidrug-resistant Pseu- Salmonella typhi. Curr Opin Gastroenterol 2018;34:25–30.
domonas aeruginosa and Acinetobacter baumannii: resistance mechanisms and [133] McDermott PF, Zhao S, Tate H. Antimicrobial Resistance in Nontyphoidal
implications for therapy. Expert Rev Anti Infect Ther 2010;8:71–93. Salmonella. Microbiol Spectr 2018;6(4).
[108] Horcajada JP, Montero M, Oliver A, Sorli L, Luque S, Gomez-Zorrilla S, [134] Armbruster CE, Mobley HLT, Pearson MM. Pathogenesis of Proteus mirabilis
et al. Epidemiology and Treatment of Multidrug-Resistant and Extensively Infection. EcoSal Plus 2018;8(1).
Drug-Resistant Pseudomonas aeruginosa Infections. Clin Microbiol Rev 2019;32 [135] Hamilton AL, Kamm MA, Ng SC, Morrison M. Proteus spp. as Putative Gas-
e0 0 031-19. trointestinal Pathogens. Clin Microbiol Rev 2018;31 e0 0 085-17.
12
S.S. Kanj, M. Bassetti, P. Kiratisin et al. International Journal of Antimicrobial Agents 60 (2022) 106633
[136] Hawkey PM, Warren RE, Livermore DM, McNulty CAM, Enoch DA, Otter JA, [140] Shen Z, Wang Y, Zhang Q, Shen J. Antimicrobial Resistance in Campylobacter
et al. Treatment of infections caused by multidrug-resistant Gram-nega- spp. Microbiol Spectr 2018;6(2).
tive bacteria: report of the British Society for Antimicrobial Chemother- [141] Su PY, Huang AH, Lai CH, Lin HF, Lin TM, Ho CH. Extensively drug-resistant
apy/Healthcare Infection Society/British Infection Association Joint Working Haemophilus influenzae - emergence, epidemiology, risk factors, and regimen.
Party. J Antimicrob Chemother 2018;73:iii2–iii78. BMC Microbiol 2020;20:102.
[137] Moradigaravand D, Boinett CJ, Martin V, Peacock SJ, Parkhill J. Re- [142] Mlynarczyk-Bonikowska B, Majewska A, Malejczyk M, Mlynarczyk G, Majew-
cent independent emergence of multiple multidrug-resistant Serratia ski S. Multiresistant Neisseria gonorrhoeae: a new threat in second decade of
marcescens clones within the United Kingdom and Ireland. Genome Res the XXI century. Med Microbiol Immunol 2020;209:95–108.
2016;26:1101–9. [143] Wong D, van Duin D. Novel Beta-Lactamase Inhibitors: Unlocking Their Po-
[138] Stock I, Grueger T, Wiedemann B. Natural antibiotic susceptibility of tential in Therapy. Drugs 2017;77:615–28.
strains of Serratia marcescens and the S. liquefaciens complex: S. liquefa- [144] van Duin D, Bonomo RA. Ceftazidime/Avibactam and Ceftolozane/Tazobactam:
ciens sensu stricto, S. proteamaculans and S. grimesii. Int J Antimicrob Agents Second-generation β -Lactam/β -Lactamase Inhibitor Combinations. Clin Infect
2003;22:35–47. Dis 2016;63:234–41.
[139] Matsumoto H, Shiotani A, Graham DY. Current and Future Treatment of Heli- [145] El-Lababidi RM, Rizk JG. Cefiderocol: A Siderophore Cephalosporin. Ann Phar-
cobacter pylori Infections. Adv Exp Med Biol 2019;1149:211–25. macother 2020;54:1215–31.
13