Aac 00256-22

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

PERSPECTIVE

New Perspectives on Antimicrobial Agents: Imipenem-


Relebactam
J. Nicholas O’Donnell,a Thomas P. Lodisea

Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
a

ABSTRACT Imipenem (IMI)/cilastatin/relebactam (REL) (I/R) is a novel b -lactam/


b -lactamase inhibitor combination with expanded microbiologic activity against
carbapenem-resistant non-Morganellaceae Enterobacterales (CR-NME) and difficult-
to-treat (DTR) Pseudomonas aeruginosa. Relebactam, a bicyclic diazabicyclooctane,
has no direct antimicrobial activity but provides reliable inhibition of many Ambler
class A and class C enzymes. It is currently approved for the treatment of adult
patients with hospital-acquired bacterial pneumonia and ventilator-associated bac-
terial pneumonia (HABP/VABP) and those with complicated urinary tract infections
(cUTIs) and complicated intra-abdominal infections (cIAIs) when limited or no alter-
native treatments are available. Given the number of recently approved b -lactams
with expanded activity against highly resistant Gram-negative pathogens, this
review summarizes the published literature on I/R, with a focus on its similar and
distinguishing characteristics relative to those of other recently approved agents.
Overall, available data support its use for the treatment of patients with HABP/
VABP, cUTI, and cIAI due to CR-NME and DTR P. aeruginosa. Data indicate that I/R
retains some activity against CR-NME and DTR P. aeruginosa isolates that are resist-
ant to the newer b -lactams and vice versa, suggesting that susceptibility testing
be performed for all the newer agents to determine optimal treatment options for
patients with CR-NME and DTR P. aeruginosa infections. Further comparative PK/PD
and clinical studies are warranted to determine the optimal role of I/R, alone and
in combination, for the treatment of patients with highly resistant Gram-negative
infections. Until further data are available, I/R is a potential treatment for patients
with CR-NME and DTR P. aeruginosa infections when the benefits outweigh the
risks.

KEYWORDS imipenem-relebactam, multidrug resistance, pharmacokinetics,


pharmacodynamics, Enterobacterales, Pseudomonas aeruginosa, clinical trials

mipenem (IMI)/cilastatin/relebactam (REL) (I/R) is a novel b -lactam/ b -lactamase inhibi-


I tor (BL/BLI) combination with expanded microbiologic activity against multidrug-resist-
ant Gram-negative pathogens, including carbapenem-resistant non-Morganellaceae
Copyright © 2022 American Society for
Enterobacterales (CR-NME) and difficult-to-treat (DTR) Pseudomonas aeruginosa. Relebactam,
Microbiology. All Rights Reserved.
a bicyclic diazabicyclooctane, has no direct antimicrobial activity but provides reliable inhibi- Address correspondence to Thomas P. Lodise,
tion of many Ambler class A and class C enzymes (1). For example, relebactam effectively Thomas.lodise@acphs.edu.
inhibits Klebsiella pneumoniae carbapenemase (KPC) and Pseudomonas-derived cephalospor- The authors declare a conflict of interest. T.P.L.
has received consultant honoraria and
inases (PDCs), which are Ambler class A and C enzymes, respectively. Relebactam differs
investigator-initiated grant support from Merck
from other diazabicyclooctane BLIs in that it carries a positively charged side chain that has & Co. J.N.O. has received investigator-initiated
been shown to limit its efflux (2). The addition of relebactam to IMI restores its in vitro activity grant support from Merck & Co.
against a substantial number of CR-NME and DTR P. aeruginosa isolates. Consistent with Received 16 February 2022
Returned for modification 28 March 2022
most new b -lactams except cefiderocol, I/R does not have expanded activity against Accepted 20 May 2022
Acinetobacter baumannii or metallo-b -lactamase (MBL)-producing Gram-negative bacteria Published 21 June 2022
relative to IMI alone (3, 4).

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 1


Perspective Antimicrobial Agents and Chemotherapy

Imipenem/cilastatin/relebactam is currently approved by the Food and Drug


Administration (FDA) for the treatment of adult patients with hospital-acquired bac-
terial pneumonia and ventilator-associated bacterial pneumonia (HABP/VABP), com-
plicated urinary tract infections (cUTI), including pyelonephritis, or complicated intra-
abdominal infections (cIAI) who have limited or no alternative treatment options (5).
In the European Union, it is approved in adult patients for the treatment of HABP/
VABP, suspected or documented bacteremia due to HABP/VABP, and infections due
to aerobic Gram-negative bacteria with limited treatment options (6). The United
States- and European Union-approved I/R dose is 500 mg/250 mg infused overed 30
min every 6 h, with renal dose adjustments for patients with creatinine clearances of
,60 mL/min (5, 6). The current FDA and Clinical and Laboratory Standards Institute
(CLSI) I/R susceptibility breakpoints for P. aeruginosa and Enterobacterales are #2/4 mg/L
and #1/4 mg/L, respectively, while the EUCAST susceptibility breakpoints for both P. aer-
uginosa and Enterobacterales are #2/4 mg/L (7, 8).
Given the number of recently approved b -lactams with activity against highly re-
sistant Gram-negative pathogens, this review on I/R summarizes the published litera-
ture on its in vitro susceptibility against highly resistant Gram-negative isolates across
surveillance programs, mechanisms of resistance, pharmacokinetics (PK), pharmacoki-
netics/pharmacodynamics (PK/PD) in preclinical models of infections, and clinical data.
As part of the review, we focus on the similar and distinguishing characteristics of I/R
relative to those of other recently approved b -lactams (i.e., BL/BLIs and cefiderocol).
We also discuss its overlapping and differential potential roles in therapy within the
current antibiotic landscape. Finally, we provide some considerations for future studies
to help better define the role of I/R in the treatment of patients with highly resistant
infections relative to those of other recently approved b -lactams.

MICROBIOLOGY
In vitro activity and mechanisms of resistance. The addition of REL to IMI expands
the in vitro spectrum of activity of I/R to include NME that produce class A b -lactamases,
including KPC and several serine extended-spectrum b -lactamases, and class C b -lacta-
mases, including the PDCs and AmpC enzymes (Table 1) (2, 9). It is largely unaffected by
porin channel-mediated resistance due to OprD loss or efflux pump-mediated resistance
(eg, MexAB, MexCD, MexXY) in P. aeruginosa (Table 2). Of note, I/R also appears to retain in
vitro activity against isolates with KPC-3 or PDC mutations that result in resistance to cefta-
zidime-avibactam (CZA) or ceftolozane-tazobactam (C/T). Resistance to I/R has been
observed in isolates expressing OXA-48 or MBLs and with combinations of porin altera-
tions and hyperexpression of carbapenemase enzymes. As with IMI alone, activity against
Acinetobacter sp. and Morganellaceae (i.e., Proteus spp., Providencia spp., and Morganella
spp.) is limited (2, 9). Among large surveillance studies, I/R typically exhibits activity against
.90% of NME and P. aeruginosa isolates (10–14). Most notably, REL provides significant
restoration of IMI activity against KPC-producing NME, lowering the MIC90 up to 16-fold (2,
9). It is also highly active against DTR P. aeruginosa isolates (15).
While I/R is highly active against most CR-NME and multidrug-resistant (MDR) P. aerugi-
nosa isolates, it does not have activity against MBL-producing isolates (e.g., NDM, IMP,
VIM), Serratia marcescens isolates that express SME carbapenemases, VEB-type extended-
spectrum b -lactamase (ESBL)-producing P. aeruginosa, and GES-like carbapenemase-pro-
ducing P. aeruginosa (2, 16–18). Relebactam also exhibits variable, but limited, inhibition of
OXA-48-like enzymes (19–22). Among isolates exhibiting I/R resistance, production of
OXA-48-like enzymes is relatively common, with the largest study to date suggesting that
only 16% (22/137) of isolates with OXA-48 production and without MBL production were
susceptible to I/R (21). Among K. pneumoniae isolates, disruption of OmpK36 combined
with production of KPC enzymes has also been associated with I/R resistance as have porin
alterations in combination with increased blaKPC copy number (i.e., increased KPC expres-
sion) (4, 23, 24). Data are more limited on the effect of disruption of Enterobacter species
porin OmpC, which serves functions similar to those of OmpK36, on I/R susceptibility.

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 2


TABLE 1 Susceptibility of carbapenem-nonsusceptible Enterobacterales to imipenem/relebactam and rate of metallo- b -lactamase productiona
Susceptibility (%)
CLSI or EUCAST %
Perspective

Study Database, location (yr[s]) breakpoints Population (n isolates) % MBL Morganellaceae IMI MERd I/R CZAd M/V COL/PMBd AMKd
Maraki 2022 (25) Greece (2017–2020) EUCAST CR-K. pneumoniae (266) 17.3% 0.0% 0.0% 0.0% 76.7% 79.7% 80.1% 65.8% NRd
Bail 2022 (37) Brazil (2013–2020) CLSI IMI/MER-R 0.0% 4.0% 0.0% 8.0% 96.0% NR NR 65.0% 67.0%
Enterobacterales (300)
Kuo 2021 (27) Taiwan (2012–2018) CLSI IMI-NS E. coli (17) 7.2%b 0.0% 0.0% 52.9% 70.6% 70.6% 94.1% 94.1% 94.1%

July 2022 Volume 66 Issue 7


IMI-NS K. pneumoniae 0.0% 0.0% 27.6% 76.1% 90.8% 81.6% 87.1% 76.7%
(163)
Bhagwat 2021 (29) Greece (2014–2018) EUCAST Enterobacterales (563) 33.0% 6.6% 9.9% 10.3% 61.0% 59.8% NR 47.4% 36.9%
Yang 2021 (83) Taiwan (2012–2015) CLSI Carbapenem-NS E. coli 4.8% 0.0% 29.3% 28.7% 87.8% NR NR NR NR
(188)
Carbapenem-NS K. 3.0% 0.0% 18.9% 23.1% 82.2% NR NR NR NR
pneumoniae (472)
Vazques-Ucha 2021 (19) Spain (2018) EUCAST CPE (401) 14.0% 0.0% 72.3% 68.8% 85.8% 83.8% NR 86.5% 75.8%
Yang 2020 (84) SMART, China (2015–2018) CLSI IMI-NS NR 0.0% 0.0% NR 66.3% NR NR 83.6% 50.9%
Enterobacteriaceae
(1,165)
Karlowsky 2020 (30) Global (2014–2016) CLSI Carbapenem-NS 21.0% 0.0% NR 4.4% 64.1% 77.5% NR 78.1% NR
Enterobacterales
(1,018)
Karlowsky 2021 (14) Global (2015–2017) CLSI KPC-producing NME 0.2% 0.0% 0.0% NR 96.2% NR NR 74.8% 71.5%
(1,081)
Nelson 2020 (32) Global (2001–2017) CLSI CPE (non-MBL) (374) 0.0% 0.3% 2.9% 24.6% 69.5% 90.6% 76.7% NR NR
Non-CP-EBE (50) 0.0% 0.0% 20.0% 51.2% 64.0% 86.0% 74.0% NR NR
Karlowsky 2021 (15) SMART, USA (2015–2017) CLSI DTR EBE (102) NR 1.0% 0.0% 0.0% 82.4% NR NR 84.3% 79.4%
Johnston 2020 (20) USA (2002–2017) CLSI CR E. coli (203) 7.9% 0.0% 44.0% 75.0% 89.0% NR NR 99.0% 80.0%
Lob 2020 (21) Europe (2015–2017) EUCAST IMI-NS NME (474) 30.8% 0.0% 0.0% NR 42.4% NR NR 67.1% 54.0%
Karlowsky 2020 (17) SMART, USA (2015–2017) CLSI IMI-R NME (170) 0.0% 0.0% 0.0% NR 66.4% NR NR NR NR
Karlowsky 2020 (85) SMART, USA (2015–2017) CLSI IMI-NS NME (154) 1.3% 0.0% 0.0% NR 77.9% NR NR 61.0% 88.3%
Carpenter 2019 (33) USA (2013–2017) CLSI IMI-NS CPE isolates (200) 0.0% 0.0% 0.0% NR 96.5% 100.0% NR 46.1%c NR
Galani 2019 (34) Greece (2014–2016) EUCAST KPC-producing K. 0.0% 0.0% 0.0% 0.3% 98.0% 99.7% NR 60.7% NR
pneumoniae (295)
Karlowsky 2018 (9) SMART, USA (2016) CLSI IMI-R NME (130) NR 0.0% 0.0% 0.0% 78.5% NR NR 41.5% 97.7%
Karlowsky 2018 (86) SMART, Europe (2015) EUCAST IMI-NS K. pneumoniae 22.9% 0.0% 0.0% NR 54.2% NR NR NR 41.9%
(179)
IMI-NS Enterobacter spp. 73.5% 0.0% 0.0% NR 26.5% NR NR NR 88.2%
(34)
Lapuebla 2015 (87) USA, (2013–2014) CLSI KPC-producing K. 0.0% 0.0% 9.0% NR 97.0% NR NR NR NR
pneumoniae (111)
aTable limited to studies with .100 carbapenem-nonsusceptible isolates.
bMBL-production reported for E. coli and K. pneumoniae together.
cTested against 180 non-Serratia species isolates.

dCOL, colistin; PMB, polymyxin B; AMK, amikacin; MER, meropenem; NR, not reported.

10.1128/aac.00256-22
Antimicrobial Agents and Chemotherapy

3
TABLE 2 P. aeruginosa susceptibility to imipenem/relebactam and prevalence of metallo- b -lactamase production
Susceptibility (%)
Perspective

Study Database, location (yr[s]) Phenotype (n isolates) % MBL IMI MER I/R C/T CAZ CZA M/V COL/PMB AMK
Bail 2022 (37) Brazil (2013–2020) IMI/MER-R (carbapenemase 0% 2% 6% 63% 80% 53% NR NR 98% 64%
non-producing) (229)
Sader 2021 (10) SENTRY Global (2020) MER-NS (54) NR NR 0% 88.9% 90.6% NR 92.6% 61.1% NR NR
IMI-R (48) NR 0% NR 87.5% 91.5% NR 93.8% 56.3% NR NR

July 2022 Volume 66 Issue 7


Sader 2022 (11) SENTRY, Europe (2020) MER-R (70) NR NR 0% 58.6% 57.1% NR 69.9% 5.7% NR NR
IMI-R (155) NR 0% NR 78.7% 81.9% NR 87% 59.4% NR NR
Zhang 2021 (3) SMART, China (2015–2018) IMI-NS (835) NR 0% NR 64.4% NR% NR NR NR NR NR
MDR (835) NR 25.2% NR 65.8% NR NR NR NR NR NR
Lob 2021 (13) SMART, USA (2018–2019) IMI-R (43) NR 0% 46.5% 79.1% NR 72.1% NR NR NR 95.4
Lob 2021 (12) SMART, USA (2017–2019) DTR ICU (72) NR 0% 0% 54.2% 61.1% 0% NR NR NR 83.3%
DTR non-ICU (56) NR 0% 0% 57.1% 69.6% 0% NR NR NR 82.1%
Kuo 2021 (27) Taiwan (2018) IMI-NS (81) 4.93% 0% 28.4% 85.2% NR 43.2% 81.5% 42% 69.3% 95%
Bhagwat 2021 (29) Greece (2014–2018) IMI-NS (172) 64% 0% 9.9% 28.5% 25.6% 0% 33.7% NR 64.5% 34.9%
Karlowsky 2020 (30) Global (2014–2016) MDR (262) 35.90% 0% 1.9% 17.2% 21.8% NR 26.3% NR 99.6% NR
Karlowsky 2021 (14) SMART, Global (2015–2017) Pediatric MDR (281) 15.3% 28.5% NR 70.5% NR 19.9% NR NR 98.6 72.2%
Adult MDR (4,927) 13.7% 28% NR 68.3% NR 23.4% NR NR 98.9% 72%
Karlowski 2021 (15) SMART, USA (2015–2017) DTR (230) NR 0% 0% 62.2% 67.5% 0% NR NR 98.7% 84.8%
Lob 2020 (21) Europe (2015–2017) IMI-NS (469) 18.30% 0% NR 74.4% NR 35.6% NR NR 64.6% 99.6%
Karlowsky 2020 (17) SMART, USA (2015–2017) IMI-R (219) NR 0% NR 77.2% NR NR NR NR NR NR
Karlowsky 2020 (85) SMART, USA (2015–2017) IMI-NS (193) NR 0% NR 85% NR NR NR NR NR NR
Young 2019 (2) SMART (2009–2016) IMI-R (3,747) 13.50% 0% NR 68% NR NR NR NR NR NR
Karlowski 2019 (88) SMART, USA (2015–2017) IMI-NS (766) NR 0% NR 78.3% NR NR NR NR NR NR
Asempa 2019 (89) USA (2017–2018) Carbapenem-NS (189) NR 6.4% 22.2% 75.7% NR 58.2% 84.7% NR NR NR
Lob 2018 (90) SMART, Europe (2015–2017) IMI-NS (1,668) NR 0% NR 75.2% NR NR NR NR NR NR
Mushtaq 2021 (18) England (2012–2019) VEB-producing (97) 0% 3.1% NR 10.3% 1% 0% 2.1% NR NR NR
GES-5 (carbapenemase) 0% 0% NR 0% 8.1% 8.1% 94.6% NR NR NR
producing (37)

10.1128/aac.00256-22
Antimicrobial Agents and Chemotherapy

4
Perspective Antimicrobial Agents and Chemotherapy

Finally, I/R exhibited very limited in vitro activity against P. aeruginosa isolates producing a
VEB-type ESBL (18).
Comparisons of in vitro susceptibility between I/R and CZA have been conducted in
several surveillance studies (10, 11, 18, 19, 22, 23, 25–34). Typically, susceptibility results
are similar, or slightly favor CZA. Differences in activity against Enterobacterales are typ-
ically ,10% of tested isolates and are mostly driven by the prevalence of OXA-48 pro-
duction, Morganellaceae, and Serratia marcescens. To date, I/R does appear unaffected
by KPC-3 mutations that render CZA ineffective (4, 24). While the exact mechanism of
these mutations’ effect on avibactam are unclear, it has been suggested that they may
result in increased affinity for ceftazidime and decreased affinity for avibactam. It is
also worth noting that these mutations are also associated with significant decreases
in carbapenem MICs, often with restoration of carbapenem susceptibility as a solitary
mechanism (35, 36). It appears, however, that these mutations in combination with
porin alterations in K. pneumoniae may result in increases in MICs with M/V but not
with I/R (24). Activity against DTR-P. aeruginosa is also similar between these agents,
though I/R does not appear to exhibit activity against GES-5-producing P. aeruginosa
whereas CZA does (18).
Comparisons with meropenem-vaborbactam (M/V) and C/T are more limited than
CZA, and scant comparative susceptibility data are available with cefiderocol (10, 11,
15, 18, 22, 24–30, 32). Overall, M/V appears to exhibit activity against NME similar to
that of I/R, though M/V may have improved activity against OXA-48 producers relative
to that of I/R (10, 11, 22, 25–27, 32). I/R exhibits significantly increased activity against
P. aeruginosa relative to that of M/V. I/R also appears to exhibit activity against KPC-
producing K. pneumoniae isolates that exhibit M/V resistance via KPC production com-
bined with OmpK35 and OmpK36 mutations (24). Against P. aeruginosa, I/R and C/T
exhibited similar susceptibility profiles (10, 11, 15, 18, 22, 28–30, 37). One study of non-
carbapenemase-producing P. aeruginosa from Brazil, however, identified activity of C/T
substantially higher than that of I/R (80 versus 63%), though the cause of this discrep-
ancy is unknown (37). Of note, some DTR-P. aeruginosa isolates that develop resistance
to C/T also develop cross-resistance to CZA (28). It appears that these mutations, often
driven by mutations in PDCs, have a limited effect on I/R susceptibility.

PHARMACOKINETICS
Most published data on the PK of I/R were ascertained in phase I and II studies of
noninfected patients. In phase I and II studies, I/R exhibited linear PK with a dose-inde-
pendent terminal half-life (t1/2) ranging from 1.35 to 1.85 h (38). Data from single- and
multiple-dose studies indicated that I/R is predominately cleared by the kidneys (IMI,
;200 mL/min; REL, ;150 mL/min) with the fraction excreted in urine ranging from
94.7% to 100%. There was no appreciable accumulation with multiple doses, and the
observed apparent volume of distribution (;20 L) indicated that I/R largely distributed
in extracellular fluid. No drug-drug interactions between IMI and REL were observed,
and their PK profiles were similar, supporting their use as coadministered agents (38).
In a phase 1 PK study of patients with impaired renal function, including patients on
hemodialysis (HD), plasma exposures of IMI and REL increased as the estimated glo-
merular filtration rate (eGFR) decreased, and all 3 compounds exhibited comparable PK
alterations in patients with mild, moderate, and severe renal impairment and were sim-
ilarly removed by HD (39). These data were used, in part, to derive the current renal
dosing recommendations for patients with renal impairment, including HD (5). In
healthy subjects, the area under the curve from zero to infinity (AUC0–1) in epithelial
lining fluid (ELF) for both IMI and REL was ;55% of free plasma AUCs (40).

PHARMACOKINETICS AND PHARMACODYNAMICS


Overview. The PK/PD profile of I/R has been informed in 8 preclinical models of infec-
tions: neutropenic murine disseminated and pulmonary infection models (K. pneumoniae
and P. aeruginosa) (41), neutropenic mouse thigh infection model (K. pneumoniae and P.

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 5


Perspective Antimicrobial Agents and Chemotherapy

aeruginosa) (42, 43), in vitro one-compartment infection model (K. pneumoniae, P. aerugi-
nosa, and Escherichia coli) (44, 45), and hollow fiber infection model (HFIM; K. pneumoniae
and P. aeruginosa) (46–48). A mix of IMI-resistant and IMI-susceptible strains were eval-
uated across the preclinical PK/PD studies. Limited information on the susceptibility of the
evaluated isolates against newer b -lactams was included in the studies. In the two studies
that provided data, most isolates were susceptible to ceftazidime-avibactam (CZA) and cef-
tolozane-tazobactam (C/T) (43, 45). Standard starting inocula were evaluated across all
studies (49). All preclinical PK/PD studies were 24 h except two in vitro PK/PD model pub-
lished studies, which evaluated durations of 3, 7, and 14 days (44, 48). Across most preclini-
cal PK/PD studies, the FDA-European Union-approved I/R dose was simulated using the I/R
free maximum concentration (fCmax) and half-life (t1/2) observed in healthy human volun-
teers (38), and none include a newer b -lactam as a comparator. Given that the dosing (50)
and PK/PD profile (51, 52) of IMI are well established, humanized REL dose escalation and
fractionation studies (46, 47) were performed (REL doses were varied from 0 to 3,000 mg
every 1.5 to 24 h) to ascertain the REL in the presence of IMI PK/PD indices for a 1- to 2-log
kill, the PK/PD targets associated with clinical outcomes (53).
PK/PD targets for REL in the presence of IMI in preclinical PK/PD models of
infections. Two PK/PD targets for REL in the presence of IMI have been identified across
the preclinical PK/PD infection models of infection (46, 47). In the 24-h dose-escalation and
fractionation translational semimechanistic HFIM of IMI-resistant P. aeruginosa (n = 5 iso-
lates with I/R MICs of 2/4 to 16/4 mg/L) study by Bhagunde and colleagues, the PK/PD
index was found to be the fAUC0–24/MIC, and the magnitude of fAUC0–24/MIC for stasis,
1-log, and 2-log kill was 2.7, 4.7, and 7.5, respectively (46). Most patient fAUC0–24/MIC at a
250 mg REL dose exceeded 7.5, suggesting that the I/R 500 mg/250 mg dose every 6 h
was sufficient to provide .2-log kill for IMI-resistant P. aeruginosa isolates with potentiated
I/R MIC (the MIC of IMI in the presence of REL 4 mg/L) of #4 mg/L. The findings from this
HFIM study aligned with an earlier mouse thigh P. aeruginosa infection model study, which
suggested that the PK/PD-linked variable for REL in the presence of IMI was fAUC (42). It is
consistent with the PK/PD driver for meropenem-vaborbactam (M/V; vaborbactam fAUC:
MIC ratio of 18 to 35 associated with 1 log10 CFU/mL killing) (54) but differs from the
PK/PD driver (i.e., percent time above a threshold) for CZA and aztreonam-AVI (ATM-AVI)
(55, 56). Further study is needed, but the differences in PK/PD drivers could be due to
altered enzyme-binding kinetics; REL is an irreversible BLI while AVI is a reversible inhibitor.
As an irreversible BLI, degree of b -lactamase inhibition is likely a function of the magni-
tude of exposure over time while maintenance of a critical concentration would be more
critical for a reversible inhibitor like AVI (57).
A second dose-escalation HFIM of IMI-resistant P. aeruginosa and K. pneumoniae
that was 72 h in duration also found an association between fAUC/MIC and bacterial
killing, and the fAUC/MIC ratios required to achieve static, 1-log, and 2-log drops using
the potentiated I/R MIC were 8.2, 12, and 18, respectively (47). However, the percent-
age of time that I/R exceeds the dynamic MIC (%T . MICdynamic) was found to be a
more informative PK/PD driver. Data from checkboard experiments performed as part
of the study indicated that the potentiated I/R MIC of an isolate changes in a concen-
tration-dependent manner with increasing concentrations of REL, and the MICdynamic
reflects the potentiated I/R MIC at a given point in time based on the REL concentra-
tion present (58). Employing a maximum-effect (Emax) model to describe the relation-
ship between the potentiated I/R MIC and the REL concentration, the authors found
that the clinically administered dose of I/R (500 mg/250 mg intravenously [i.v.] every
6 h as a 0.5-h infusion) demonstrated a 65.5% fT . MICdynamic, which was well above
the identified 48- and 72-h fT . MICdynamic thresholds for sustained $2-log killing and
regrowth suppression.
Probability of target attainment profiles of United States- and European
Union-approved I/R dosing regimens. The probability of target attainment (PTA) pro-
file of I/R was evaluated as part of the population PK study that included PK data from
patients in the multicenter, randomized, double-blind trial comparing efficacy and
safety of imipenem/relebactam (RESTORE-IMI-1 and RESTORE-IMI-2) randomized

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 6


Perspective Antimicrobial Agents and Chemotherapy

clinical trials (RCTs) (59). Using data from 12 completed phase I to III clinical studies
(1,197 total patients with quantifiable plasma PK samples), a two-compartment zero-
order i.v. infusion model with first-order linear elimination with a proportional error
term was identified as the optimal base PK model (59). Covariates retained in the final
REL PK model included pneumonia and creatinine clearance (CLCR), expressed as a
power function, on CL and ventilation status, pneumonia, and total weight (power
function) on apparent central volume (V1). For IMI, significant covariates included
pneumonia, CLCR (power function), and body weight (power function) on CL and venti-
lation status, pneumonia, and total body weight (power function) on V1.
With the final population PK model (59), simulations of I/R exposure profiles in non-
ventilated and ventilated pneumonia patients (n = 1,000 per renal function category)
based on the covariate patterns observed in RESTORE-IMI-2 and MODIFY I/II (end-stage
renal disease [ESRD] patients only) (60) were performed. For the I/R 500/250-mg and
renal function-adjusted dosing regimens, free plasma concentrations at steady-state
conditions were simulated to characterize the joint I/R PTA profile. The PK/PD targets
in the joint PTA analyses were 30% fT . MIC for IMI and fAUC/MIC $ 8 for REL (46).
Safety analyses were also performed to assess the percentage of patients within each
renal function group that remained below the upper limit safety exposure thresholds
for IMI (AUC0–24, 3229.8 m Mh; Cmax, 625.1 m M) and REL (AUC0–24, 2941.0 m Mh; Cmax,
367.9 m M) (38, 61). In the primary analysis, joint PTA was .90% for all renal function
subgroups at the current I/R MIC breakpoint of 2/4 mg/L, regardless of ventilation sta-
tus. In a sensitivity joint PTA analysis that employed an IMI PK/PD target of 40%
fT . MIC, PTA across renal function subgroups still exceeded 90% for I/R MIC values of
#2/4 mg/L. In the safety analysis, less than 1% of patients in any renal function group
exceeded the AUC0–24 and Cmax thresholds when renal function-adjusted doses of I/R
were simulated, except the ESRD group in which 12.2% of patients exceeded the
threshold for REL AUC0–24.
Performance of approved I/R dosing regimens in preclinical PK/PD infection
model studies. Several preclinical PK/PD studies have been performed to assess the effi-
cacy of the now-approved I/R regimen when it is administered alone and in combination.
In the neutropenic mouse thigh infection model of P. aeruginosa study that examined
humanized I/R exposures, a $2-log reduction in bacterial density was observed in 27/29
(93%) of the IMI-resistant isolates with 500 mg/250 mg every 6 h (43). For the two isolates
with ,2-log reduction in CFU, one was VIM producing (I/R MIC . 32 mg/L) and the sec-
ond produced a GES-20 carbapenemase (I/R MIC = 32 mg/L). In a 24-h in vitro one-com-
partment PK/PD model of P. aeruginosa, the mean reductions in CFU/mL at hour 24 were
22.52, 21.49, 21.15, and 20.61 log10 CFU/mL against isolates with potentiated I/R MICs
of 1, 2, 4, and 8 mg/L, respectively (45). However, regrowth at hour 24 with I/R was com-
monplace and I/R-resistant subpopulations (at 3 the baseline MIC) were observed in one
isolate that exhibited intermediate susceptibility (4/4 mg/L) to I/R at baseline. The combi-
nation of I/R plus colistin resulted in synergistic or additive effects against three of the six
isolates, while the addition of amikacin to I/R achieved only indifferent effects against all
isolates. For the I/R and colistin combination regimen, no resistant subpopulations devel-
oped to either drug, whereas the addition of amikacin to I/R prevented the emergence of
amikacin-resistant subpopulations but the I/R-resistant populations remained for the one
isolate. Similarly, P. aeruginosa regrowth was observed in the 7-day one-compartment
PK/PD infection model study by Noel and colleagues with I/R 500 mg/250 mg i.v. every
6 h despite significant drops in CFU/mL counts during the first 24 h (44). Regrowth
through 7 days was not associated with an increase in I/R MICs. However, regrowth of
P. aeruginosa was greater than the starting inoculum by day 14 and increases in I/R MICs
among evaluated isolates were observed. Addition of amikacin to this infection model
study prevented development of resistance to I/R and regrowth. In a 72-h HFIM by Hirsch
et al. that evaluated 500 mg of REL in combination with 500 to 1,000 mg of IMI every 6 h,
I/R demonstrated rapid bacterial killing and sustained suppression of bacterial growth for
up to 72 h against a K. pneumoniae isolate harboring the KPC-2 carbapenemase and TEM-
and SHV-like extended-spectrum b -lactamases. Against P. aeruginosa, $2-log reduction in

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 7


Perspective Antimicrobial Agents and Chemotherapy

bacterial burden was observed consistently at 24 h but regrowth by 72-h was observed in
2 of the 3 P. aeruginosa isolates evaluated with I/R 500 mg/500 mg i.v. every 6 h (48).

CLINICAL EXPERIENCE
Clinical trial data. To date, I/R has been evaluated in 2 prospective, randomized, dou-
ble-blind, dose-ranging phase II RCTs (62, 63), 2 prospective, randomized, double-blind
phase III RCTs (64, 65), one open-labeled noncomparator phase III trial (66), and 2 real-
world evidence studies (Table 3) (67). The FDA and European Union approvals of I/R were
based, in large part, on the results of RESTORE-IMI-1 and RESTORE-IMI-2 (64, 65). RESTORE-
IMI-1 was a pathogen-focused randomized, double-blind phase III RCT that investigated
the efficacy of I/R and IMI plus colistin against IMI-nonsusceptible infections (65). In this
noninferential, descriptive phase III RCT, hospitalized patients with HABP/VABP, cUTIs, or
cIAIs caused by IMI-nonsusceptible (colistin- and I/R-susceptible) pathogens were random-
ized 2:1 to 5 to 21 days of I/R or IMI plus colistin (loading dose to achieve 300 mg colistin
base activity [CBA] and then up to 150 mg CBA every 12 h). The safety population included
enrolled patients who received $1 dose of study treatment. The microbiologic modified
intent-to-treat (mMITT) population was the primary efficacy population, which comprised
all randomized patients who received $1 dose of study treatment and had a central labo-
ratory-confirmed IMI Gram-negative pathogen from the primary infection site. The primary
efficacy endpoint was overall response, which was defined based on the infection type
per regulatory guidance (HABP/VABP, 28-day all-cause mortality [ACM]; cIAI, day 28 clinical
response; and cUTI, composite clinical and microbiologic response at early follow-up
[EFU]) (68–70). Secondary endpoints were day 28 clinical response (mMITT population), 28-
day all-cause mortality (mMITT population), and treatment-emergent nephrotoxicity
(safety population).
Overall, 50 patients were enrolled, 47 patients (31 I/R versus 16 IMI plus colistin)
received $1 dose of assigned study treatment (safety population), and 31 patients (21
I/R versus 10 IMI plus colistin) met the criteria for the MITT population. Treatment
groups were reasonably well matched at baseline in the MITT population: 36%
were $ 65 years of age, 23% had renal impairment (CLCR , 60 mL/min), 29% had an
APACHE-II score of .15 (71), 36% had HABP/VABP, 51% had a cUTI, and 13% had a
cIAI. The most common pathogens were P. aeruginosa (77%), Klebsiella spp. (16%), and
other Enterobacterales (6%). Detected b -lactamases included AmpC (84% of mMITT
patients), ESBLs (35%), KPC (16%), and OXA-48 (3%).
In the MITT population, favorable outcomes were observed in 71% of patients in
the I/R group versus 70% of patients in the IMI plus colistin group (90% confidence
interval [CI], 227.5, 21.4). Favorable overall response against P. aeruginosa was
observed in 81% (13/16) of patients in the I/R group and 63% (5/8) of patients in the
IMI plus colistin group. Against Enterobacterales, favorable response rates were 40% (2/5)
for I/R and 100% (2/2) for IMI plus colistin, respectively. Day 28 ACM overall was 9.5% (2/
21) and 30% (3/10) in the I/R and IMI plus colistin (adjusted difference, 220.5 [90% CI,
246.5, 6.7]), respectively. Clinical response at day 28 was 71% in the I/R group and 40%
in the IMI plus colistin group (adjusted difference, 31.4 [90% CI, 1.3, 51.5]). All-cause mor-
tality and clinical response at day 28 by pathogen were not provided. Prior meropenem
therapy did not affect overall response, and there were no instances of treatment-emer-
gent I/R-nonsusceptible isolates. Serious adverse events (AEs) occurred in 10% and 31%
of I/R and IMI plus colistin patients, respectively. Three patients (19%) in the IMI plus coli-
stin arm (drug-related blood creatinine increase, drug-related CLCR increase, and treat-
ment-emergent infection; n = 1 each) and none in the I/R arm discontinued treatment
due to AEs. Treatment-emergent nephrotoxicity was significantly less frequent with I/R
than with IMI plus colistin (10% and 56%, respectively; adjusted difference, 245.9 [95%
CI, –69.1% to –18.4%). By the Kidney Disease Improving Global Outcomes (KDIGO) crite-
ria, none of the patients in the I/R arm and 31.3% in the IMI plus colistin group experi-
enced stage III acute kidney injury, and two patients who received IMI plus colistin
required initiation of dialysis during the study period. In a secondary analysis in

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 8


TABLE 3 Summary of published clinical dataa
Key primary and secondary
Study design and population Comparators endpoints Key baseline characteristics Major findings Other findings and comments
Perspective

Prospective, randomized, double- I/R 250 mg, I/R 125 mg, or IMI The primary endpoint was favorable Of the 302 patients randomized, At DCIV, .95% of ME patients in Four patients had treatment-related
blind, dose-ranging phase II study 500 mg alone microbiological response rate at 298 were treated and 230 each treatment arm had AEs leading to discontinuation of
comparing efficacy and safety of I/R Treatment was administered over DCIV in the ME population (77.2%) were ME at DCIV favorable microbiological study drug
with IMI alone in patients with cUTIs 30 minutes every 6 hours for 4– Secondary efficacy endpoints, all  71 in the I/R 250 mg group responses, and both I/R dosing  2.0% with I/R 250 mg
(63) 14 days (step-down to oral assessed in the ME population:  79 in the I/R 125 mg group regimens (500/250 mg and the  1.0% with I/R 125 mg
ciprofloxacin after 4 days was  Microbiological responses at EFU  80 in the IMI group 500/125 mg) were found to be  1% with IMI
permitted) and LFU In the ME at DCI population, groups noninferior to IMI One patient each in the 250 and

July 2022 Volume 66 Issue 7


 Microbiological response at DCIV were well balanced at baseline: All patients with an IMI-NS Gram- 125 mg I/R groups experienced
in patients with IMI-resistant  40% were $65 yrs negative baseline pathogen in liver enzyme elevation (i.e., ALT or
pathogens  52% were female the ME population achieved a AST) of $5% ULN
 Clinical response at DCIV, EFU and  52% had a cUTI vs AP only. favorable microbiological
LFU The most common baseline response at DCIV
pathogens: A favorable clinical response in ME
 E. coli (62%) population at DCIV exceeded
 K. pneumoniae (15%) 95% for all treatment groups
Among the patients with a Microbiological and clinical
confirmed Gram-negative response rates were similar
pathogen (n = 220): between all three treatment
 34.1% were NS to at least one groups at the EFU and LFU visits
third-generation cephalosporin
 11.4% had an IMI NS isolate
 6.8% had an I/R NS isolate
Prospective, randomized, double- I/R 250 mg, I/R 125 mg, or IMI The primary endpoint was favorable Of the 351 subjects randomized, At the DCIV visit, the proportion of The most common AEs were nausea,
blind, dose-ranging phase II study 500 mg alone clinical response at DCIV in the 277 were included in the MITT subjects in the ME population vomiting, and diarrhea
comparing efficacy and safety of I/R Treatment was administered over ME population at 28–42 days population, and 255 (72.6%) with a favorable clinical response Drug-related AE events leading to
with IMI alone in patients with cIAI 30 minutes every 6 hours for after i.v. study therapy (LFU) were included in the ME was .95% for the 3 treatment discontinuation:
(62) 4–14 days in all groups Secondary endpoints included population groups, and both I/R doses were  1 subject who received I/R 125 mg
clinical response at EFU and LFU  83 in the I/R 250 mg group found to be noninferior to IMI (decreased CLCR)
visits and global response  87 in the I/R 125 mg group alone  3 subjects who received IMI alone
at day 28  85 in the IMI group All patients with an IMI (thrombocytosis, nausea, and
In the ME population, groups were nonsusceptible pathogen in the increased ALT)
well balanced at baseline: ME population (n = 34) had a 4 patients experienced liver enzyme
 55% were male favorable microbiological elevations (i.e., AST and ALT
 79% were ,65 yrs old response at the DCIV visit of $5)
 95% had an APACHE-II score of Clinical response rates at the EFU  2 with I/R 250
#15 and LFU visits were similar  2 with IMI alone
 53% had complicated between all treatment groups One patient who received I/R
appendicitis and 17% had Global response at 28 days 250 mg met the criteria for Hy’s
complicated cholecystitis postrandomization in the MITT law (day 2 of treatment), and it
The most common pathogens population was similar (.85%) was deemed to be unrelated to
identified at baseline: between all treatment groups treatment
 E. coli (171 isolates) Three fatal adverse events occurred
 K. pneumoniae (38 isolates) (septic shock, ventricular
(Continued on next page)

10.1128/aac.00256-22
Antimicrobial Agents and Chemotherapy

9
TABLE 3 (Continued)
Key primary and secondary
Study design and population Comparators endpoints Key baseline characteristics Major findings Other findings and comments
Perspective

 P. aeruginosa (37 isolates) fibrillation, and intestinal


36 subjects (13%) had IMI- infarction) among patients in the
resistant Gram-negative I/R 125 mg group, and all deaths
infections at baseline in MITT were considered non-drug
population related
Pathogen-focused randomized, Patients were randomized 2:1 to 5– The primary efficacy endpoint was 50 patients were enrolled: Favorable outcomes were observed Prior meropenem therapy did not
double-blind descriptive phase III 21 days of I/R 250 mg over overall response in the mMITT  47 patients (31 I/R vs 16 IMI plus in 71% of patients in the I/R affect overall response
study that investigated the efficacy 30 minutes every 6 hours or IMI population (comprised all colistin) received $1 dose of group vs 70% of patients in the There were no instances of

July 2022 Volume 66 Issue 7


of I/R and IMI plus colistin in 500 mg over 30 minutes every randomized patients who assigned study treatment (safety IMI plus colistin group (90% CI, treatment-emergent I/R
hospitalized patients with HABP/ 6 hours plus colistin (loading received $1 dose of study population) 227.5, 21.4) in mMITT population nonsusceptibility
VABP, cUTIs, or cIAIs caused by dose to achieve 300 mg CBA then treatment and had a central  31 patients (21 I/R vs 10 IMI plus Favorable overall response against Serious adverse events occurred in
IMI-non-susceptible (colistin- up to 150 mg CBA every laboratory-confirmed IMI non- colistin) met the criteria for the P. aeruginosa was observed in 10% and 31% of I/R and IMI plus
and I/R-susceptible) pathogens 12 hours) susceptible Gram-negative MITT population 81% (13/16) of patients in the I/R colistin patients, respectively
(RESTORE-IMI-1) (65) pathogen from the primary Treatment groups were reasonably group and 63% (5/8) of patients 3 patients (19%) in the IMI plus
infection site) well matched at baseline in the in the IMI plus colistin group colistin arm (drug-related blood
The primary efficacy endpoint was MITT population: Favorable response against creatinine increase, drug-related
defined based on the infection  36% were $65 yrs old Enterobacterales was 40% (2/5) CLCR increase, and treatment-
type per regulatory guidance  23% had renal impairment for I/R and 100% (2/2) for IMI plus emergent infection; n = 1 each)
(HABP/VABP: 28-day all-cause (creatinine clearance ,60 mL/ colistin and 0 in the I/R arm discontinued
mortality; cIAI, day 28 clinical min) Day 28 ACM was 9.5% (2/21) and treatment due to AEs
response; and cUTI, composite  29% had an APACHE-II score of 30% (3/10) in the I/R and IMI plus Treatment-emergent nephrotoxicity
clinical and microbiologic .15 colistin (adjusted difference, was significantly less frequent
response at EFU)  36% had HABP/VABP 220.5 [90% CI, 246.5, 6.7]), with I/R than with IMI plus colistin
Secondary endpoints were day 28  51% had a cUTI respectively (10% and 56%, respectively,
clinical response (mMITT  13% had a cIAI Clinical response at day 28 was 71% adjusted difference 245.9, [95%
population), 28-day all-cause The most common pathogens were in the I/R group and 40% in the CI, –69.1% to –18.4%])
mortality (mMITT population), P. aeruginosa (77%), Klebsiella IMI plus colistin group (adjusted KDIGO criteria, 0 of the patients in
and treatment-emergent spp. (16%), and other difference, 31.4 [90% CI 1.3, 51.5]) the I/R arm and 31.3% in the IMI
nephrotoxicity (safety Enterobacterales (6%) plus colistin group experienced
population) Detected b -lactamases included stage 3 acute kidney injury
AmpC (84% of mMITT patients), 2 patients who received IMI plus
ESBLs (35%), KPC (16%), and colistin required initiation of
OXA-48 (3%) dialysis during the study period
(day 3)
Randomized, double-blind, Patients were randomized in a 1:1 The primary endpoint was day 28 Of 537 randomized patients: In the MITT population, 28-day ACM The most reported drug-related AEs
multicenter trial comparing efficacy ratio to I/R 250 mg over all-cause mortality (ACM) in the  535 (266 I/R, 269 TZP) received $1 was 15.9% with I/R and 21.3% with I/R were diarrhea, increased
and safety of I/R vs TZP in adults 30 minutes every 6 hours or TZP modified intent-to-treat (MITT) dose of study treatment TZP (adjusted treatment aspartate aminotransferase, and
with HABP or VABP (RESTORE-IMI 2 4.5 g over 30 minutes every population (patients who  531 (264 IMI/REL, 267 TZP) were difference, 25.3% [95% CI, increased alanine
study) (64) 6 hours for 7–14 days received study therapy, excluding included in the MITT population 211.9% to 1.2%]); noninferiority aminotransferase (each drug-
All patients received empirical those with only Gram-positive Treatment groups were reasonably P , .001) related AE was ,3%)
intravenous linezolid for $7 days cocci at baseline) well matched at baseline in the For patients who had all confirmed Six I/R patients (2.3%) and 4 TZP
unless baseline LRT cultures Key secondary endpoint was clinical MITT population baseline LRT pathogens patients (1.5%) discontinued
confirmed the absence of MRSA response at the EFU (7–14 days  43% were .65 yrs old susceptible to both study drugs, treatment due to drug-related
after completing therapy) in the  66% were in the ICU 28-day ACM was 16% for I/R and AEs
MITT population  49% had ventilated HABP/VABP 19% for TZP (adjusted treatment Serious drug-related AEs were
Other secondary endpoints included  48% had APACHE-II score of $15 difference, 24.8 (95% CI, 216.1, reported in 1.1% of I/R vs 0.7% of
28-day ACM in mMITT (70) 6.1) TZP patients
population), microbiologic  17% had augmented renal In clinically relevant patient There were 40 deaths (15%) in the
response at EOT and EFU (mMITT clearance and 25% had moderate/ subgroup analyses, 28-day ACM I/R group and 57 deaths (21%) in
population), and clinical response severe renal impairment was similar between treatment the TZP group; no deaths were
at EFU (clinically evaluable [CE] Most frequent Gram-negative groups except for patients with considered drug related
population) pathogens: ventilated HABP/VABP and
 K. pneumoniae (26%) APACHE-II score of $15
 P. aeruginosa (19%)  Ventilated HABP/VABP: 20% for I/R
 Acinetobacter calcoaceticus- and 31% for TZP (adjusted
baumannii complex (16%) treatment difference, 211.2 [95%
 E. coli (16%) CI, 221.6, 20.5)
Among MITT patients with $1  APACHE-II score $ 15: 20% for I/R
identified Gram-gram LRT and 35% for TZP (adjusted

10.1128/aac.00256-22
(Continued on next page)
Antimicrobial Agents and Chemotherapy

10
TABLE 3 (Continued)
Key primary and secondary
Study design and population Comparators endpoints Key baseline characteristics Major findings Other findings and comments
Perspective

pathogen and susceptibility treatment difference, 215.4 [95%


interpretation available CI, 226.2, 24.4])
(n = 380), 80% of I/R patients and Baseline pathogen subgroup
66% of TZP patients had all analyses of 28-day ACM
susceptible isolates
 Enterobacterales: 12% for I/R and
20% for TZP (adjusted treatment
difference, 28.5 [95% CI, 221.9,

July 2022 Volume 66 Issue 7


23.3])
 P. aeruginosa: 33% for I/R and 12%
for TZP (adjusted treatment
difference, 221.3 [95% CI, 24.5,
48.9])
In the secondary endpoint analyses,
favorable clinical response at EFU
was 61.0% with I/R and 55.8%
with TZP (adjusted treatment
difference, 5.0% [95% CI, 23.2%
to 13.2%]); noninferiority
P , .001
Other secondary endpoints (i.e., 28-
day ACM in MITT population,
favorable microbiologic response
at EFU in mMITT population, and
favorable clinical response at EFU
in CE population) were
comparable between treatment
groups
Phase 3, multicenter, open-label, I/R 250 mg over 30 minutes every Primary efficacy endpoints were Of 87 patients screened, 83 (cIAI, Favorable clinical and Drug-related AEs occurred in 18.5%
noncomparative study of I/R in 6 hours for 5–14 days assessed in the ME population: n = 39; cUTI, n = 44) were microbiological responses in the of patients in the MITT
Japanese patients with cUTIs or cIAIs  Clinical response at EOT for enrolled in the study ME population at EOT: population
suspected or confirmed to be patients with cIAI  81 patients (cIAI, n = 37; cUTI,  cUTI: favorable clinical response 3.7% of patients discontinued
caused by I/R-susceptible Gram-  Microbiological response at EOT n = 44) were included in the MITT was 100% and favorable therapy due to a drug-related AE
negative pathogens (66) for patients with cUTI population microbiologic response was 100% (hypoesthesia [two patients],
Secondary efficacy endpoints:  67 patients (cIAI, n = 28; cUTI,  cIAI: favorable clinical response dizziness, tremor, and blurred
 Clinical response at TOC for cIAI n = 39) were included in the ME was 85.7% and favorable vision)
 Microbiological response at TOC population microbiologic response was 85.7% Serious AEs occurred in 9 patients,
for cUTI Baseline characteristics in cIAI Favorable clinical and including one death, but none
patients: microbiological responses in the were considered treatment
 62% were male ME population at TOC: related
 46% were $ 65 yrs  cUTI: favorable clinical response
 62% had a complicated was 92.3% and favorable
appendicitis microbiologic response was 59.0%
Baseline characteristics in cIAI  cIAI: favorable clinical response
patients was 82.1% and favorable
 68% were female microbiologic response was 82.1%
 62% were $ 65 yrs
 50% had an uncomplicated AP
 The most common baseline
pathogen isolated was E. coli
 51% of cIAI patients
 71% of cUTI patients
aDCIV:discontinuation of intravenous therapy; EFU: early follow-up; LFU; late follow-up; ME: microbiologic evaluable; NS: nonsusceptible; ALT: alanine transaminase; AST: aspartate aminotransferase; MRSA: methicillin-resistant
Staphylococcus aureus; EOT: end of treatment; TOC: test of cure; CE: clinically evaluable; I/R: imipenem/relebactam; IMI: imipenem; cUTI: complicated urinary tract infections; DCIV: discontinuation of intravenous therapy; AEs:
adverse events; ULN: upper limit of normal; cIAI: complicated intra-abdominal infections; IV: intravenous; MITT: microbiologic intention to treat; HABP; hospital-acquired bacterial pneumonia; VABP: ventilator-associated bacterial
pneumonia; CBA: colistin base activity; mMITT: modified microbiologic intent-to-treat; CI: confidence interval; KDIGO: Kidney Disease Improving Global Outcomes; ACM: all-cause mortality; TZP: piperacillin/tazobactam; ICU:
intensive care unit; APACHE-II: acute physiology and chronic health evaluation-II.

10.1128/aac.00256-22
Antimicrobial Agents and Chemotherapy

11
Perspective Antimicrobial Agents and Chemotherapy

RESTORE-IMI-1, similar outcomes were observed in primary mMITT population (n = 31),


where eligibility was based on central laboratory susceptibility testing, and the supple-
mental mMITT (SmMITT) population, where eligibility was based on local, site-level test-
ing (n = 41) (72).
I/R was also compared with piperacillin-tazobactam (TZP) in RESTORE-IMI 2, a multi-
center, randomized, phase III noninferiority trial of adult patients with HABP/VABP (64).
In RESTORE-IMI 2, adult patients with nonventilated HABP, ventilated HABP, or VABP
were randomized (stratified by nonventilated HABP versus ventilated HABP/VABP and
by APACHE-II score [,15 versus $15] [71]) in a 1:1 ratio to I/R or TZP 44.5 g, intrave-
nously (30-minute infusion) every 6 h for 7 to 14 days. All patients received empirical
intravenous linezolid for $7 days unless baseline LRT cultures confirmed the absence
of methicillin-resistant Staphylococcus aureus. The primary endpoint was day 28 all-
cause mortality (ACM) in the MITT population (patients who received study therapy,
excluding those with only Gram-positive cocci at baseline). The key secondary end-
point was clinical response at the EFU (7 to 14 days after completing therapy) in the
MITT population. Other secondary endpoints included 28-day ACM in mMITT popula-
tion, microbiologic response at EOT and EFU (mMITT population), and clinical response
at EFU (clinically evaluable [CE] population).
Overall, 537 patients were randomized and 531 (264 I/R, 267 TZP) were included in
the MITT population. Among patients in the MITT population, 43% were .65 years old,
66% were in the intensive care unit (ICU), 49% had ventilated HABP/VABP, 48% had
APACHE-II score of $15 (71), 17% had augmented renal clearance, and 25% had mod-
erate to severe renal impairment. Most frequent Gram-negative pathogens were K.
pneumoniae (26%), P. aeruginosa (19%), Acinetobacter calcoaceticus-baumannii complex
(16%), and Escherichia coli (16%). Among MITT patients with $1 identified Gram-gram
LRT pathogens and susceptibility interpretation available (n = 380), 80% of I/R patients
and 66% of TZP patients had all susceptible isolates. Mean treatment duration was
8.7 days with I/R and 8.3 days with TZP.
In the MITT population, 28-day ACM was 15.9% with I/R and 21.3% with TZP
(adjusted treatment difference, 25.3% [95% CI, 211.9% to 1.2%]; noninferiority P ,
0.001). For patients who had all confirmed baseline LRT pathogens susceptible to both
study drugs, 28-day ACM was 16% for I/R and 19% for TZP (adjusted treatment differ-
ence, 24.8 [95% CI, 216.1, 6.1]). No data were provided on 28-ACM rates for patients
with nonsusceptible LRT Gram-negative pathogens to treatment received. In the clini-
cally relevant patient subgroups analyses, 28-day ACM was similar between treatment
groups except for patients with ventilated HABP/VABP and APACHE-II score of $15.
Among ventilated HABP/VABP patients, 28-ACM was 20% for I/R and 31% for TZP
(adjusted treatment difference, 211.2 [95% CI, 221.6, 20.5]). For the subgroup of
patients with an APACHE-II score of $15, 28-ACM was 20% for I/R and 35% for TZP
(adjusted treatment difference, 215.4 [95% CI, 226.2, 24.4]). In the baseline pathogen
subgroup analyses, 28-day ACM for patients with Enterobacterales (I/R [n = 68] versus
TZP [n = 66]) was 12% for I/R and 20% for TZP (adjusted treatment difference, 28.5
[95% CI, 221.9, 23.3]). For patients with P. aeruginosa (I/R [n = 15] versus TZP [n = 25]),
28-day ACM was 33% for I/R and 12% for TZP (adjusted treatment difference, 221.3
[95% CI, 24.5, 48.9]).
In the secondary endpoint analyses, I/R was found to be noninferior to TZP for
favorable clinical response at EFU (61.0% with I/R and 55.8% with TZP [adjusted treat-
ment difference, 5.0% {95% CI, 23.2% to 13.2%}; noninferiority P , .001]). Other sec-
ondary endpoints (i.e., 28-day ACM in MITT population, favorable microbiologic
response at EFU in mMITT population, and favorable clinical response at EFU in CE
population) were also comparable between treatment groups. Drug-related AEs
occurred in 11.7% of patients who received I/R and 9.7% of patients who received TZP.
The most reported drug-related AEs with I/R were diarrhea, increased aspartate amino-
transferase, and increased alanine aminotransferase (each drug-related AE was ,3%).
Six I/R patients (2.3%) and 4 TZP patients (1.5%) discontinued treatment due to drug-

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 12


Perspective Antimicrobial Agents and Chemotherapy

related AEs. Serious drug-related AEs were reported in 1.1% of I/R versus 0.7% of TZP
patients. There were 40 deaths (15%) in the I/R group and 57 deaths (21%) in the TZP
group; no deaths were considered drug related.
Real-world data. Published real-world evidence with I/R is currently limited to two
noncomparator case reviews (67, 73). The first was a multicenter observational study of
21 patients with doses of I/R ranging from 500/250 to 1,250/625 mg every 6 h (67). The
most common infection types were respiratory tract infections, including HABP and
VABP (52%), UTIs (14%), and invasive prosthetic device infections (14%). Pseudomonas
aeruginosa was the predominant pathogen (76%), followed by K. pneumoniae; 29%
were polymicrobial infections and 29% had concurrent bacteremia. Multidrug-resistant
pathogens were common; 3/8 (38%) patients with Enterobacterales had a carbapenem-
resistant Enterobacterales (CRE) infection, and 15/16 (94%) P. aeruginosa cases were
MDR. Only 52% of cases had I/R MICs performed (mostly Etest), with an MIC range of
0.125/4 to $32/4 (73% were susceptible). The median (IQR) duration of I/R therapy was
8 (4.5 to 14) days, and combination therapy was commonplace (29%). Thirty-day mor-
tality and clinical cure were 33% and 62%, respectively. Nonsusceptibility to I/R devel-
oped on treatment in 1 of 9 isolates with subsequent MIC testing post-index culture
(nonsusceptible organism not identified, but the patient had a CRE/CR P. aeruginosa
pneumonia). Microbiological recurrence occurred in 24% (5/21) of patients, and 2 of
the 5 patients with recurrence had an initial P. aeruginosa I/R-susceptible isolate but a
nonsusceptible I/R isolate on subsequent culture. Two adverse events were reported (1
gastrointestinal [nausea, vomiting, diarrhea] and 1 encephalopathic [altered mental
status, somnolence, new-onset seizures]), but neither resulted in drug discontinuation.
The second real-world evidence study described the experience of 19 patients who
were treated with I/R for an MDR P. aeruginosa HABP/VABP (73). Among these 19
patients, 5 had emergence of I/R-nonsusceptible P. aeruginosa during treatment or
within 30 days posttreatment. All 5 patients with recovered emergent I/R-nonsuscepti-
ble P. aeruginosa were managed in the ICU and had failed prior antibiotic regimens,
including two patients who received I/R after treatment-emergent resistance to C/T.
Baseline I/R MICs ranged from 0.5 to 1 mg/L, and postexposure I/R MICs ranged from 4
to 32 mg/L following 10- to 28-day treatment courses. Whole-genome sequencing
(WGS) was performed, and each recovered P. aeruginosa isolate across the 5 patients
had a unique sequence type (ST), including two patients with high-risk MDR clones
ST235 and ST244 (74). Genomic sequence analyses identified treatment-emergent
mutations in MexAB-OprM and/or MexEF-OprN efflux operons (mutations in both
membrane transporter and transcriptional regulator genes) that arose independently
in each patient and across the distinct P. aeruginosa STs. There were no clear associations
between emergent I/R-nonsusceptible isolates and mutations in porin genes or genes
encoding penicillin-binding proteins. Furthermore, no isolates harbored MBLs or other
b -lactamase enzymes known to confer I/R resistance (74). Testing with efflux-inhibitor
PA b N restored I/R susceptibility against all but 1 I/R-nonsusceptible isolate that had a
mutation in ampC, suggesting that the MexAB-OprM and MexEF-OprN operons potentially
work in concert to extrude REL and result in I/R-nonsusceptible strains, most likely in the
presence of other upregulated resistance mechanisms (i.e., downregulated OprD porin
channels and ampC overexpression). Alternatively, as REL differs from other diazabicy-
clooctane BLIs in that it carries a positively charged side chain believed to limit drug efflux
(5), an alternative explanation for the observed findings was that mutations in MexEF-
OprN may have limited REL entry into P. aeruginosa isolates largely by downregulating
OprD porin channels.

ROLE OF IMIPENEM-RELEBACTAM IN CLINICAL PRACTICE


Overall, published preclinical PK/PD infection model, susceptibility surveillance, and
clinical data for I/R are highly favorable and indicate that I/R is a welcomed addition to
our antibiotic armamentarium against CR-NME and DTR P. aeruginosa. Consistent with
best antibiotic stewardship practices, I/R use should be limited to clinical situations in

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 13


Perspective Antimicrobial Agents and Chemotherapy

which a CR-NME or DTR P. aeruginosa infection is highly suspected or documented.


Like the other recently approved b -lactams, I/R demonstrated noninferior efficacy for
the treatment of adults with HABP/VABP, and its safety profile was consistent with
those of other b -lactams (5, 6, 62–66). Although it was a descriptive, small-scale RCT,
RESTORE-IMI-2 suggested that I/R may be associated with similar or improved 28-day
response rates and less nephrotoxicity relative to those of IMI and colistin combination
therapy for the treatment of adult patients with IMI-resistant NME and P. aeruginosa
infections (65). Available preclinical PK/PD (43, 46, 47) and PTA data (59) provide
adequate support for the current I/R and renal function-adjusted dosing regimens at
the current CLSI and EUCAST susceptibility breakpoints for P. aeruginosa and NME.
Across its phase II and III RCTs, no cases of treatment-emergent I/R resistance were
reported, lending further support for the adequacy of the I/R FDA- and European
Union-approved I/R dosing regimen. However, regrowth and resistance emergence
were observed in two in vitro PK/PD models of P. aeruginosa infections (44, 45), and
two recent case series revealed treatment-emergent I/R resistance during therapy
among extensively antibiotic experienced patients with infections due to CR/DTR P.
aeruginosa (67, 73). The preclinical infection model and clinical I/R treatment-emergent
resistance findings with MDR/CR/DTR P. aeruginosa are highly consistent with the other
recently approved novel b -lactams (28, 75–78) and highlight the critical importance of
susceptibility testing of baseline and subsequent isolates when considering one of the
newer b -lactam agents, especially among patients not responding to therapy or with
recurrent infections.
Given the current lack of comparator clinical data between the newer b -lactams,
the most informative way to differentiate between the recently approved b -lactams is
by comparison of their microbiologic in vitro susceptibility profiles. Unlike the other
recently approved b -lactams, I/R lacks activity against Morganellaceae (5). It also exhib-
its limited microbiologic activity against MBL-producing NME and P. aeruginosa and CR
Acinetobacter sp., which is consistent with the recently approved b -lactams except
cefiderocol (3, 17, 21). In comparative surveillance programs, I/R in vitro activity was
similar to that of CZA and M/V for CR-NME and C/T and CZA for MDR/CR/DTR P. aerugi-
nosa. Subtle differences in susceptibility results among the newer b -lactams were
observed across several comparative surveillance studies with I/R retaining some
microbiologic activity against CR-NME and MDR/CR/DTR P. aeruginosa isolates that are
resistant to some of the newer b -lactams and vice versa. Data suggest that I/R may
retain activity against CR-NME isolates that develop resistance to CZA via KPC-3 muta-
tions and DTR P. aeruginosa isolates that develop resistance to C/T and CZA due to
PDC mutations (37). Conversely, I/R is typically nonsusceptible to NME and/or P. aeruginosa
isolates that produce SME b -lactamases, OXA-48-like b -lactamases, GES-like carbapenemase,
and VEB-producing b -lactamases in contrast to some newer b -lactams (2, 16–18).
Expression of multiple resistance mechanisms simultaneously (i.e., downregulated or deleted
porins, b -lactamase hyperproduction, upregulated efflux pumps, etc.) may also result in dis-
cordant susceptibility patterns for the newer b -lactams, as some resistance expression pat-
terns have a higher propensity to result in resistance to certain b -lactams more so than
others (28, 73, 75). Additional data are needed to fully elucidate the role of concurrently
expressed resistance mechanisms in causing nonsusceptibility against each of the newer
b -lactam agents (2, 4, 14, 23).
Unfortunately, the resistance mechanism(s) present in a CR-NME or DTR P. aerugi-
nosa infection are infrequently known at baseline, even with use of rapid diagnostics
(rapid diagnostic testing can detect the presence of only select resistance genes). From
a clinical practice standpoint, susceptibility testing should ideally be performed for all
the newer agents for patients with CR-NME and DTR P. aeruginosa infections to deter-
mine the best available treatment options. In the absence of near real-time antibiotic
susceptibility results, institution-specific antibiogram data should guide early targeted
treatment decisions for patients with CR-NME and CR P. aeruginosa infections. In a clin-
ical situation in which some or all newer agents are active against the recovered CR-

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 14


Perspective Antimicrobial Agents and Chemotherapy

NME and/or DTR P. aeruginosa isolate(s), there are no comparator data to indicate that
one should be preferentially used over another, and selection of a I/R relative to
another agent should be made on a case-by-case basis at this time. Of note, unlike the
newer b -lactams, I/R had reliable microbiologic activity against ampicillin-susceptible
Enterococcus sp. and Gram-negative anaerobes, making it a potentially preferred agent
in patients with polymicrobial infections that include Enterococcus sp., CR-NME, and/or
DTR P. aeruginosa (5).

FUTURE DIRECTIONS
Additional real-world data are needed to determine the optimal place for I/R ther-
apy in our antibiotic arsenal. Comparative preclinical PK/PD and clinical data are
needed between newer BL/BLIs to determine optimal treatment of CR-NME and DTR P.
aeruginosa infections. Currently, differences in in vitro activity among newer BL/BLIs
appear to be limited, and overall differences in coverage are driven by the intrinsic ac-
tivity of the b -lactam agent (i.e., IMI exhibits coverage against Enterococci but does not
have activity against Morganellaceae and CR-NME that express certain b -lactamases).
An emerging theme with newer agents is the development of resistance during or
shortly after therapy, particularly among patients with DTR P. aeruginosa infections
and extensive antibiotic exposure (28, 73, 76). In patients with DTR P. aeruginosa infec-
tions who develop resistance on C/T, there is some suggestion that I/R retains activity
in treatment-emergent infections; however, a recent case series suggests that subse-
quent I/R resistance may develop (73). There is a need to better understand the PK/PD
target for resistance selection against borderline-resistant and hypermutable strains,
particularly for P. aeruginosa. To date, most I/R preclinical PK/PD infection model stud-
ies have examined only standard inocula, short courses, and isolates that were not re-
sistant to the newer agents at baseline. There may be some flexibility with use of a
higher dose of I/R (safety has been evaluated with doses of IMI 1,000 mg and REL up to
1,000 mg every 6 h), and it may be that higher doses are ultimately needed to treat
patients with high inoculum infections that are resistant to other newer b -lactams at
baseline (38).
There is also a need to establish optimal combination therapy given reports of
treatment-emergent resistance and overall lack of activity against certain resistance ge-
notypes. Limited preclinical PK/PD with amikacin and colistin suggests that use of com-
bination therapy may enhance killing and minimize resistance emergence against P.
aeruginosa in vitro (43, 45). However, while both combinations showed synergy in
static concentration time kills, only combinations with colistin were additive or syner-
gistic against six MDR P. aeruginosa isolates tested in a dynamic infection model (45).
Ultimately, it may be that newer agents need to be paired with a second active drug,
especially with P. aeruginosa infections, given the high rates of resistance emergence
observed in recent real-world evidence studies. As part of combination therapy studies,
there needs to be an assessment of aztreonam (ATM) with I/R for isolates producing
MBLs. Limited in vitro studies have suggested that aztreonam plus I/R exhibits synergy
against MBL-producing K. pneumoniae and P. aeruginosa that constitutively produce
PDCs (79, 80).
I/R may also have a role in the treatment of Mycobacterium abscessus complex infec-
tions (81, 82). IMI remains one of the mainstays of care for the treatment of this highly
resistant pathogen, and REL appears to have a beneficial, though modest, effect on IMI
susceptibilities. Combinations of I/R with amoxicillin have shown particularly good ac-
tivity in vitro (82). To date, data on I/R against M. abscessus are limited to in vitro stud-
ies, though this may prove to be a promising step forward in the treatment of these
infections.

CONCLUSIONS
Available preclinical PK/PD infection model, microbiologic surveillance, PTA, and
clinical data for I/R support the use and current dosing of I/R for the treatment of

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 15


Perspective Antimicrobial Agents and Chemotherapy

patients with HABP/VABP, cUTI, and cIAI due to highly resistant NME and P. aeruginosa.
From a microbiologic in vitro activity perspective, I/R is most comparable to CZA and
M/V for CR-NME and C/T and CZA for MDR/CR/DTR P. aeruginosa. Data indicate that I/R
retains some activity against CR-NME and MDR/CR/DTR P. aeruginosa isolates that are
resistant at baseline or emerge during therapy to the newer b -lactams and vice versa,
suggesting that susceptibility testing be performed for all the newer agents for
patients with CR-NME and DTR P. aeruginosa infections to identify the most viable
treatment options. Further comparative PK/PD and clinical studies are warranted to
determine the optimal role of I/R, alone and in combination, for the treatment of
patients with highly resistant Gram-negative infections. Until further data are available,
I/R is a reasonable early targeted treatment option in patients with suspected or docu-
mented infections due to CR-NME and DTR P. aeruginosa infections when the benefits
outweigh the risks.

ACKNOWLEDGMENTS
No funding was received for this publication.
T.P.L. has received consultant honoraria and investigator-initiated grant support from
Merck & Co. J.N.O. has received investigator-initiated grant support from Merck & Co.

REFERENCES
1. Blizzard TA, Chen H, Kim S, Wu J, Bodner R, Gude C, Imbriglio J, Young K, 11. Sader HS, Carvalhaes CG, Shortridge D, Castanheira M. 2022. Comparative
Park YW, Ogawa A, Raghoobar S, Hairston N, Painter RE, Wisniewski D, activity of newer beta-lactam/beta-lactamase inhibitor combinations
Scapin G, Fitzgerald P, Sharma N, Lu J, Ha S, Hermes J, Hammond ML. against Pseudomonas aeruginosa from patients hospitalized with pneu-
2014. Discovery of MK-7655, a beta-lactamase inhibitor for combination monia in European medical centers in 2020. Eur J Clin Microbiol Infect Dis
with Primaxin(R). Bioorg Med Chem Lett 24:780–785. https://doi.org/10 41:319–324. https://doi.org/10.1007/s10096-021-04363-7.
.1016/j.bmcl.2013.12.101. 12. Lob SH, DePestel DD, DeRyke CA, Kazmierczak KM, Young K, Motyl MR,
2. Young K, Painter RE, Raghoobar SL, Hairston NN, Racine F, Wisniewski D, Sahm DF. 2021. Ceftolozane/tazobactam and imipenem/relebactam cross-
Balibar CJ, Villafania A, Zhang R, Sahm DF, Blizzard T, Murgolo N, susceptibility among clinical isolates of Pseudomonas aeruginosa from
Hammond ML, Motyl MR. 2019. In vitro studies evaluating the activity of patients with respiratory tract infections in ICU and non-ICU wards-SMART
imipenem in combination with relebactam against Pseudomonas aerugi- United States 2017–2019. Open Forum Infect Dis 8:ofab320. https://doi.org/
nosa. BMC Microbiol 19:150. https://doi.org/10.1186/s12866-019-1522-7. 10.1093/ofid/ofab320.
3. Zhang H, Jia P, Zhu Y, Zhang G, Zhang J, Kang W, Duan S, Zhang W, Yang 13. Lob SH, Hackel MA, Young K, Motyl MR, Sahm DF. 2021. Activity of imipe-
Q, Xu Y. 2021. Susceptibility to imipenem/relebactam of Pseudomonas nem/relebactam and comparators against Gram-negative pathogens
aeruginosa and Acinetobacter baumannii isolates from Chinese intra-ab- from patients with bloodstream infections in the United States and Can-
dominal, respiratory and urinary tract infections: SMART 2015 to 2018. ada - SMART 2018–2019. Diagn Microbiol Infect Dis 100:115421. https://
Infect Drug Resist 14:3509–3518. https://doi.org/10.2147/IDR.S325520. doi.org/10.1016/j.diagmicrobio.2021.115421.
4. Haidar G, Clancy CJ, Chen L, Samanta P, Shields RK, Kreiswirth BN, Nguyen 14. Karlowsky JA, Lob SH, Young K, Motyl MR, Sahm DF. 2021. In vitro activity
MH. 2017. Identifying spectra of activity and therapeutic niches for cefta- of imipenem/relebactam against Gram-negative bacilli from pediatric
patients-study for monitoring antimicrobial resistance trends (SMART)
zidime-avibactam and imipenem-relebactam against carbapenem-resist-
global surveillance program 2015–2017. J Pediatric Infect Dis Soc 10:
ant Enterobacteriaceae. Antimicrob Agents Chemother 61. https://doi
274–281. https://doi.org/10.1093/jpids/piaa056.
.org/10.1128/AAC.00642-17.
15. Karlowsky JA, Lob SH, Raddatz J, DePestel DD, Young K, Motyl MR, Sahm DF.
5. Anonymous. 2019. RECARBRIOTM (imipenem, cilastatin, and relebactam)
2021. In vitro activity of imipenem/relebactam and ceftolozane/tazobactam
for injection, for intravenous use. Merck Sharp & Dohme LLC, Rahway, NJ, USA.
against clinical isolates of Gram-negative bacilli with difficult-to-treat resist-
https://www.merck.com/product/usa/pi_circulars/r/recarbrio/recarbrio_pi.
ance and multidrug-resistant phenotypes-study for monitoring antimicrobial
pdf. Accessed on February 1, 2022.
resistance trends, United States 2015–2017. Clin Infect Dis 72:2112–2120.
6. Anonymous. 2020. Recarbrio - EMEA/H/C/004808 - N/0010. European Medi-
https://doi.org/10.1093/cid/ciaa381.
cines Agency, Amsterdam, The Netherlands. https://www.ema.europa.eu/en/ 16. Biagi M, Shajee A, Vialichka A, Jurkovic M, Tan X, Wenzler E. 2020. Activity
documents/product-information/recarbrio-epar-product-information_en.pdf. of imipenem-relebactam and meropenem-vaborbactam against carbape-
Accessed on February 1, 2022. nem-resistant, SME-producing Serratia marcescens. Antimicrob Agents
7. Clinical and Laboratory Standards Institute. 2005. The Clinical and Laboratory Chemother 64. https://doi.org/10.1128/AAC.02255-19.
Standards Institute (CLSI) has published M100—performance standards for 17. Karlowsky JA, Lob SH, Kazmierczak KM, Young K, Motyl MR, Sahm DF.
antimicrobial susceptibility testing, 31st ed. Clinical and Laboratory Stand- 2020. In-vitro activity of imipenem/relebactam and key beta-lactam
ards Institute, Wayne, PA. agents against Gram-negative bacilli isolated from lower respiratory tract
8. EUCAST. 2022. Clinical breakpoints–breakpoints and guidance. https:// infection samples of intensive care unit patients - SMART Surveillance
www.eucast.org/clinical_breakpoints/. United States 2015–2017. Int J Antimicrob Agents 55:105841. https://doi
9. Karlowsky JA, Lob SH, Kazmierczak KM, Young K, Motyl MR, Sahm DF. .org/10.1016/j.ijantimicag.2019.10.022.
2018. In vitro activity of imipenem-relebactam against clinical isolates of 18. Mushtaq S, Meunier D, Vickers A, Woodford N, Livermore DM. 2021. Activity
Gram-negative bacilli isolated in hospital laboratories in the United States of imipenem/relebactam against Pseudomonas aeruginosa producing ESBLs
as part of the SMART 2016 program. Antimicrob Agents Chemother 62. and carbapenemases. J Antimicrob Chemother 76:434–442. https://doi.org/
https://doi.org/10.1128/AAC.00169-18. 10.1093/jac/dkaa456.
10. Sader HS, Castanheira M, Duncan LR, Mendes RE. 2021. Antimicrobial activ- 19. Vazquez-Ucha JC, Seoane-Estevez A, Rodino-Janeiro BK, Gonzalez-Bardanca
ities of ceftazidime/avibactam, ceftolozane/tazobactam, imipenem/relebac- M, Conde-Perez K, Martinez-Guitian M, Alvarez-Fraga L, Arca-Suarez J, Lasarte-
tam, meropenem/vaborbactam, and comparators against Pseudomonas Monterrubio C, Gut M, Gut I, Alvarez-Tejado M, Oviano M, Beceiro A, Bou G,
aeruginosa from patients with skin and soft tissue infections. Int J Infect Dis GEMARA-SEIMC/REIPI Enterobacterales Study Group. 2021. Activity of imipe-
113:279–281. https://doi.org/10.1016/j.ijid.2021.10.022. nem/relebactam against a Spanish nationwide collection of carbapenemase-

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 16


Perspective Antimicrobial Agents and Chemotherapy

producing Enterobacterales. J Antimicrob Chemother 76:1498–1510. https:// 33. Carpenter J, Neidig N, Campbell A, Thornsberry T, Truex T, Fortney T, Zhang
doi.org/10.1093/jac/dkab043. Y, Bush K. 2019. Activity of imipenem/relebactam against carbapenemase-
20. Johnston BD, Thuras P, Porter SB, Anacker M, VonBank B, Vagnone PS, producing Enterobacteriaceae with high colistin resistance. J Antimicrob Che-
Witwer M, Castanheira M, Johnson JR. 2020. Activity of imipenem-rele- mother 74:3260–3263. https://doi.org/10.1093/jac/dkz354.
bactam against carbapenem-resistant Escherichia coli isolates from the 34. Galani I, Souli M, Nafplioti K, Adamou P, Karaiskos I, Giamarellou H,
United States in relation to clonal background, resistance genes, coresist- Antoniadou A, Study Collaborators. 2019. In vitro activity of imipenem-
ance, and region. Antimicrob Agents Chemother 64. https://doi.org/10 relebactam against non-MBL carbapenemase-producing Klebsiella pneu-
.1128/AAC.02408-19. moniae isolated in Greek hospitals in 2015–2016. Eur J Clin Microbiol
21. Lob SH, Karlowsky JA, Young K, Motyl MR, Hawser S, Kothari ND, Sahm Infect Dis 38:1143–1150. https://doi.org/10.1007/s10096-019-03517-y.
DF. 2020. In vitro activity of imipenem-relebactam against resistant phe- 35. Shields RK, Chen L, Cheng S, Chavda KD, Press EG, Snyder A, Pandey R,
notypes of Enterobacteriaceae and Pseudomonas aeruginosa isolated Doi Y, Kreiswirth BN, Nguyen MH, Clancy CJ. 2017. Emergence of ceftazi-
from intraabdominal and urinary tract infection samples—SMART Surveil- dime-avibactam resistance due to plasmid-borne blaKPC-3 mutations
lance Europe 2015–2017. J Med Microbiol 69:207–217. https://doi.org/10 during treatment of carbapenem-resistant Klebsiella pneumoniae Infec-
.1099/jmm.0.001142. tions. Antimicrob Agents Chemother 61. https://doi.org/10.1128/AAC
22. Senchyna F, Gaur RL, Sandlund J, Truong C, Tremintin G, Kultz D, Gomez .02097-16.
CA, Tamburini FB, Andermann T, Bhatt A, Tickler I, Watz N, Budvytiene I, 36. Haidar G, Clancy CJ, Shields RK, Hao B, Cheng S, Nguyen MH. 2017. Muta-
Shi G, Tenover FC, Banaei N. 2019. Diversity of resistance mechanisms in tions in blaKPC-3 that confer ceftazidime-avibactam resistance encode
carbapenem-resistant Enterobacteriaceae at a health care system in novel KPC-3 variants that function as extended-spectrum beta-lacta-
Northern California, from 2013 to 2016. Diagn Microbiol Infect Dis 93: mases. Antimicrob Agents Chemother 61. https://doi.org/10.1128/AAC
250–257. https://doi.org/10.1016/j.diagmicrobio.2018.10.004. .02534-16.
23. Gomez-Simmonds A, Stump S, Giddins MJ, Annavajhala MK, Uhlemann 37. Bail L, Ito CAS, Arend L, Nogueira KDS, Tuon FF. 2022. Activity of imipenem-
AC. 2018. Clonal background, resistance gene profile, and porin gene relebactam and ceftolozane-tazobactam against carbapenem-resistant Pseu-
mutations modulate in vitro susceptibility to imipenem-relebactam in domonas aeruginosa and KPC-producing Enterobacterales. Diagn Microbiol
diverse Enterobacteriaceae. Antimicrob Agents Chemother 62. https:// Infect Dis 102:115568. https://doi.org/10.1016/j.diagmicrobio.2021.115568.
doi.org/10.1128/AAC.00573-18. 38. Rhee EG, Rizk ML, Calder N, Nefliu M, Warrington SJ, Schwartz MS, Mangin
24. Lombardo D, Ambretti S, Lazzarotto T, Gaibani P. 2022. In vitro activity of E, Boundy K, Bhagunde P, Colon-Gonzalez F, Jumes P, Liu Y, Butterton JR.
imipenem-relebactam against KPC-producing Klebsiella pneumoniae re- 2018. Pharmacokinetics, safety, and tolerability of single and multiple
sistant to ceftazidime-avibactam and/or meropenem-vaborbactam. Clin doses of relebactam, a beta-lactamase inhibitor, in combination with imi-
Microbiol Infect 28:749–751. https://doi.org/10.1016/j.cmi.2022.01.025. penem and cilastatin in healthy participants. Antimicrob Agents Chemo-
25. Maraki S, Mavromanolaki VE, Magkafouraki E, Moraitis P, Stafylaki D, Kasimati ther 62:e00280-18. https://doi.org/10.1128/AAC.00280-18.
A, Scoulica E. 2022. Epidemiology and in vitro activity of ceftazidime-avibac- 39. Bhagunde P, Colon-Gonzalez F, Liu Y, Wu J, Xu SS, Garrett G, Jumes P,
tam, meropenem-vaborbactam, imipenem-relebactam, eravacycline, plazo- Lasseter K, Marbury T, Rizk ML, Lala M, Rhee EG, Butterton JR, Boundy K.
micin, and comparators against Greek carbapenemase-producing Klebsiella 2020. Impact of renal impairment and human organic anion transporter
pneumoniae isolates. Infection 50:467–474. https://doi.org/10.1007/s15010 inhibition on pharmacokinetics, safety and tolerability of relebactam
-021-01735-1. combined with imipenem and cilastatin. Br J Clin Pharmacol 86:944–957.
26. Castanheira M, Doyle TB, Deshpande LM, Mendes RE, Sader HS. 2021. Ac- https://doi.org/10.1111/bcp.14204.
tivity of ceftazidime/avibactam, meropenem/vaborbactam and imipe- 40. Rizk ML, Rhee EG, Jumes PA, Gotfried MH, Zhao T, Mangin E, Bi S, Chavez-
nem/relebactam against carbapenemase-negative carbapenem-resistant Eng CM, Zhang Z, Butterton JR. 2018. Intrapulmonary pharmacokinetics
Enterobacterales isolates from US hospitals. Int J Antimicrob Agents 58: of relebactam, a novel beta-lactamase inhibitor, dosed in combination
106439. https://doi.org/10.1016/j.ijantimicag.2021.106439. with imipenem-cilastatin in healthy subjects. Antimicrob Agents Chemo-
27. Kuo SC, Wang YC, Tan MC, Huang WC, Shiau YR, Wang HY, Lai JF, Huang ther 62. https://doi.org/10.1128/AAC.01411-17.
IW, Lauderdale TL. 2021. In vitro activity of imipenem/relebactam, mero- 41. Powles MA, Galgoci A, Misura A, Colwell L, Dingley KH, Tang W, Wu J,
penem/vaborbactam, ceftazidime/avibactam, cefepime/zidebactam and Blizzard T, Motyl M, Young K. 2018. In vivo efficacy of relebactam (MK-7655)
other novel antibiotics against imipenem-non-susceptible Gram-negative in combination with imipenem-cilastatin in murine infection models. Antimi-
bacilli from Taiwan. J Antimicrob Chemother 76:2071–2078. https://doi crob Agents Chemother 62. https://doi.org/10.1128/AAC.02577-17.
.org/10.1093/jac/dkab141. 42. Mavridou E, Melchers RJ, van Mil AC, Mangin E, Motyl MR, Mouton JW.
28. Rubio AM, Kline EG, Jones CE, Chen L, Kreiswirth BN, Nguyen MH, Clancy CJ, 2015. Pharmacodynamics of imipenem in combination with beta-lacta-
Cooper VS, Haidar G, Van Tyne D, Shields RK. 2021. In vitro susceptibility of mase inhibitor MK7655 in a murine thigh model. Antimicrob Agents Che-
multidrug-resistant Pseudomonas aeruginosa following treatment-emer- mother 59:790–795. https://doi.org/10.1128/AAC.03706-14.
gent resistance to ceftolozane-tazobactam. Antimicrob Agents Chemother 43. Reyes S, Abdelraouf K, Nicolau DP. 2020. In vivo activity of human-simu-
65. https://doi.org/10.1128/AAC.00084-21. lated regimens of imipenem alone and in combination with relebactam
29. Bhagwat SS, Legakis NJ, Skalidis T, Loannidis A, Goumenopoulos C, Joshi against Pseudomonas aeruginosa in the murine thigh infection model. J
PR, Shrivastava R, Palwe SR, Periasamy H, Patel MV, Chatzipanagiotou S, Antimicrob Chemother 75:2197–2205.
Hellenic Cefepime/Zidebactam Study Group. 2021. In vitro activity of 44. Noel AR, Bowker KE, Attwood M, MacGowan AP. 2019. Antibacterial effect
cefepime/zidebactam (WCK 5222) against recent Gram-negative isolates of imipenem/relebactam on aerobic Gram-negative bacilli: in vitro simu-
collected from high resistance settings of Greek hospitals. Diagn Micro- lations of 7 or 14 day human exposures. J Antimicrob Chemother 74:
biol Infect Dis 100:115327. https://doi.org/10.1016/j.diagmicrobio.2021 1945–1951. https://doi.org/10.1093/jac/dkz114.
.115327. 45. Chen IH, Nicolau DP, Kuti JL. 2020. Imipenem/cilastatin/relebactam alone
30. Karlowsky JA, Hackel MA, Bouchillon SK, Sahm DF. 2020. In vitro activity and in combination against Pseudomonas aeruginosa in the in vitro phar-
of WCK 5222 (cefepime-zidebactam) against worldwide collected Gram- macodynamic model. Antimicrob Agents Chemother 64. https://doi.org/
negative bacilli not susceptible to carbapenems. Antimicrob Agents Che- 10.1128/AAC.01764-20.
mother 64. https://doi.org/10.1128/AAC.01432-20. 46. Bhagunde P, Zhang Z, Racine F, Carr D, Wu J, Young K, Rizk ML. 2019. A
31. Bhagwat SS, Hariharan P, Joshi PR, Palwe SR, Shrivastava R, Patel MV, translational pharmacokinetic/pharmacodynamic model to characterize
Devanga Ragupathi NK, Bakthavatchalam YD, Ramesh MS, Soman R, bacterial kill in the presence of imipenem-relebactam. Int J Infect Dis 89:
Veeraraghavan B. 2020. Activity of cefepime/zidebactam against MDR 55–61. https://doi.org/10.1016/j.ijid.2019.08.026.
Escherichia coli isolates harbouring a novel mechanism of resistance 47. Wu J, Racine F, Wismer MK, Young K, Carr DM, Xiao JC, Katwaru R, Si Q,
based on four-amino-acid inserts in PBP3. J Antimicrob Chemother 75: Harradine P, Motyl M, Bhagunde PR, Rizk ML. 2018. Exploring the pharma-
3563–3567. https://doi.org/10.1093/jac/dkaa353. cokinetic/pharmacodynamic relationship of relebactam (MK-7655) in
32. Nelson K, Rubio-Aparicio D, Sun D, Dudley M, Lomovskaya O. 2020. In vitro combination with imipenem in a hollow-fiber infection model. Antimi-
activity of the ultrabroad-spectrum-beta-lactamase inhibitor QPX7728 crob Agents Chemother 62. https://doi.org/10.1128/AAC.02323-17.
against carbapenem-resistant Enterobacterales with varying intrinsic and 48. Hirsch EB, Ledesma KR, Chang KT, Schwartz MS, Motyl MR, Tam VH. 2012.
acquired resistance mechanisms. Antimicrob Agents Chemother 64. https:// In vitro activity of MK-7655, a novel beta-lactamase inhibitor, in combi-
doi.org/10.1128/AAC.00757-20. nation with imipenem against carbapenem-resistant Gram-negative

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 17


Perspective Antimicrobial Agents and Chemotherapy

bacteria. Antimicrob Agents Chemother 56:3753–3757. https://doi.org/ 65. Motsch J, Murta de Oliveira C, Stus V, Koksal I, Lyulko O, Boucher HW,
10.1128/AAC.05927-11. Kaye KS, File TM, Brown ML, Khan I, Du J, Joeng HK, Tipping RW, Aggrey
49. Bulitta JB, Hope WW, Eakin AE, Guina T, Tam VH, Louie A, Drusano GL, A, Young K, Kartsonis NA, Butterton JR, Paschke A. 2020. RESTORE-IMI 1: a
Hoover JL. 2019. Generating robust and informative nonclinical in vitro multicenter, randomized, double-blind trial comparing efficacy and safety
and in vivo bacterial infection model efficacy data to support translation of imipenem/relebactam vs colistin plus imipenem in patients with imi-
to humans. Antimicrob Agents Chemother 63. https://doi.org/10.1128/ penem-nonsusceptible bacterial infections. Clin Infect Dis 70:1799–1808.
AAC.02307-18. https://doi.org/10.1093/cid/ciz530.
50. Anonymous. 2016. PRIMAXIN (imipenem and cilastatin) for injection, for in- 66. Kohno S, Bando H, Yoneyama F, Kikukawa H, Kawahara K, Shirakawa M,
travenous use. Merck Sharp & Dohme Corp., Whitehouse Station, NJ, USA. Aoyama N, Brown M, Paschke A, Takase A. 2021. The safety and efficacy
https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/050587s074lbl of relebactam/imipenem/cilastatin in Japanese patients with complicated
.pdf. intra-abdominal infection or complicated urinary tract infection: a multi-
51. Drusano GL. 2004. Antimicrobial pharmacodynamics: critical interactions center, open-label, noncomparative phase 3 study. J Infect Chemother
of 'bug and drug'. Nat Rev Microbiol 2:289–300. https://doi.org/10.1038/ 27:262–270. https://doi.org/10.1016/j.jiac.2020.09.032.
nrmicro862. 67. Rebold N, Morrisette T, Lagnf AM, Alosaimy S, Holger D, Barber K, Justo
52. Craig WA. 1998. Pharmacokinetic/pharmacodynamic parameters: ration- JA, Antosz K, Carlson TJ, Frens JJ, Biagi M, Kufel WD, Moore WJ, Mercuro
ale for antibacterial dosing of mice and men. Clin Infect Dis 26:1–10. N, Raux BR, Rybak MJ. 2021. Early multicenter experience with imipenem-
https://doi.org/10.1086/516284. cilastatin-relebactam for multidrug-resistant Gram-negative infections.
53. European Medicines Agency. 2016. Guideline on the use of pharmacoki- Open Forum Infect Dis 8:ofab554. https://doi.org/10.1093/ofid/ofab554.
netics and pharmacodynamics in the development of antimicrobial me- 68. US Department of Health and Human Services, Food and Drug Adminis-
dicinal products (EMA/CHMP/594085/2015). Committee for Medicinal tration Center for Drug Evaluation and Research. 2014. Community-
Products for Human Use, London, UK. acquired bacterial pneumonia: developing drugs for treatment, guidance
54. Griffith DC, Sabet M, Tarazi Z, Lomovskaya O, Dudley MN. 2019. Pharma- for industry. Center for Drug Evaluation and Research, Silver Spring, MD.
cokinetics/pharmacodynamics of vaborbactam, a novel beta-lactamase 69. US Department of Health and Human Services, Food and Drug Adminis-
inhibitor, in combination with meropenem. Antimicrob Agents Chemo- tration Center for Drug Evaluation and Research. 2018. Guidance for
ther 63. https://doi.org/10.1128/AAC.01659-18. industry. Complicated intra-abdominal infections: developing drugs for
55. Berkhout J, Melchers MJ, van Mil AC, Seyedmousavi S, Lagarde CM, treatment. Center for Drug Evaluation and Research, Silver Spring, MD.
Schuck VJ, Nichols WW, Mouton JW. 2016. Pharmacodynamics of ceftazi- 70. US Department of Health and Human Services, Food and Drug Adminis-
dime and avibactam in neutropenic mice with thigh or lung infection. tration Center for Drug Evaluation and Research. 2015. Guidance for
Antimicrob Agents Chemother 60:368–375. https://doi.org/10.1128/AAC industry. Complicated urinary tract infections: developing drugs for treat-
.01269-15. ment. Center for Drug Evaluation and Research, Silver Spring, MD.
56. Singh R, Kim A, Tanudra MA, Harris JJ, McLaughlin RE, Patey S, O’Donnell JP, 71. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. 1985. APACHE II: a se-
Bradford PA, Eakin AE. 2015. Pharmacokinetics/pharmacodynamics of a verity of disease classification system. Crit Care Med 13:818–829. https://
doi.org/10.1097/00003246-198510000-00009.
beta-lactam and beta-lactamase inhibitor combination: a novel approach for
72. Kaye KS, Boucher HW, Brown ML, Aggrey A, Khan I, Joeng HK, Tipping
aztreonam/avibactam. J Antimicrob Chemother 70:2618–2626. https://doi
RW, Du J, Young K, Butterton JR, Paschke A. 2020. Comparison of treat-
.org/10.1093/jac/dkv132.
ment outcomes between analysis populations in the RESTORE-IMI 1
57. Crass RL, Pai MP. 2019. Pharmacokinetics and pharmacodynamics of
phase 3 trial of imipenem-cilastatin-relebactam versus colistin plus imipe-
beta-lactamase inhibitors. Pharmacotherapy 39:182–195. https://doi.org/
nem-cilastatin in patients with imipenem-nonsusceptible bacterial infec-
10.1002/phar.2210.
tions. Antimicrob Agents Chemother 64. https://doi.org/10.1128/AAC
58. Bhagunde P, Chang KT, Hirsch EB, Ledesma KR, Nikolaou M, Tam VH.
.02203-19.
2012. Novel modeling framework to guide design of optimal dosing strat-
73. Shields RK, Stellfox ME, Kline EG, Samanta P, Van Tyne D. 2022. Evolution
egies for beta-lactamase inhibitors. Antimicrob Agents Chemother 56:
of imipenem-relebactam resistance following treatment of multi-drug re-
2237–2240. https://doi.org/10.1128/AAC.06113-11.
sistant Pseudomonas aeruginosa pneumonia. Clin Infect Dis ciac097.
59. Patel M, Bellanti F, Daryani NM, Noormohamed N, Hilbert DW, Young K,
https://doi.org/10.1093/cid/ciac097.
Kulkarni P, Copalu W, Gheyas F, Rizk ML. 2021. Population pharmacoki-
74. Del Barrio-Tofino E, Lopez-Causape C, Oliver A. 2020. Pseudomonas aeru-
netic/pharmacodynamic assessment of imipenem/cilastatin/relebactam
ginosa epidemic high-risk clones and their association with horizontally-
in patients with hospital-acquired/ventilator-associated bacterial pneu- acquired beta-lactamases: 2020 update. Int J Antimicrob Agents 56:
monia. Clin Transl Sci 15:396–408. https://doi.org/10.1111/cts.13158. 106196. https://doi.org/10.1016/j.ijantimicag.2020.106196.
60. Yee KL, Kleijn HJ, Kerbusch T, Matthews RP, Dorr MB, Garey KW, Wrishko 75. Tamma PD, Beisken S, Bergman Y, Posch AE, Avdic E, Sharara SL,
RE. 2019. Population pharmacokinetics and pharmacodynamics of bezlo- Cosgrove SE, Simner PJ. 2021. Modifiable risk factors for the emergence
toxumab in adults with primary and recurrent Clostridium difficile infec- of ceftolozane-tazobactam resistance. Clin Infect Dis 73:e4599–e4606.
tion. Antimicrob Agents Chemother 63. https://doi.org/10.1128/AAC https://doi.org/10.1093/cid/ciaa1306.
.01971-18. 76. Simner PJ, Beisken S, Bergman Y, Posch AE, Cosgrove SE, Tamma PD.
61. Anonymous. 2016. PRIMAXIN (imipenem and cilastatin). US Prescribing 2021. Cefiderocol activity against clinical Pseudomonas aeruginosa iso-
information. Merck Sharp & Dohme Corp, Whitehouse Station, NJ, USA. lates exhibiting ceftolozane-tazobactam resistance. Open Forum Infect
62. Lucasti C, Vasile L, Sandesc D, Venskutonis D, McLeroth P, Lala M, Rizk ML, Dis 8:ofab311. https://doi.org/10.1093/ofid/ofab311.
Brown ML, Losada MC, Pedley A, Kartsonis NA, Paschke A. 2016. Phase 2, 77. Naseer S, Weinstein EA, Rubin DB, Suvarna K, Wei X, Higgins K, Goodwin
dose-ranging study of relebactam with imipenem-cilastatin in subjects A, Jang SH, Iarikov D, Farley J, Nambiar S. 2021. US Food and Drug Admin-
with complicated intra-abdominal infection. Antimicrob Agents Chemo- istration (FDA): benefit-risk considerations for cefiderocol (Fetroja(R)). Clin
ther 60:6234–6243. https://doi.org/10.1128/AAC.00633-16. Infect Dis 72:e1103–e1111. https://doi.org/10.1093/cid/ciaa1799.
63. Sims M, Mariyanovski V, McLeroth P, Akers W, Lee YC, Brown ML, Du J, 78. Poirel L, Sadek M, Kusaksizoglu A, Nordmann P. 2022. Co-resistance to
Pedley A, Kartsonis NA, Paschke A. 2017. Prospective, randomized, dou- ceftazidime-avibactam and cefiderocol in clinical isolates producing KPC
ble-blind, phase 2 dose-ranging study comparing efficacy and safety of variants. Eur J Clin Microbiol Infect Dis 41:677–680. https://doi.org/10
imipenem/cilastatin plus relebactam with imipenem/cilastatin alone in .1007/s10096-021-04397-x.
patients with complicated urinary tract infections. J Antimicrob Chemo- 79. Maraki S, Mavromanolaki VE, Moraitis P, Stafylaki D, Kasimati A,
ther 72:2616–2626. https://doi.org/10.1093/jac/dkx139. Magkafouraki E, Scoulica E. 2021. Ceftazidime-avibactam, meropenen-
64. Titov I, Wunderink RG, Roquilly A, Rodriguez Gonzalez D, David-Wang A, vaborbactam, and imipenem-relebactam in combination with aztreonam
Boucher HW, Kaye KS, Losada MC, Du J, Tipping R, Rizk ML, Patel M, against multidrug-resistant, metallo-beta-lactamase-producing Klebsiella
Brown ML, Young K, Kartsonis NA, Butterton JR, Paschke A, Chen LF. 2021. pneumoniae. Eur J Clin Microbiol Infect Dis 40:1755–1759. https://doi
A randomized, double-blind, multicenter trial comparing efficacy and .org/10.1007/s10096-021-04197-3.
safety of imipenem/cilastatin/relebactam versus piperacillin/tazobactam 80. O'Donnell JN, Putra V, Belfiore GM, Maring BL, Young K, Lodise TP. 2022.
in adults with hospital-acquired or ventilator-associated bacterial pneu- In vitro activity of imipenem/relebactam plus aztreonam against metallo-
monia (RESTORE-IMI 2 study). Clin Infect Dis 73:e4539–e4548. https://doi b -lactamase producing, OprD-deficient Pseudomonas aeruginosa with
.org/10.1093/cid/ciaa803. varying levels of Pseudomonas-derived cephalosporinase production. Int

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 18


Perspective Antimicrobial Agents and Chemotherapy

J Antimicrob Agents 59:106595. https://doi.org/10.1016/j.ijantimicag urinary tract infection samples: SMART Surveillance United States 2015–
.2022.106595. 2017. J Glob Antimicrob Resist 21:223–228. https://doi.org/10.1016/j.jgar
81. Le Run E, Atze H, Arthur M, Mainardi JL. 2020. Impact of relebactam-medi- .2019.10.028.
ated inhibition of Mycobacterium abscessus BlaMab beta-lactamase on 86. Karlowsky JA, Lob SH, Kazmierczak KM, Hawser SP, Magnet S, Young K,
the in vitro and intracellular efficacy of imipenem. J Antimicrob Chemo- Motyl MR, Sahm DF. 2018. In vitro activity of imipenem/relebactam
ther 75:379–383. https://doi.org/10.1093/jac/dkz433. against Gram-negative ESKAPE pathogens isolated in 17 European coun-
82. Lopeman RC, Harrison J, Rathbone DL, Desai M, Lambert PA, Cox JAG. 2020. tries: 2015 SMART surveillance programme. J Antimicrob Chemother 73:
Effect of amoxicillin in combination with imipenem-relebactam against 1872–1879. https://doi.org/10.1093/jac/dky107.
Mycobacterium abscessus. Sci Rep 10:928. https://doi.org/10.1038/s41598 87. Lapuebla A, Abdallah M, Olafisoye O, Cortes C, Urban C, Landman D,
-020-57844-8. Quale J. 2015. Activity of imipenem with relebactam against Gram-nega-
83. Yang TY, Hsieh YJ, Kao LT, Liu GH, Lian SH, Wang LC, Lin IL, Wang HY, tive pathogens from New York City. Antimicrob Agents Chemother 59:
Tseng SP, Lu PL. 2021. Activities of imipenem-relebactam combination 5029–5031. https://doi.org/10.1128/AAC.00830-15.
against carbapenem-nonsusceptible Enterobacteriaceae in Taiwan. J 88. Karlowsky JA, Lob SH, Young K, Motyl MR, Sahm DF. 2019. Activity of imi-
Microbiol Immunol Infect 55:86–94. https://doi.org/10.1016/j.jmii.2021.02 penem-relebactam against multidrug-resistant Pseudomonas aeruginosa
.001. from the United States—SMART 2015–2017. Diagn Microbiol Infect Dis
84. Yang Q, Zhang H, Yu Y, Kong H, Duan Q, Wang Y, Zhang S, Sun Z, Liao K, Gu 95:212–215. https://doi.org/10.1016/j.diagmicrobio.2019.05.001.
L, Jiang X, Wu A, Huang W, Shan B, Kang M, Hu F, Yu H, Zhang W, Xu Y. 2020. 89. Asempa TE, Nicolau DP, Kuti JL. 2019. Carbapenem-nonsusceptible Pseu-
In vitro activity of imipenem/relebactam against Enterobacteriaceae isolates domonas aeruginosa isolates from intensive care units in the United
obtained from intra-abdominal, respiratory tract, and urinary tract infections States: a potential role for new beta-lactam combination agents. J Clin
in china: study for monitoring antimicrobial resistance trends (SMART), 2015– Microbiol 57. https://doi.org/10.1128/JCM.00535-19.
2018. Clin Infect Dis 71:S427–S435. https://doi.org/10.1093/cid/ciaa1519. 90. Lob SH, Karlowsky JA, Young K, Motyl MR, Hawser S, Kothari ND, Gueny ME,
85. Karlowsky JA, Lob SH, Kazmierczak KM, Young K, Motyl MR, Sahm DF. Sahm DF. 2019. Activity of imipenem/relebactam against MDR Pseudomo-
2020. In vitro activity of imipenem/relebactam against Enterobacteria- nas aeruginosa in Europe: SMART 2015–17. J Antimicrob Chemother 74:
ceae and Pseudomonas aeruginosa isolated from intraabdominal and 2284–2288. https://doi.org/10.1093/jac/dkz191.

July 2022 Volume 66 Issue 7 10.1128/aac.00256-22 19

You might also like