Update of Clinical Application in Ceftazidime-Avibactam For Multidrug Resistant Gram Negative Bacteria Infections

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Infection (2022) 50:1409–1423

https://doi.org/10.1007/s15010-022-01876-x

REVIEW

Update of clinical application in ceftazidime–avibactam


for multidrug‑resistant Gram‑negative bacteria infections
Sisi Zhen1 · Hui Wang1,2 · Sizhou Feng1

Received: 22 April 2022 / Accepted: 14 June 2022 / Published online: 4 July 2022
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany 2022

Abstract
Purpose  Multidrug-resistant Gram-negative bacteria (MDR-GNB) have become a major global public health threat. Ceftazi-
dime–avibactam (CAZ–AVI) is a newer combination of β-lactam/β-lactamase inhibitor, with activity against carbapenem-
resistant Enterobacterales (CRE) and carbapenem-resistant Pseudomonas aeruginosa (CRPA). The aim of this review is to
describe the recent real-world experience of CAZ–AVI for the infections due to MDR-GNB.
Methods  We searched PubMed, Embase and Google Scholar for clinical application in CAZ-AVI for MDR-GNB infections.
Reference lists were reviewed and synthesized for narrative review.
Results  MDRGNB infections are associated with higher mortality significantly comparing to drug-susceptible bacterial
infections. Fortunately, CAZ–AVI shows significant benefits for infections due to KPC or OXA-48 CRE, comparing to
colistin, carbapenem, aminoglycoside and other older agents, even in those with immunocompromised status. The efficacy of
CAZ–AVI varies in different infection sites due to CRE, which is lower in pneumonia. Early use is associated with improved
clinical outcomes. Noteworthy, when adopted as salvage therapy, CAZ–AVI is still superior to other GNB active antibiot-
ics. CAZ–AVI plus aztreonam is recommended as the first line of MBL-CRE infections. However, for infections caused by
KPC- and OXA-48-producing isolates, further investigations are needed to demonstrate the benefit of combination therapy.
Besides CRE, CAZ-AVI is also active to MDR-PA. However, the development of resistance in CRE and MDR-PA against
CAZ–AVI is alarming, and more investigations and studies are needed to prevent, diagnose, and treat infections due to
CAZ–AVI-resistant pathogens.
Conclusions  CAZ-AVI appears to be a valuable therapeutic option in MDR-GNB infections. Using CAZ-AVI appropriately
to improve efficacy and decrease the emergence of resistance is important.

Keywords  Ceftazidime–avibactam · Carbapenem-resistant Enterobacterales · Multidrug-resistant Pseudomonas


aeruginosa · Multidrug-resistant Gram-negative bacteria

Introduction

Since initial emergence in 1980s, Enterobacterales produc-


ing extended-spectrum beta lactamase (ESBL) was reported
Sisi Zhen and Hui Wang are co-authors.
with increased frequency and became a major antimicrobial-
* Sizhou Feng resistant pathogen [1]. Carbapenem was the last resort of
szfeng@ihcams.ac.cn; doctor_szhfeng@163.com ESBL-producing Enterobacterales infections. However, the
extensive use of carbapenem led to a more serious problem:
1
State Key Laboratory of Experimental Hematology, the emergence of carbapenem-resistant Enterobacterales
National Clinical Research Center for Blood Diseases, Haihe
Laboratory of Cell Ecosystem, Institute of Hematology & (CRE). One of the most comprehensive continental surveys
Blood Diseases Hospital, Chinese Academy of Medical is the European Survey of Carbapenemase-Producing Enter-
Sciences, Peking Union Medical College, Tianjin 300020, obacterales (CPE) initiative, which reported 850 of 2301
China (37%) Klebsiella pneumoniae samples and 77 of 402 (19%)
2
Department of Hematology, The Affiliated Yantai E coli samples were carbapenemase (KPC, NDM, VIM, or
Yuhuangding Hospital of Qingdao University, OXA-48-like) producers. The infection prevalence of CPE
Yantai 264000, Shandong Province, China

13
Vol.:(0123456789)

1410 S. Zhen et al.

was shown to be 1.3 patients per 10,000 hospital admis- enzymes [18], but cannot inhibit the activity of metallo-β-
sions and 2.5 patients per 100,000 hospital patient-days lactamases (MBLs) [19]. The addition of avibactam restores
[2]. Similarly, multidrug-resistant Gram-negative bacteria ceftazidime activity against isolates previously resistant to
(MDR-GNB) other than CRE, such as carbapenem-resistant ceftazidime, including Enterobacterales and Pseudomonas
Pseudomonas aeruginosa (CRPA) or carbapenem-resistant aeruginosa. However, Acinetobacter baumannii strains are
Acinetobacter baumannii (CRAB), are also prevalent [3–6]. resistant to CAZ–AVI, because avibactam cannot inhibit
MDR-GNB infections were associated with higher mor- OXA-51, 23, 40, 24-like enzymes, which are always pro-
tality significantly comparing to drug-susceptible bacterial duced by Acinetobacter baumannii [20, 21]. Avibactam
infections. In the United States, it was estimated that more is renally excreted, and its pharmacokinetics are similar
than 2 million people suffered infections by multidrug-resist- to those of ceftazidime, allowing for coformulation [22].
ant organism each year and about 23,000 died as a direct CAZ–AVI was approved by FDA for the use in complicated
result of these infections [7]. A number of studies had been intra-abdominal infections (cIAI), complicated urinary tract
conducted to assess the outcomes of infections due to CRE infections (cUTI) including pyelonephritis, and hospital-
[8–12]. A meta-analysis suggested that mortality was higher acquired bacterial pneumonia/ventilator-associated bacte-
significantly in carbapenem-resistant Klebsiella pneumoniae rial pneumonia (HABP/VABP) with recommended dose
(CRKP) (466/1093, 42.6%) than carbapenem-susceptible of 2.5 g q8h for 2 h. According to the Infectious Diseases
Klebsiella pneumoniae patients (231/859, 26.9%) (OR 2.18, Society of America Guidance on the Treatment of Antimi-
95% CI 1.60–2.90) [13]. crobial-Resistant Gram-Negative Infections published in
Clinical outcomes of infections due to CRE or CRPA in 2022, CAZ–AVI was recommended as preferred treatment
hematological patients had been described [14–17]. Tre- for these infections: 1. pyelonephritis and cUTI caused by
carichi et al. [16] conducted a prospective study including CRE; 2. infections outside of the urinary tract caused by
278 episodes of Klebsiella pneumoniae bacteremia in 13 CRE; 3. KPC-producing CRE infections outside of the uri-
hematological units, and found that the overall mortality at nary tract and OXA-48-like-producing CRE infections if
21 days was significantly higher for patients with CRKP carbapenemase production is present; 4. difficult-to-treat
bacteremia than those with carbapenem-susceptible Kleb- infections due to Pseudomonas aeruginosa; furthermore,
siella pneumoniae bacteremia (84/161, 52.2% vs 17/117, CAZ–AVI together with aztreonam was recommended as
14.5%, P < 0.001). Similar outcomes were observed in 81 first-line therapy for MBL-producing CRE infections [23].
hematological malignancies patients infected with Pseu-
domonas aeruginosa [17]. Overall mortality rate at 30 days The clinical data in CRE infections
was significantly higher in patients with CRPA (n = 23) com-
paring to those with carbapenem-susceptible Pseudomonas Clinical efficacy comparing to other drugs
aeruginosa (n = 58) (60.9% vs 17.2%, P < 0.001).
Considering the increasing prevalence, higher mortality, Before newer β-lactam/β-lactamase inhibitors entered the
and longer stay of hospital, WHO identified CRE, CRPA, market, traditional drugs, including colistin, carbapenem,
and CRAB as critical priority pathogens. Because of lack- aminoglycoside, tigecycline monotherapy, or combination
ing alternative drugs, patients are often dead without effec- therapy, were common choices for CRE infections. Till now,
tive treatment, indicating that it is of utmost importance to one prospective and several retrospective cohorts have been
choose newer agents against them. Ceftazidime–avibactam conducted to compare the efficacy between CAZ–AVI and
(CAZ–AVI) is one of the new antibiotics approved by FDA colistin exclusively in different countries, among which
and EMA based on the promising outcomes in clinical trials. bacteremia and HABP were the most common types of
In this review, we describe the actual evidence of CAZ–AVI infections [24–27]. The first prospective, multicenter study
in patients with infections due to CRE and multidrug-resist- comparing CAZ–AVI to colistin for CRE infections was
ant Pseudomonas aeruginosa (MDR-PA), aiming to provide conducted by Van Duin et al. involving 137 patients [24].
insights into the use of CAZ–AVI for these infections. Bacteremia (n = 63, 46%) and respiratory infections (n = 30,
22%) were most common types. Among 54 CRKP isolates
tested for carbapenemase gene, 28 (52%) and 24 (44%)
The clinical data of ceftazidime–avibactam were positive for ­blaKPC-2 and ­blaKPC-3. 98 CRE isolates
were tested the susceptibility to colistin. The susceptible
CAZ–AVI is a new β-lactam/β-lactamase inhibitor combi- rates to colistin in CAZ–AVI group and colistin group were
nation. Different from other β-lactamase inhibitor, avibac- 77% (23/30) and 93% (63/68), respectively. IPTW-adjusted
tam is a novel non-β-lactam β-lactamase inhibitor, which all-cause hospital mortality at day 30 was 9% versus 32%
can inhibit ESBL, Ambler class A (GEM, SHV, CTX-M (difference, 23%, 95% CI, 9% ~ 35%, P = 0.001) in patients
and KPC), class C (AmpC), and some class D (OXA-48) treated with CAZ–AVI and colistin. Almangour et al. [26]

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1411

conducted a larger retrospective, multicenter study including mortality significantly than comparator (21.9%, 16/73
230 patients with CRE infections. The most common infec- vs 46.9%, 23/49, P = 0.0004). This study described less
tion type was HABP and VABP (n = 82, 36%), followed by adverse events in CAZ–AVI group (11/189, 5.8% vs 30/150,
UTI (n = 44, 19%), wound infection (n = 37, 16%), and IAI 20%, P < 0.001). Gu et al. [30] also reported the advantage
(n = 30, 13%). Carbapenemase type was reported for 49% of CAZ–AVI than others for CRE infections in a single-
(73/149) and 46% (37/81) isolates in CAZ–AVI and colistin center, retrospective study. The study included 90 infections
group, in which 98% were b­ laOXA-48. If isolates were identi- due to CRKP. Forty two were treated with CAZ–AVI and
fied as non-susceptible to the study drug, the patient was forty eight were treated with others (carbapenem, colis-
excluded. Clinical cure (71% vs 52%, P = 0.004, OR: 2.29, tin, tigecycline, fosfomycin, fluroquinolone). The crude
95% CI 1.31–4.01) was significantly higher in CAZ–AVI 30-day mortality (8/42, 19.0% vs 22/48, 45.8%, P = 0.007)
group than colistin-based group. Mortality was lower (35% and 14-day clinical failure (14/42, 33.3% vs 24/48, 50.0%,
vs 44%, P = 0.156, OR: 0.67, 95% CI 0.39–1.16) in patients P = 0.046) were lower in CAZ–AVI group. Moreover, the
treated with CAZ–AVI. As for the safety, acute kidney injury subgroup analysis showed CAZ–AVI was still associated
(15% vs 33%, P = 0.002, OR 0.37, 95% CI 0.19–0.69) was with decreased 30-day mortality in the bacteremia (OR 0.23,
significantly less common in patients receiving CAZ–AVI. 95% CI 0.08–0.63, p = 0.004), septic shock (OR 0.23, 95%
Although larger, better designed studies are needed, for CI 0.07–0.78, p = 0.019), SOFA score > 5 (OR 0.13, 95%
infections due to KPC or OXA-48 CRE, especially for CI 0.04–0.36, P < 0.000), mechanical ventilation (OR 0.13,
colistin resistant strains infections, CAZ–AVI is a reason- 95% CI 0.04–0.36), and Charlson comorbidity index (> 3,
able alternative to colistin. However, for MBL-CRE, which OR 0.15, 95% CI 0.04–0.55, P = 0.004). Available data indi-
is naturally resistant to CAZ–AVI, CAZ–AVI monotherapy cate that CAZ–AVI is associated with improved clinical and
is not recommended. microbiology outcomes for KPC or OXA-48 CRE infections
When comparing to other antibiotics such as carbap- comparing to traditional drugs and may be more valuable
enem, aminoglycoside and so on, similar outcomes were for severe cases than mild cases. (The clinical data about
observed [28–34]. One meta-analysis encompassed 12 stud- comparative efficacy between CAZ–AVI and others are sum-
ies involving 4951 patients evaluating the efficacy and safety marized in Table 1.)
of CAZ–AVI for GNB infections [35]. In the subgroup anal- The efficacy between CAZ–AVI and other newer
ysis of causative pathogen, CAZ–AVI demonstrated a higher β-lactam/β-lactamase inhibitors for the treatment of CRE
clinical response (RR 1.61, 95% CI 1.13–2.29, I2 = 61.7%) infections is similar. Ackley et al. [36] conducted a multi-
and a lower mortality (RR 0.29, 95% CI 0.13–0.63, I2 = 0) center, retrospective cohort study to compare the efficacy of
than control treatment (carbapenem, colistin, tigecycline, CAZ–AVI (n = 105) and meropenem–vaborbactam (n = 26)
and so on) for infections caused by CRE. Moreover, in the for CRE infections. 61% of the patients in the CAZ–AVI
subgroup analysis by infection type, CAZ–AVI was supe- group received combination therapy (with aminoglycosides,
rior in clinical response for bacteremia from two studies colistin, tigecycline, and others) and 15% in the merope-
(140 patients) (RR 2.11, 95% CI 1.54–2.88, I2 = 0) and in nem–vaborbactam group, respectively. Among 32 and
microbiological eradication in cUTI from three studies 13 isolates tested for genetic mechanism in CAZ–AVI
(1186 patients) (RR 1.13, 95% CI 1.05–1.21, I2 = 43.6%). and meropenem–vaborbactam group, 23 (71.9%) and 10
Castón et al. [28] conducted the largest comparative series (76.9%) were positive for KPC. Clinical success at day 30
of patients (n = 339) with infections caused by CPE treated (61.9% vs 69.2%, P = 0.49), mortality at day 30 (19.1% vs
with CAZ–AVI or best available therapy (BAT). The most 11.5%, P = 0.57), and mortality at day 90 (28.6% vs 26.9%,
common type of carbapenemase was OXA-48 (n = 255), fol- P = 0.48) were similar between CAZ–AVI group and mero-
lowed by KPC (n = 84). UTI was the most frequent type of penem–vaborbactam group. Development of resistance
infection (n = 129, 38.1%), followed by bloodstream infec- occurred in three patients who received CAZ–AVI mono-
tion (BSI) (n = 111, 32.7%), IAI (n = 60, 17.7%), and pneu- therapy and in none-patients in the meropenem–vaborbac-
monia (n = 39, 11.5%). Although CAZ–AVI was always tam group.
used as monotherapy than BAT (133/189, 70.4% vs 64/150, Totally, although more data are needed, CAZ–AVI offers
42.6%, P < 0.001), the 30-day crude mortality was signifi- therapeutic advantages comparing to other traditional anti-
cantly lower in CAZ–AVI group (26/189, 13.7% vs 33/150, biotics for the treatment of KPC- and OXA-48-producing
22.0%, P = 0.04). In multivariate analysis, CAZ–AVI-con- Enterobacterales infections, especially for severe patients.
taining regimen was the only independent factor of 21-day
clinical response (OR 2.43, 95% CI 1.16–5.12, P = 0.02) and Clinical experience in patients with pneumonia
microbiological eradication (OR 0.40, 95% CI 0.18 ~ 0.85,
P = 0.02). In patients with INCREMENT-CPE scores > 7 The clinical and microbiological outcomes among
points, CAZ–AVI treatment was associated with lower patients treated with CAZ–AVI for CRE infections vary in

13

Table 1  Overview of retrospective/prospective comparative/case–control studies involving CAZ–AVI for the treatment of CRE infections
1412

Publication Design Number of patients (% with combination) No. (%) and Baseline pathogens Resistance mecha- Outcomes (No. %) (CAZ–AVI vs. compara-
sources of infec- nisms tor)

13
tion
CAZ–AVI Comparator Clinical cure/success Mortality

van Duin 2018 [24] Multicenter, pro- 38 (NR) 99 colistin (NR) BSI 46%, K. pneumoniae 97%; KPC 38%, NR Day 30: 3/38 (8%)
spective Pneumonia 22%, Enterobacter spp. 3% non-carbapenemase vs 33/99 (33%),
UTI 14%, producing 1%; P = 0.64
Wound 10%, NR 61%,
Others 8%
Hakeam 2021 [25] Multicenter, retro- 32 (72%) 29 colistin (100%) 100% BSI K. pneumoniae 79%; OXA-48 51%; 15/32 (47%) vs 6/29 Day 30: 12/32 (38%)
spective E coli. 11%; NDM 21%; (21%), P = 0.049 vs 12/29 (41%),
Other 10% KPC 10%; P = 0.75
NR 18%
Fang 2021 [27] Multicenter, retro- 37 (70%) 78 colistin (88%) Pneumonia 57%; K. pneumoniae 100% NR 19/37 (51%) vs 9/78 Day 28: 3/37 (8%)
spective BSI 50%; (12%) vs 23/78 (29%),
IAI 28%; P = 0.013
UTI 10%;
Others 13%
Almangour 2022 Multicenter, retro- 149 (23%) 81 colistin (70%) HAP/VAP 36%; K. pneumoniae 87%; OXA-48 47%; 106/149 (71%) vs In hospital mortal-
[26] spective BSI 26%; E coli. 9%; KPC 1%; 42/81 (52%), ity: 52/149 (35%)
UTI 19%; Other 3% NR 52% P = 0.004 vs 36/81 (44%),
Wound 16%; P = 0.156
IAI 13%;
Others 13%
Shi 2021 [40] Sing center, retro- 43 (79%) 62 tigecycline (94%) HAP/VAP 100% K. pneumoniae 100% NR 22/43 (51%) vs Day 28: 13/43 (30%)
spective 18/62 (29%), vs 21/62 (34%),
P = 0.022 P = 0.695
Ackley 2020 [36] Multicenter, retro- 105 (61%) 26 MV (15%) BSI 40%; Klebsiella spp. 67%; KPC 24%; 65/105 (62%) vs Day 30: 20 /105
spective Non-BSI: Enterobacter spp. NR 76% 18/26 (69%), (19%) vs 3 /26
Pneumonia 30%; 21%; P = 0.49 (12%), P = 0.57;
SSTI 15%; E coli. 9%; Day 90: 30 /105
IAI 13%; Other 3% (29%) vs 7 /26
Others 1% (27%), P = 0.48
Tumbarello 2019 Multicenter, retro- 138 (79%) (104 104 (74%) BSI 86%, K. pneumoniae 100% KPC 100% NR Day 30: 47/138
[38] spective, matched matched) Pneumonia 5%, (34%);
IAI 5%, 38/104 (37%) vs
2%, 58/104 (56%),
others 1% P = 0.005
Karaiskos I (2021) Multicenter prospec- 147 (71 matched) 71 (NR) BSI 76%, K. pneumoniae 100% KPC 97%, Day 14: 119/147 Day 28:30/147
[53] tive, matched (54%) UTI 15%, OXA-48 3% (81%) (20%);
HAP/VAP 17%, 13/71 (18%) vs 29/71
IAI 7%, (41%), P = 0.005
others 5%
S. Zhen et al.
Table 1  (continued)
Publication Design Number of patients (% with combination) No. (%) and Baseline pathogens Resistance mecha- Outcomes (No. %) (CAZ–AVI vs. compara-
sources of infec- nisms tor)
tion
CAZ–AVI Comparator Clinical cure/success Mortality

Shields 2017 [29] Single center, retro- 13 (38%) 96 (70%) BSI 100% K. pneumoniae 100% KPC 97%; Day 30: 11/13 (85%) Day 30: 1/13 (8%)
spective NR 3% vs 39/96 (41%), vs 30/96 (31%),
P = 0.006 P = 0.1;
Day 90: 1/13 (8%)
vs 43/96 (45%),
P = 0.01
Alraddadi 2019 [31] Single center, retro- 10 28 (89%) BSI 58%, K. pneumoniae 79%, OXA-48 71%, Day 30: 4/10 (40%) Day 30: 5/10 (50%)
spective HAP 50%; E coli. 21% NDM 16%, vs 11/28 (39%), vs 16/28 (57%),
UTI 29%; NDM + OXA-48 P > 0.99 P = 0.7
IAI 21%, 3%,
SSTI 13% non-carbapenemase
producing 11%
Tsolaki 2020 [33] Two centers, retro- 41 (78%) 36 (97%) BSI 65%, K. pneumoniae 100% KPC 94%; 33/41(80%) vs Day 28: 6/41 (15%)
spective VAP 34%, NR 6% 19/36(53%), vs 14/36 (39%),
IAI 10%, P = 0.01 P = 0.035
UTI 4%,
Others 3%
Gu 2021 [30] Sing center, retro- 42 (62%) 48 (NR) Pneumonia 74%; K. pneumoniae 100% NR 28/42 (67%) vs Day 30: 8/42 (19%)
spective BSI 50%; 24/48 (50%), vs 22/48 (46%),
UTI 7%; P = 0.046 P = 0.007
CNS 6%
IAI 2%;
SSTI 4%
Chen 2021 [32] Multicenter, retro- 35 (NR) 152 (NR) BSI 100% Enterobacterales NR 25/35 (71%) vs Day 30: 6/35 (17%)
spective 100% 48/152 (32%), vs 72/152 (47%),
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative…

P = 0.002
Shen 2021 [34] Sing center, retro- 9 (56%) 80 (31%) BSI 100% K. pneumoniae 100% NR NR Day 28: 2/9 (22%) vs
spective 40/80 (50%)
Castón 2022 [28] Multicenter, retro- 189 (30%) 150 (57%) UTI 38%; K. pneumoniae 92%; OXA-48 75%; 169/189 (89%) vs Day 30: 26/189
spective BSI 33%; Enterobacter spp. 4%; KPC 25% 119/150 (79%), (14%) vs 33/150
IAI 18%; E. coli. 3%; P = 0.01 (22%), P = 0.04
Pneumonia 12% Other 1%
Caston 2017 [41] Multicenter, retro- 8 (NR) 23 (74%) BSI 100% K. pneumoniae 81%; OXA-48 61%; 6/8 (75%) vs 8/23 Day 30: 2/8 (25%),
spective (hemato- Other 19% KPC 39% (35%), P = 0.031 vs 12/23 (52%),
logical patients) P = 0.19

CAZ–AVI ceftazidime–avibactam, CRE carbapenem-resistant Enterobacterales, NR unreported; BSI: bloodstream infection, UTI urinary tract infection, KP Klebsiella pneumonia, K. pneumo-
niae Klebsiella pneumonia, E. coli Escherichia coli, HAP/VAP hospital-acquired pneumonia/ventilator-associated pneumonia, SSTI skin and soft-tissue infection, IAI intra-abdominal infection,
MV meropenem–vaborbactam

13
1413

1414 S. Zhen et al.

different types of infection. In previous studies, pneumonia in those with febrile neutropenia. CAZ–AVI was adminis-
or mechanical ventilation had been illustrated to be a risk tered in 10 of 109 patients who received hematopoietic stem
factor for CAZ–AVI treatment failure [37–39]. Preclinical cell transplantation for empirical or definitive therapy. They
human studies (phase I) suggested that CAZ–AVI pen- found that CAZ–AVI was effective in bacteremia clearance.
etrates in the long with PK patterns profile similar between We conducted a retrospective cohort in our hospital, includ-
epithelial lining fluid and plasma, and the epithelial lining ing 47 hematological patients with CRE or CRPA BSI. A
fluid/plasma ratio of CAZ–AVI concentrations ranges from favorable survival rate was observed as 83.8% in 37 patients
0.31 to 0.35. Although the epithelial lining fluid exposures with CRE BSI, supporting more clinical experiences for the
of CAZ–AVI exceed level required for efficacy in plasma, application of CAZ–AVI in hematological diseases patients
there is no evidence to support similar exposures in pneu- (the data are being published).
monia patients, especially in those with mechanical ventila- Real-world clinical practice of using CAZ–AVI for
tion. This study also found that epithelial lining fluid con- CRE infections in solid organ transplantation receptors
centrations increased dose-proportionally for both drugs, was also described recently in several observational stud-
with twofold increase in area under curve for patients who ies and case series [43–46]. Zhang et al. [44] compared
received 3 g ceftazidime plus 1 g avibactam comparing the efficacy of CAZ–AVI and other antibiotics for CRKP
to who received 2 g ceftazidime plus 0.5 g avibactam. If infection after kidney transplantation and found patients
higher doses of CAZ–AVI are more effective in pneumonia, receiving CAZ–AVI had lower 30-day mortality (3/22,
patients still need evaluated. 13.6% vs 14/32, 43.8%, P = 0.019) and higher 14-day clini-
Fortunately, when comparing to tigecycline-based cal cure (18/22, 81.8% vs 17/32, 53.1%, P = 0.030) signifi-
regimen, CAZ–AVI still shows better clinical outcomes cantly. Chen et al. [45] included 10 (9 infected with CRKP
for patients with pneumonia due to CRE. Shi et al. [40] and 1 with CRPA) lung transplant recipients treated with
conducted a retrospective cohort to compare the efficacy CAZ–AVI. The 30-day clinical survival was high to 100%
between CAZ–AVI-based regimen (n = 43) and tigecycline- and nine patients achieve negative of microbiological culture
based regimen (n = 62) for HABP or VABP due to CRKP in with a median time of 6.7 days. Another study reported that
intensive-care units. Most population received CAZ–AVI the clinical response at 30 days among liver transplanta-
or tigecycline combination therapy (79.1% vs 93.5%, tion recipients treated with CAZ–AVI as salvage therapy
P = 0.027). Although there was no significant difference in for CRKP infections was 58.8% (10/17) [43]. It seems that
28-day survival rates between the two groups, CAZ–AVI CAZ–AVI can be a promising choice for CRE infections,
regimen was significantly associated with 28-day clinical even in those with impairment of immune system. Larger
success (OR 3.405, 95% CI 1.304–8.889, P = 0.012) and multicenter studies are needed to be conducted to solidate
microbiological success (OR 7.778, 95% CI 2.717–22.265, this.
P < 0.001) in propensity score analysis.
CAZ–AVI-based therapy for pneumonia due to CRE The optimization of usage
shows lower clinical success comparing to other types of
infections, while comparing to tigecycline, it seems to be a CAZ–AVI monotherapy or  CAZ–AVI‑based combination
better choice. The optimal dose for pneumonia or those with therapy  Prior to the introduction of CAZ–AVI into the
mechanical ventilation needs further exploration. market, combination therapy with two or more antibiotics
with in vitro activity was deemed to be superior to mono-
Clinical experience in special populations therapy, especially in those with septic shock or high mor-
tality scores. While for CAZ–AVI-containing regimen,
Patients with hematological malignancies and solid organ monotherapy supports similar efficacy as combination with
transplantation receptors are in high risk of severe infections traditional drugs (carbapenem, colistin, and so on) for infec-
because of impairment of the immune system and frequent tions due to KPC- or OXA-48-producing Enterobacterales
neutropenia. True-world clinical experience considering the according to available experience [37, 47–50]. In a recent
experience of CAZ–AVI in hematological patients is limited meta-analysis, including 13 studies and 503 patients with
till now. A multicenter, retrospective study conducted by carbapenem-resistant pathogens infections, there was no dif-
Castón et al. [41] retrospectively reported the outcomes of 31 ference in mortality regarding CAZ–AVI monotherapy and
hematological patients infected with CRE BSI. The 14-day CAZ–AVI-based combination therapy (OR 0.96, 95% CI
clinical success rate (75.0% vs 34.8%) and 30-day survival 0.65–1.41) [51]. Tumbarello et al. [52] conducted the larg-
rate (75.0% vs 47.8%) of 8 patients in CAZ–AVI group were est retrospective study involving 577 adults infected with
higher than the other patients (n = 23) in the control group. KPC-KP. Bacteremia was the most frequent type of infec-
Clerici et al. [42] conducted a retrospective study including tion (n = 391), and non-bacteremia included cUTIs (n = 71),
126 hematological patients and evaluate CAZ–AVI efficacy pneumonia (n = 59), IAIs (n = 35), and others (n = 21). Mor-

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1415

tality at day 30 was 25.3% (146/577), which did not differ with higher proportion of MBL-CRE (The data are being
between those with CAZ–AVI alone and those with combi- published.)
nation therapy (3/165, 26.1% vs 103/412, 25.0%, P = 0.79). Moreover, combination therapy may decrease the recur-
Notably, CAZ–AVI combination therapy was more fre- rence of CRE infections and CAZ–AVI resistance. Ackley
quently in patients with higher Charlson comorbidity et al. [36] retrospectively included 105 patients treated with
indexes and in those with pneumonia significantly (P < 0.01 CAZ–AVI for CRE infections. The recurrence of CRE infec-
for both). A prospective cohort also described similar out- tions (6/64, 9.4% vs 9/41, 22.0%) and CAZ–AVI resistance
come when comparing CAZ–AVI-containing monotherapy (0/64, 0% vs 3/41, 7.3%) were numerically lower in combi-
and combination therapy [53]. On the contrary, a retrospec- nation therapy group than monotherapy group.
tive cohort performed at two tertiary hospitals in China first In conclusion, CAZ–AVI plus aztreonam shows signifi-
reported that 30-day mortality rates for patients receiving cant dominance in clinical efficacy for MBL-CRE infections.
CAZ–AVI-based combination therapy were significantly For KPC- or OXA-48 CRE infections, whether combination
lower than those with CAZ–AVI monotherapy (10/41, CAZ–AVI with an in vitro non-susceptible antimicrobial
24.4% vs 11/21, 47.6%, P = 0.028) for CRKP infection [54]. drug could have better clinical effectiveness than CAZ–AVI
The multivariate analysis still revealed that combination monotherapy is controversial. Well-designed prospective
with another in vitro non-susceptible antimicrobial, such as studies or randomized control trials with more participants
carbapenems, tigecycline, and fosfomycin, was significantly should be designed for further investigation.
associated with lower 30-day mortality (HR 0.167, 95% CI
0.060–0.465, P = 0.001). Considering the existing of con- Timing of CAZ–AVI therapy  As is known, appropriate and
founders such as illness severity and types of infections, timely treatment is critical to patients with bacteremia [62].
more evidence is required to explore the potential benefit of A retrospective study enrolling 102 KPC-CRE bacteremia
combination for KPC- or OXA-48 CRE infections. found that the survivors had shorter median time (8.5 h vs
For MBL-producing strains infections, clinical data 48 h, P = 0.014) from blood cultures collection to appropri-
have shown promising outcomes with the combination of ate antibiotic therapy than those who died [63]. Receipt of
CAZ–AVI and aztreonam in several case reports and small appropriate treatment within 48 h decreased 30-day mortal-
cohort studies [55–60]. Falcone et al. [61] conducted the first ity rate (29.1% vs 63.8%, P < 0.001), and was independently
prospective study involving 102 patients with bacteremia negatively associated with the overall 30-day mortality by
due to MBL-producing Enterobacterales, with 82 produc- Cox regression analysis (HR 1.382, 95% CI 1.132–1.687,
ing NDM and the others producing VIM. 51.0% (n = 52) of P = 0.001). Consistent with these reports, Jorgense et al. [64]
patients receiving CAZ–AVI plus aztreonam, while 49.0% demonstrated receiving CAZ–AVI within 48 h of infection
(n = 50) are receiving other active antibiotics consisting of onset was associated with lower clinical failure (OR 0.409,
colistin, tigecycline, aminoglycosides, and carbapenem. 95% CI 0.180–0.930, P = 0.0329) for patients infected with
The 30-day mortality (10/52, 19.2% vs 22/50, 44.0%, MDR-GNB. Therefore, to improve the prognosis of infec-
P = 0.007) and acute kidney injury (1/52, 1.9% vs 10/50, tions due to CRE, it is of great importance to use appropri-
20.0%, P = 0.003) were significantly lower in CAZ–AVI ate antibiotics including CAZ–AVI as soon as possible after
plus aztreonam group than comparative group. A PS-based blood culture collection.
matched analysis including 50 pairs of patients reported that Although early use is critical, it is noteworthy that after
the regimen of CAZ–AVI plus aztreonam was associated failure of other regimens, CAZ–AVI as salvage therapy is
with a lower mortality rate at day 30 (HR 0.31, 95% CI still associated with clear benefits comparing to other com-
0.15–0.66, P = 0.002), lower clinical failure at day 14 (OR mon regimens. Tumbarello et al. [38] reviewed 104 patients
0.36, 95% CI 0.18–0.70, P = 0.003), and shorter length of infected with bacteremia due to KPC-KP, who receiving
hospital stay (HR 0.48, 95% CI 0.29–0.78, P = 0.003) com- CAZ–AVI as a salvage regimen after a first-line treatment
pared to other active antibiotics. The clinical data support with other antimicrobials. CAZ–AVI was administrated with
that CAZ–AVI plus aztreonam is a potential therapeutic other active antibiotics in 82 patients. When comparing to a
choice in MBL-producing isolates infections. The data in matched cohort involving 104 KPC-KP bacteremia treated
our hospital indicated that MBL-CRE accounted for 56% with drugs other than CAZ–AVI, the data indicated that
(n = 37) in 66 CRE isolated from blood and tested by PCR receipt of CAZ–AVI was associated with a lower 30-day
for identification of carbapenemase genes. In terms of the mortality significantly (36.5% vs 55.7%, P = 0.005). Mul-
types of isolates, the proportions of MBL-CRE were 77% tivariate analysis of the 208 cases of KPC-KP bacteremia
(24/31), 33% (9/27), 43% (3/7) and 100% (1/1) in Escheri- identified receipt of CAZ–AVI as the sole independent pre-
chia coli, K. pneumonia, Enterobacter cloacae, and Raoul- dictor of survival. Even among patients with single-drug
tella planticola, respectively. Therefore, combination with salvage regimens (n = 49), CAZ–AVI group still displayed
aztreonam actively is important, especially in those hospitals lower 30-day mortality (9/22, 40.9% vs 21/27, 77.8%,

13

1416 S. Zhen et al.

P = 0.008). Guimarães et al. [65] also demonstrated that reported. After exposure to CAZ–AVI, 92 isolates were
CAZ–AVI was a promising salvage therapy for KPC-KP reported, in which 55 were detected for the mechanism of
infections. CAZ–AVI as salvage therapy for KPC-KP infec- resistance to CAZ–AVI. Acquired resistance was mostly
tions was superior to alternative drugs, even in patients with associated with substitution of D179Y in KPC-3 (n = 32) or
bacteremia. KPC-2 (n = 5), alone or together with other substitutions or
Similarly, favorable outcomes were observed in patients resistance determinants.
with OXA-48-CRE infections when using CAZ–AVI as Reported mechanisms of resistance in CRE include enzy-
salvage therapy. Sousa et al. [66] prospectively enrolled matic resistance causing antibiotics inactivation, membrane
57 patients with OXA-48-CRE infections, who received impermeability of porin mutation, or increased efflux activ-
CAZ–AVI after previous treatment failure. Most patients ity and target protein mutations [87]. As is mentioned above,
received CAZ–AVI as monotherapy (46/57). Sepsis or septic numerous mutations in b­ laKPC genes, especially b­ laKPC-3 and
shock occurred in 31 (54%) patients. Clinical and micro- ­blaKPC-2 genes conferring CAZ–AVI resistance, have been
biological cure were achieved in 77% and 65% of patients. published. Most of KPC variants reported have substitutions,
Mortality rates at 14 days and 30 days were 14% and 22%, insertions or deletions in four loops, including the Ω-loop
respectively. Six patients experienced recurrence of OXA- (Arg164 to Asp179), the Val240 loop located between β3-
48-CRE infections within 90 days from the end of treatment, and β4-strand (Cys238 to Thr243), and the Lys270 loop
without increasing MIC (the minimum inhibitory concentra- located between the β5-strand, the α11- helix (Ala267 to
tion) to CAZ–AVI comparing to the original isolates. After Ser275), and Trp105 loop. The Ω-loop region in CAZ–AVI
retreated with CAZ–AVI monotherapy, all but one patient is an important activity site of lactamase [88]. The muta-
achieved clinical and microbiological cure. The remain- tions of KPC-3 or KPC-2 occurring at 164–179 site, such
ing patient, who had a complicated sternal osteomyelitis, as Asp179Tyr, Asp179Ala, Asp164Ser, and so on, result in
was clinically and microbiologically cured following three flexibility of the Ω-loop region, which decrease the bind-
courses with CAZ–AVI monotherapy. Moreover, none of ing of avibactam and ceftazidime hydrolysis activity, lead-
these episodes developed resistance to CAZ–AVI. ing to CAZ–AVI resistance [89]. Besides KPC, amino acid
Early use of CAZ–AVI is associated with improved substitutions in other Class A β-lactamase such as CTX-M-
clinical outcomes in CRE infections. If other antimicrobials 14 and VEB-1 also contribute to CAZ–AVI resistance [78,
had failed, CAZ–AVI as salvage therapy still shows clear 90]. Class B β-lactamase is naturally resistant to CAZ–AVI,
benefits in KPC- and OXA-48-producing Enterobacterales because avibactam could not inhibit the activity of MBLs
infections. [19]. Chromosome- or plasmid-mediated AmpC β-lactamase
mutations have been reported in CRE. The changes in the
Limitation: the emergence of resistance to CAZ–AVI Ω-loop region (such as amino acids 168, 176, 309–314, and
366) cause resistance in C. freundii and E. cloacae [91].
As is shown above, CAZ–AVI brings large clinical benefits Structural alterations in or surrounding the R2 binding site
for the CPE infections. However, as the popular use of this lead to non-susceptibility in E. coli [92]. As for Class D-β-
drug, resistance to CAZ–AVI has been reported in multi- lactamase, avibactam can inhibit OXA-48, but cannot inhibit
ple centers. The overall resistance rate after exposure to some OXA-48 variants (OXA-163, OXA-405 and so on)
CAZ–AVI ranged between 1.4 and 12.8% in studies enroll- [93]. Permeability defects related to non-functional porins
ing more than 20 patients with CRE infections [36–39, 48, also contribute to the resistance. Take K. pneumoniae for
52, 53, 67]. To support more information of treatment- example, OmpK 35 and OmpK 36 allow diffusion of avi-
associated resistance to CAZ–AVI, we collected available bactam across the outer membrane, while mutation of the
literature online, including 8 cohorts and 18 case reports OmpK 35/36 gene invalidates [94]. However, this resistance
[36–39, 48, 52, 53, 67–86]. mechanism alone cannot increase MIC of CAZ–AVI. Only
Seventy-one CRE (susceptible to CAZ–AVI) were together with other mechanisms, mutation of porins can sig-
observed developing resistance to CAZ–AVI after exposure nificantly increase the MIC to CAZ–AVI [95]. The presence
to this drug till now, which harbored KPC-3 (n = 30) mostly, of penicillin binding protein 3 formed by four amino acid
followed by KPC-2 (n = 11), and the rest of isolates (n = 30) insertion is also associated with decreasing CAZ–AVI sus-
were not reported for subtypes of KPC. The isolates before ceptibility in E. coli [96].
developing resistance to CAZ–AVI mainly belonged to The infection due to CAZ–AVI-resistant isolates becomes
ST258 (n = 13) and ST512 (n = 12). Other strains belonged a concerning issue with higher mortality. As was reported
to ST39 (n = 5), ST151 (n = 2), ST307 (n = 2), ST11 (n = 2), by Tumbarello and colleagues [52], 30-day mortality rates
and ST101 (n = 1). The ST strains were not reported in 34 among patients infected with CRE who developed resist-
isolates. Regarding to the species, most were KP (n = 67), ance and those susceptible to CAZ–AVI were 45% (9/20)
and 1 was Citrobacter freundii, while 3 isolates were not and 24% (137/557), respectively. Lacking valuable choice

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1417

is a concerning problem. Notably, the mutation of D179Y this drug. Isolates belonging to ST258 or ST512, or pro-
restored susceptibility to carbapenems. Theoretically, car- ducing KPC, especially KPC-3, are more likely to develop
bapenems can be a choice of these infections. However, resistance to CAZ–AVI after exposure to this drug. Preferred
this is not recommended, because the susceptibility to car- choice for infections due to CAZ–AVI-resistant pathogens
bapenem is unstable, which can be rapidly restored under has not been determined. Optimizing usage of CAZ–AVI
the selective pressure of carbapenem. Some suggested dual to prevent the emergence of resistance and improving sur-
carbapenem and CAZ–AVI therapy, while the clinically effi- veillance methods is necessary. For isolates without prior
cacy and safety of this regimen is not determined. Athans exposure to CAZ–AVI but developing resistance, further
et al. [72] reported that one case was clinically cured using investigation is warranted.
meropenem–vaborbactam as salvage therapy for CAZ–AVI-
resistant Klebsiella pneumonia infection, which indicated The clinical data in MDR‑PA infections
that meropenem–vaborbactam may be an alternative choice.
Currently, there is no best choice for treating CAZ–AVI- Besides CRE, CAZ–AVI was also active to MDR-PA. Cur-
resistant CRE infections. Therefore, increasing hospital sur- rently, some published studies from different countries have
veillance and preventing the transmission of resistance are illustrated the therapeutic effect in CRPA or MDR-PA infec-
essential. Moreover, selecting factors associated with resist- tions. Corbella et al. [101] conducted a retrospective cohort
ance and optimizing antibiotics to decrease the emergency on patients with MDR or extremely resistant (XDR) PA
of resistance are also beneficial. infections treated with CAZ–AVI in Spain. The source of
Many laboratories usually detect CRE by susceptibility infection included pneumonia (21/61, 34.4%), UTI (16/61,
tests and phenotypic carbapenemase tests such as mCIM 26.2%), skin and soft-tissue infection (14/61, 23.3%), IAI
and β-CarbaTM  test or  immunochromatography  assays. (7/61, 11.5%), and others. Half of them had sepsis at presen-
Studies have found that some of the strains (such as those tation. CAZ–AVI was initiated at a median of 7.0 days from
with KPC-31, KPC-94, and KPC-95) cannot be detected by symptom onset. The 14-day clinical cure rate and 30-day
the traditional phenotypic carbapenemase tests [75, 81, 97]. all-cause mortality rate were 54.1% and 13.1%, respectively.
Accurate molecular approach—PCR—should be imple- Chen et al. [102] enrolled 136 patients with CRPA infec-
mented together with traditional methods to prevent CRE tions in a single center in China to provide comparison of
from escaping regular surveillance. CAZ–AVI and polymyxin. Almost all patients had pneu-
In terms of the risk factors of CAZ–AVI resistance, monia infections (n = 135, 99.3%), while 49 (36.0%) had
some authors proposed that suboptimal dose exposure to BSI. The 14-day mortality rate (5.9% vs 27.1%, P = 0.002),
CAZ–AVI is one of the predictors [98]. Shields et al. [39] 30-day mortality rate (13.7% vs 47.1%, P < 0.001), and in-
found that renal replacement therapy was independently hospital rate (29.4% vs 60.0%, P = 0.001) were lower in
associated with emergence of resistance to CAZ–AVI the CAZ–AVI group than in the polymyxin B group. Fur-
(P = 0.009) in a retrospective study including 77 patients thermore, after adjusting for confounders, the difference in
with CRE infections, suggesting that renal replacement 30-day mortality between the two groups was still signifi-
therapy may lead to suboptimal dose. Fontana et al. [97] cant (14.3% vs 42.9%, P = 0.018). Regarding to the bacte-
reported that resistance to CAZ–AVI tended to occur among rial eradication, the frequency was significantly higher in
those with a long previous exposure time to this drug. How- those with CAZ–AVI than polymyxin B (45.1% vs 12.9%,
ever, Shen et al. [83] reported a strain developed resistance P < 0.001). Multivariate Cox analysis identified CAZ–AVI
only after 3 days exposure to CAZ–AVI in one case. The therapy and central venous catheterization as independ-
exposure duration associated with resistance is difficult to ent predictors of 30-day survival, and age as the predic-
define. Moreover, Ackley et al. [36] reported higher resist- tor of 30-day mortality. A multicenter retrospective study
ance rate in those treated with monotherapy than combina- conducted in 6 medical centers in the USA enrolled 203
tion therapy numerically (3/41, 7.3% vs 0/64, 0%), but this patients [64]. CRE and CRPA were isolated from 117 and
was limited by its small sample and confounding factors 63 culture specimens, respectively. For CRPA infections, the
between the two groups. most frequent infection source was respiratory tract (60.3%),
Notably, CAZ–AVI resistance in KPC-KP producers followed by urinary tract (11.1%) and so on. The 30-day
developed independently from previous exposure. Hum- mortality and 30-day recurrence rates were 17.5% (11/63)
phries et al. [99] and Gaibani et al. [100] reported 1 and 3 and 6.3% (4/63). Another multicenter retrospective study
cases infected with KPC-KP isolates resistant to CAZ–AVI involving 41 MDR-GNB (other than CRE) patients treated
without prior treatment of CAZ–AVI, respectively. with CAZ–AVI monotherapy (8/41, 19.5%) or combination
The emergence of CAZ–AVI resistance among CRE is therapy (33/41, 80.5%) from 13 hospitals in Italy [103].
becoming a concerning problem, decreasing the efficacy of The most common causative episodes were Pseudomonas

13

1418 S. Zhen et al.

aeruginosa (33/41, 80.5%) and ESBL-producing Enterobac- Author contributions  SZ and HW drafted the manuscript, and Sizhou
terales (4/41, 9.8%), whereas the remaining four patients had Feng critically revised and approved the final version to be published.
polymicrobial infections. The clinical cure rate was 87.8%
Funding  This work received grant from funding CAMS Innovation
(29/33) in patients infected with Pseudomonas aeruginosa, Fund for Medical Sciences (CIFMS) 2021-I2M-C&T-B-080 and
and among 4 patients experienced clinical failure, 3 patients 2021-I2M-017.
presented with sepsis or septic shock. Several case series and
case reports also highlighted the efficacy of CAZ–AVI for Declarations 
CRPA infections, even in central nervous system infection
had been described [104–108]. Conflict of interest  There are no conflicts of interest to report.
With the increasing use of this drug, the emergence of
Ethical approval  Not required.
resistance to CAZ–AVI in vivo or in vitro studies is concern-
ing. Ruedas-López et al. [109] and Fraile-Ribot et al. [110]
reported the resistance after CAZ–AVI treatment of Pseu-
domonas aeruginosa infections. Moreover, some studies also References
reported that Pseudomonas aeruginosa developed cross-
resistance to ceftazidime, CAZ–AVI and ceftolozane/tazobac- 1. Doi Y, Iovleva A, Bonomo R. The ecology of extended-spectrum
tam during treatment of cephalosporins or other antibiotics β-lactamases (ESBLs) in the developed world. J Travel Med.
[111–113]. Generally, like CRE, the most common resistance 2017;24:S44-S51. https://​doi.​org/​10.​1093/​jtm/​taw102.
2. Grundmann H, Glasner C, Albiger B, Aanensen D, Tomlinson
mechanism to CAZ–AVI in Pseudomonas aeruginosa is enzy- C, Andrasević A, et al. Occurrence of carbapenemase-produc-
matic inactivation. The hyperproduction or structural modifi- ing Klebsiella pneumoniae and Escherichia coli in the Euro-
cation of AmpC, and the variants of OXA-2 or OXA-10 are pean survey of carbapenemase-producing Enterobacteriaceae
common causes [111, 113, 114]. Moreover, one recent study (EuSCAPE): a prospective, multinational study. Lancet Infect
Dis. 2017;17:153–63. https://​doi.​org/​10.​1016/​s1473-​3099(16)​
reported that KPC-90 variants confer resistance to CAZ–AVI 30257-2.
in CRPA [115]. Another study in China found that CAZ–AVI 3. Shortridge D, Gales A, Streit J, Huband M, Tsakris A, Jones
resistance was associated with ­blaKPC-2 copy-number variation R. Pseudomonas aeruginosageographic and temporal patterns
[116]. In addition to β-lactamase, the membrane impermeabil- of antimicrobial resistance in over 20 years from the SENTRY
antimicrobial surveillance program, 1997–2016. Open Forum
ity due to porin mutations and high rates of OprD loss play a Infect Dis. 2019;6:S63–8. https://​doi.​org/​10.​1093/​ofid/​ofy343.
role in CAZ–AVI resistance [87]. Efflux pumps do not appear 4. Eichenberger E, Thaden J. Epidemiology and Mechanisms of
to play a major role in CAZ–AVI resistance. Resistance of Extensively Drug Resistant Gram-Negative Bac-
CAZ–AVI appears to be a valuable therapeutic option for teria. Antibiotics (Basel, Switzerland). 2019;8(2). https://d​ oi.o​ rg/​
10.​3390/​antib​iotic​s8020​037.
infections due to MDR-PA, but the emergence of resistance 5. Weiner L, Webb A, Limbago B, Dudeck M, Patel J, Kallen
is alarming. A, et  al. Antimicrobial-resistant pathogens associated with
healthcare-associated infections: summary of data reported to
the national healthcare safety network at the centers for disease
control and prevention, 2011–2014. Infect Control Hosp Epide-
Summary miol. 2016;37:1288–301. https://​doi.​org/​10.​1017/​ice.​2016.​174.
6. https://w ​ ww.​ecdc.​europa.​eu/s​ ites/​portal/​files/​docum​ents/​EARS-​
Net-​report-​2017-​update-​jan-​2019.​pdf.
The increasing prevalence of CRE and MDR-PA is a major 7. Centers for Disease Control and Prevention (CDC). Antimicro-
threat to human health globally. Fortunately, CAZ–AVI shows bial Resistance. Atlanta: CDC; 2015. http://​www.​cdc.​gov/​drugr​
significant benefits for infections due to CRE and MDR-PA, esist​ance/​index.​html. https://​doi.​org/​10.​3389/​fmed.​2019.​00074.
even in those with immunocompromised status, supporting a 8. Vardakas K, Matthaiou D, Falagas M, Antypa E, Koteli A, Anto-
niadou E. Characteristics, risk factors and outcomes of carbape-
new therapeutic option. The efficacy of CAZ–AVI varies in nem-resistant Klebsiella pneumoniae infections in the intensive
different infection sites due to CRE. Early use is associated care unit. J Infect. 2015;70:592–9. https://​doi.​org/​10.​1016/j.​jinf.​
with improved clinical outcomes. Noteworthy, when adopted 2014.​11.​003.
as salvage therapy, CAZ–AVI is still superior to other GNB 9. Villegas M, Pallares C, Escandón-Vargas K, Hernández-Gómez
C, Correa A, Álvarez C, et al. Characterization and clinical
active antibiotics. CAZ–AVI plus aztreonam is the first line impact of bloodstream infection caused by carbapenemase-pro-
of MBL-CRE infections. However, for infections caused by ducing enterobacteriaceae in seven Latin American Countries.
KPC- and OXA-48-producing isolates, further investigations PLoS ONE. 2016;11: e0154092. https://d​ oi.o​ rg/1​ 0.1​ 371/j​ ourna​ l.​
are needed to demonstrate the benefit of combination therapy. pone.​01540​92.
10. Veeraraghavan B, Shankar C, Karunasree S, Kumari S, Ravi
The development of resistance in CRE and MDR-PA against R, Ralph R. Carbapenem resistant Klebsiella pneumoniae iso-
CAZ–AVI is alarming, and more investigations and studies lated from bloodstream infection: Indian experience. Pathogens
are needed to prevent, diagnose, and treat infections due to Global Health. 2017;111:240–6. https://​doi.​org/​10.​1080/​20477​
CAZ–AVI-resistant pathogens. 724.​2017.​13401​28.

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1419

11. Paczosa M, Mecsas J. Klebsiella pneumoniae: going on infections due to carbapenem-resistant enterobacteriaceae. Clin
the offense with a strong defense. Microbiol Mol Biol Rev. Infect Dis. 2018;66:163–71. https://​doi.​org/​10.​1093/​cid/​cix783.
2016;80:629–61. https://​doi.​org/​10.​1128/​mmbr.​00078-​15. 25. Hakeam H, Alsahli H, Albabtain L, Alassaf S, Al Duhailib Z,
12. Iredell J, Brown J, Tagg K. Antibiotic resistance in Enterobac- Althawadi S. Effectiveness of ceftazidime–avibactam versus
teriaceae: mechanisms and clinical implications. BMJ (Clin Res colistin in treating carbapenem-resistant Enterobacteriaceae
ed). 2016;352: h6420. https://​doi.​org/​10.​1136/​bmj.​h6420. bacteremia. Int J Infect Dis. 2021;109:1–7. https://​doi.​org/​10.​
13. Maraolo A, Corcione S, Grossi A, Signori A, Alicino C, Hus- 1016/j.​ijid.​2021.​05.​079.
sein K, et al. The impact of carbapenem resistance on mortal- 26. Almangour T, Ghonem L, Aljabri A, Alruwaili A, Al Musawa
ity in patients with klebsiella pneumoniae bloodstream infec- M, Damfu N, et al. Ceftazidime–avibactam versus colistin for
tion: an individual patient data meta-analysis of 1952 patients. the treatment of infections due to carbapenem-resistant entero-
Infect Dis Therapy. 2021;10(1):541–58. https://​doi.​org/​10.​1007/​ bacterales: a multicenter cohort study. Infect Drug Resistance.
s40121-​021-​00408-8. 2022;15:211–21. https://​doi.​org/​10.​2147/​idr.​S3490​04.
14. Garcia-Vidal C, Cardozo-Espinola C, Puerta-Alcalde P, Marco F, 27. Fang J, Li H, Zhang M, Shi G, Liu M, Wang Y, et al. Klebsiella
Tellez A, Agüero D, et al. Risk factors for mortality in patients pneumonia efficacy of ceftazidime–avibactam versus polymyxin
with acute leukemia and bloodstream infections in the era of b and risk factors affecting clinical outcomes in patients with
multiresistance. PLoS ONE. 2018;13: e0199531. https://​doi.​org/​ carbapenem-resistant infections a retrospective study. Front
10.​1371/​journ​al.​pone.​01995​31. Pharmacol. 2021;12: 780940. https://d​ oi.o​ rg/1​ 0.3​ 389/f​ phar.2​ 021.​
15. Micozzi A, Gentile G, Minotti C, Cartoni C, Capria S, Ballarò D, 780940.
et al. Carbapenem-resistant Klebsiella pneumoniae in high-risk 28. Castón J, Cano A, Pérez-Camacho I, Aguado J, Carratalá J,
haematological patients: factors favouring spread, risk factors Ramasco F, et al. Impact of ceftazidime/avibactam versus best
and outcome of carbapenem-resistant Klebsiella pneumoniae available therapy on mortality from infections caused by car-
bacteremias. BMC Infect Dis. 2017;17:203. https://​doi.​org/​10.​ bapenemase-producing Enterobacterales (CAVICOR study). J
1186/​s12879-​017-​2297-9. Antimicrob Chemother. 2022. https://​doi.​org/​10.​1093/​jac/​dkac0​
16. Trecarichi EM, Pagano L, Martino B, Candoni A, Di Blasi R, 49.
Nadali G, et al. Bloodstream infections caused by Klebsiella 29. Shields R, Nguyen M, Chen L, Press E, Potoski B, Marini R,
pneumoniae in onco-hematological patients: clinical impact et al. Ceftazidime–avibactam Is Superior to Other Treatment
of carbapenem resistance in a multicentre prospective survey. Regimens against Carbapenem-Resistant Klebsiella pneumoniae
Am J Hematol. 2016;91:1076–81. https://​doi.​org/​10.​1002/​ajh.​ Bacteremia. Antimicrob Agents Chemother. 2017;61. https://d​ oi.​
24489. org/​10.​1128/​aac.​00883-​17.
17. Wei J, Zhu Q, Sun Z, Wang C. Pseudomonas aeruginosa[The 30. Gu J, Xu J, Zuo T, Chen Y. Ceftazidime–avibactam in the treat-
impact of carbapenem-resistance infections on mortality of ment of infections from carbapenem-resistant Klebsiella pneu-
patients with hematological disorders]. Zhonghua Nei Ke Za Zhi. moniae: Ceftazidime–avibactam against CR-KP infections. J
2020;59:353–9. https://​doi.​org/​10.​3760/​cma.j.​cn112​138-​20191​ Global Antimicrobial Resistance. 2021;26:20–5. https://​doi.​org/​
104-​00728. 10.​1016/j.​jgar.​2021.​04.​022.
18. de Jonge B, Karlowsky J, Kazmierczak K, Biedenbach D, Sahm 31. Alraddadi B, Saeedi M, Qutub M, Alshukairi A, Hassanien
D, Nichols W. In vitro susceptibility to ceftazidime–avibactam A, Wali G. Efficacy of ceftazidime–avibactam in the treat-
of carbapenem-non-susceptible enterobacteriaceae isolates ment of infections due to Carbapenem-resistant Enterobacte-
collected during the inform global surveillance study (2012 to riaceae. BMC Infect Dis. 2019;19:772. https://​doi.​org/​10.​1186/​
2014). Antimicrob Agents Chemother. 2016;60:3163–9. https://​ s12879-​019-​4409-1.
doi.​org/​10.​1128/​aac.​03042-​15. 32. Chen L, Han X, Li Y, Li M. Enterobacterales assessment of mor-
19. Zhanel G, Lawson C, Adam H, Schweizer F, Zelenitsky tality-related risk factors and effective antimicrobial regimens
S, Lagacé-Wiens P, et  al. Ceftazidime–avibactam: a novel for treatment of bloodstream infections caused by carbapenem-
cephalosporin/β-lactamase inhibitor combination. Drugs. resistant. Antimicrob Agents Chemother. 2021;65: e0069821.
2013;73:159–77. https://​doi.​org/​10.​1007/​s40265-​013-​0013-7. https://​doi.​org/​10.​1128/​aac.​00698-​21.
20. Turton J, Woodford N, Glover J, Yarde S, Kaufmann M, Pitt T. 33. Tsolaki V, Mantzarlis K, Mpakalis A, Malli E, Tsimpoukas
Identification of Acinetobacter baumannii by detection of the F, Tsirogianni A, et al. Ceftazidime–avibactam To Treat Life-
blaOXA-51-like carbapenemase gene intrinsic to this species. Threatening Infections by Carbapenem-Resistant Pathogens
J Clin Microbiol. 2006;44:2974–6. https://​doi.​org/​10.​1128/​jcm.​ in Critically Ill Mechanically Ventilated Patients. Antimicrob
01021-​06. Agents Chemother. 2020;64. https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 128/ ​ a ac.​
21. Savov E, Trifonova A, Kovachka K, Kjosseva E, Strateva T. 02320-​19.
in vitroAntimicrobial activities of ceftazidime–avibactam, mero- 34. Shen L, Lian C, Zhu B, Yao Y, Yang Q, Zhou J, et al. Kleb-
penem–vaborbactam and plazomicin against multidrug-resistant siella pneumonia bloodstream infections due to carbapenem-
and—a pilot Bulgarian study. Infect Dis (London, England). resistant : a single-center retrospective study on risk factors and
2019;51:870–3. https://​doi.​org/​10.​1080/​23744​235.​2019.​16534​ therapy options. Microbial drug resistance (Larchmont, NY).
91. 2021;27:227–33. https://​doi.​org/​10.​1089/​mdr.​2019.​0455.
22. Merdjan H, Rangaraju M, Tarral A. Safety and pharmacokinet- 35. Zhong H, Zhao X, Zhang Z, Gu Z, Zhang C, Gao Y, et  al.
ics of single and multiple ascending doses of avibactam alone Evaluation of the efficacy and safety of ceftazidime/avibac-
and in combination with ceftazidime in healthy male volun- tam in the treatment of Gram-negative bacterial infections: a
teers: results of two randomized, placebo-controlled studies. systematic review and meta-analysis. Int J Antimicrob Agents.
Clin Drug Investig. 2015;35:307–17. https://​doi.​org/​10.​1007/​ 2018;52:443–50. https://​doi.​org/​10.​1016/j.​ijant​imicag.​2018.​07.​
s40261-​015-​0283-9. 004.
23. https://​www.​idsoc​iety.​org/​pract​ice-​guide​line/​amr-​guida​nce/. 36. Ackley R, Roshdy D, Meredith J, Minor S, Anderson W, Capraro
Accessed 22 March 2022. G, et al. Enterobacteriaceae meropenem–vaborbactam versus
24. van Duin D, Lok J, Earley M, Cober E, Richter S, Perez F, ceftazidime–avibactam for treatment of carbapenem-resistant
et al. Colistin versus ceftazidime–avibactam in the treatment of

13

1420 S. Zhen et al.

infections. antimicrob agents chemother. 2020;64. https://​doi.​ 49. King M, Heil E, Kuriakose S, Bias T, Huang V, El-Beyrouty C,
org/​10.​1128/​aac.​02313-​19. et al. Outcomes With Ceftazidime/Avibactam in Patients With
37. Castón J, Gallo M, García M, Cano A, Escribano A, Machuca Carbapenem-Resistant Enterobacteriaceae (CRE) Infections: A
I, et al. Ceftazidime–avibactam in the treatment of infections Multicenter Study. Open Forum Infectious Diseases. 2016;3.
caused by KPC-producing Klebsiella pneumoniae: factors asso- https://​doi.​org/​10.​1093/​ofid/​ofw172.​1595.
ciated with clinical efficacy in a single-center cohort. Int J Anti- 50. De la Calle C, Rodríguez O, Morata L, Marco F, Cardozo C,
microb Agents. 2020;56: 106075. https://​doi.​org/​10.​1016/j.​ijant​ García-Vidal C, et al. Clinical characteristics and prognosis of
imicag.​2020.​106075. infections caused by OXA-48 carbapenemase-producing Entero-
38. Tumbarello M, Trecarichi E, Corona A, De Rosa F, Bassetti M, bacteriaceae in patients treated with ceftazidime–avibactam. Int
Mussini C, et al. Efficacy of Ceftazidime–avibactam Salvage J Antimicrob Agents. 2019;53:520–4. https://​doi.​org/​10.​1016/j.​
Therapy in Patients With Infections Caused by Klebsiella pneu- ijant​imicag.​2018.​11.​015.
moniae Carbapenemase-producing K. pneumoniae. Clin Infect 51. Fiore M, Alfieri A, Di Franco S, Pace M, Simeon V, Ingoglia G,
Dis. 2019;68:355–64. https://​doi.​org/​10.​1093/​cid/​ciy492. et al. Ceftazidime–avibactam Combination Therapy Compared
39. Shields R, Nguyen M, Chen L, Press E, Kreiswirth B, Clancy C. to Ceftazidime–avibactam Monotherapy for the Treatment of
Pneumonia and Renal Replacement Therapy Are Risk Factors Severe Infections Due to Carbapenem-Resistant Pathogens: A
for Ceftazidime–avibactam Treatment Failures and Resistance Systematic Review and Network Meta-Analysis. Antibiotics
among Patients with Carbapenem-Resistant Enterobacteriaceae (Basel, Switzerland). 2020;9. https://​doi.​org/​10.​3390/​antib​iotic​
Infections. Antimicrob Agents Chemother. 2018;62. https://​doi.​ s9070​388.
org/​10.​1128/​aac.​02497-​17. 52. Tumbarello M, Raffaelli F, Giannella M, Mantengoli E, Mularoni
40. Shi Y, Hu J, Liu P, Wang T, Wang H, Liu Y, et al. Ceftazi- A, Venditti M, et al. Ceftazidime–avibactam use for KPC-Kp
dime–avibactam-based versus tigecycline-based regimen for infections: a retrospective observational multicenter study. Clin
the treatment of carbapenem-resistant klebsiella pneumoniae- Infect Dis. 2021. https://​doi.​org/​10.​1093/​cid/​ciab1​76.
induced pneumonia in critically Ill patients. Infect Dis Therapy. 53. Karaiskos I, Daikos G, Gkoufa A, Adamis G, Stefos A, Symbardi
2021;10:2721–34. https://d​ oi.​org/1​ 0.1​ 007/​s40121-​021-0​ 0542-3. S, et al. Ceftazidime/avibactam in the era of carbapenemase-
41. Castón J, Lacort-Peralta I, Martín-Dávila P, Loeches B, Tabares producing Klebsiella pneumoniae: experience from a national
S, Temkin L, et al. Clinical efficacy of ceftazidime/avibactam registry study. J Antimicrob Chemother. 2021;76:775–83. https://​
versus other active agents for the treatment of bacteremia due doi.​org/​10.​1093/​jac/​dkaa5​03.
to carbapenemase-producing Enterobacteriaceae in hematologic 54. Zheng G, Zhang J, Wang B, Cai J, Wang L, Hou K, et  al.
patients. Int J Infect Dis. 2017;59:118–23. https://​doi.​org/​10.​ Ceftazidime–avibactam in combination with in  vitro non-
1016/j.​ijid.​2017.​03.​021. susceptible antimicrobials versus ceftazidime–avibactam in
42. Clerici D, Oltolini C, Greco R, Ripa M, Giglio F, Mastaglio monotherapy in critically Ill patients with carbapenem-resistant
S, et al. The place of ceftazidime/avibactam and ceftolozane/ klebsiella pneumoniae infection: a retrospective cohort study.
tazobactam for therapy of haematological patients with febrile Infect Dis Therapy. 2021;10:1699–713. https://​doi.​org/​10.​1007/​
neutropenia. Int J Antimicrob Agents. 2021;57: 106335. https://​ s40121-​021-​00479-7.
doi.​org/​10.​1016/j.​ijant​imicag.​2021.​106335. 55. Shaw E, Rombauts A, Tubau F, Padullés A, Càmara J, Lozano
43. Chen F, Zhong H, Yang T, Shen C, Deng Y, Han L, et al. Kleb- T, et al. Clinical outcomes after combination treatment with cef-
siella pneumoniaeCeftazidime–avibactam as Salvage Treatment tazidime/avibactam and aztreonam for NDM-1/OXA-48/CTX-
for Infections Due to Carbapenem-Resistant in Liver Transplan- M-15-producing Klebsiella pneumoniae infection. J Antimicrob
tation Recipients. Infect Drug Resistance. 2021;14:5603–12. Chemother. 2018;73:1104–6. https://d​ oi.o​ rg/1​ 0.1​ 093/j​ ac/d​ kx496.
https://​doi.​org/​10.​2147/​idr.​S3421​63. 56. Davido B, Fellous L, Lawrence C, Maxime V, Rottman M, Dinh
44. Zhang F, Zhong J, Ding H, Liao G. Klebsiella pneumonia effi- A. Ceftazidime–avibactam and Aztreonam, an Interesting Strat-
cacy of ceftazidime–avibactam in the treatment of carbapenem- egy To Overcome β-Lactam Resistance Conferred by Metallo-β-
resistant infection after kidney transplantation. Infect Drug Lactamases in Enterobacteriaceae and Pseudomonas aeruginosa.
Resistance. 2021;14:5165–74. https://​doi.​org/​10.​2147/​idr.​S3435​ Antimicrob Agents Chemother. 2017;61. https://d​ oi.o​ rg/1​ 0.1​ 128/​
05. aac.​01008-​17.
45. Chen W, Sun L, Guo L, Cao B, Liu Y, Zhao L, et al. Clinical 57. Bocanegra-Ibarias P, Camacho-Ortiz A, Garza-González E,
outcomes of ceftazidime–avibactam in lung transplant recipi- Flores-Treviño S, Kim H, Perez-Alba E. Aztreonam plus ceftazi-
ents with infections caused by extensively drug-resistant gram- dime–avibactam as treatment of NDM-1-producing Klebsiella
negative bacilli. Ann Trans Med. 2020;8:39. https://​doi.​org/​10.​ pneumoniae bacteraemia in a neutropenic patient: Last resort
21037/​atm.​2019.​10.​40. therapy? J Global Antimicrobial Resistance. 2020;23:417–9.
46. Kuang H, Zhong C, Wang Y, Ye H, Ao K, Zong Z, et al. Clinical https://​doi.​org/​10.​1016/j.​jgar.​2020.​10.​019.
characteristics and outcomes of patients with multidrug-resist- 58. Benchetrit L, Mathy V, Armand-Lefevre L, Bouadma L, Timsit
ant Gram-negative bacterial infections treated with ceftazidime/ J. Successful treatment of septic shock due to NDM-1-producing
avibactam. J Global Antimicrobial Resistance. 2020;23:404–7. Klebsiella pneumoniae using ceftazidime/avibactam combined
https://​doi.​org/​10.​1016/j.​jgar.​2020.​10.​023. with aztreonam in solid organ transplant recipients: report of two
47. King M, Heil E, Kuriakose S, Bias T, Huang V, El-Beyrouty C, cases. Int J Antimicrob Agents. 2020;55: 105842. https://d​ oi.o​ rg/​
et al. Multicenter Study of Outcomes with Ceftazidime–avibac- 10.​1016/j.​ijant​imicag.​2019.​10.​023.
tam in Patients with Carbapenem-Resistant Enterobacteriaceae 59. Yasmin M, Fouts D, Jacobs M, Haydar H, Marshall S, White R,
Infections. Antimicrob Agents Chemother. 2017;61. https://​doi.​ et al. Monitoring Ceftazidime–avibactam and Aztreonam Con-
org/​10.​1128/​aac.​00449-​17. centrations in the Treatment of a Bloodstream Infection Caused
48. Shields R, Potoski B, Haidar G, Hao B, Doi Y, Chen L, et al. by a Multidrug-Resistant Enterobacter sp. Carrying Both Kleb-
Clinical outcomes, drug toxicity, and emergence of ceftazi- siella pneumoniae Carbapenemase-4 and New Delhi Metallo-β-
dime–avibactam resistance among patients treated for carbap- Lactamase-1. Clinical infectious diseases : an official publication
enem-resistant enterobacteriaceae infections. Clin Infect Dis. of the Infectious Diseases Society of America. 2020;711095–8.
2016;63:1615–8. https://​doi.​org/​10.​1093/​cid/​ciw636. https://​doi.​org/​10.​1093/​cid/​ciz11​55.

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1421

60. Sieswerda E, van den Brand M, van den Berg R, Sträter J, Sch- Bacteremia and Abscess in a Liver Transplant Recipient. Anti-
ouls L, van Dijk K, et al. Successful rescue treatment of sepsis microb Agents Chemother. 2019;63. https://d​ oi.o​ rg/1​ 0.1​ 128/a​ ac.​
due to a pandrug-resistant, NDM-producing Klebsiella pneu- 01551-​18.
moniae using aztreonam powder for nebulizer solution as intra- 73. Hemarajata P, Humphries R. Ceftazidime/avibactam resistance
venous therapy in combination with ceftazidime/avibactam. J associated with L169P mutation in the omega loop of KPC-2. J
Antimicrob Chemother. 2020;75:773–5. https://d​ oi.o​ rg/1​ 0.1​ 093/​ Antimicrob Chemother. 2019;74(5):1241–3. https://​doi.​org/​10.​
jac/​dkz495. 1093/​jac/​dkz026.
61. Falcone M, Daikos G, Tiseo G, Bassoulis D, Giordano C, Galfo 74. Gaibani P, Campoli C, Lewis R, Volpe S, Scaltriti E, Giannella
V, et al. Efficacy of ceftazidime–avibactam plus aztreonam in M, et al. In vivo evolution of resistant subpopulations of KPC-
patients with bloodstream infections caused by MBL- producing producing Klebsiella pneumoniae during ceftazidime/avibactam
Enterobacterales. Clin Infect Dis. 2020. https://​doi.​org/​10.​1093/​ treatment. J Antimicrob Chemother. 2018;73:1525–9. https://d​ oi.​
cid/​ciaa5​86. org/​10.​1093/​jac/​dky082.
62. Bernhard M, Lichtenstern C, Eckmann C, Weigand M. The 75. Antonelli A, Giani T, Di Pilato V, Riccobono E, Perriello G,
early antibiotic therapy in septic patients–milestone or stick- Mencacci A, et al. KPC-31 expressed in a ceftazidime/avibac-
ing point? Crit Care. 2014;18:671. https://​doi.​org/​10.​1186/​ tam-resistant Klebsiella pneumoniae is associated with relevant
s13054-​014-​0671-1. detection issues. J Antimicrob Chemother. 2019;74:2464–6.
63. Falcone M, Bassetti M, Tiseo G, Giordano C, Nencini E, Russo https://​doi.​org/​10.​1093/​jac/​dkz156.
A, et al. Time to appropriate antibiotic therapy is a predictor of 76. Venditti C, Nisii C, D’Arezzo S, Vulcano A, Capone A, Antonini
outcome in patients with bloodstream infection caused by KPC- M, et al. blaMolecular and phenotypical characterization of two
producing Klebsiella pneumoniae. Crit Care. 2020;24:29. https://​ cases of antibiotic-driven ceftazidime–avibactam resistance in
doi.​org/​10.​1186/​s13054-​020-​2742-9. -harboring. Infect Drug Resistance. 2019;12:1935–40. https://​
64. Jorgensen S, Trinh T, Zasowski E, Lagnf A, Bhatia S, Melvin S, doi.​org/​10.​2147/​idr.​S2079​93.
et al. Real-World Experience With Ceftazidime–avibactam for 77. Gaibani P, Ambretti S, Campoli C, Viale P, Re M. Genomic
Multidrug-Resistant Gram-Negative Bacterial Infections. Open characterization of a Klebsiella pneumoniae ST1519 resistant to
forum infectious diseases. 2019;6:ofz522. https://​doi.​org/​10.​ ceftazidime/avibactam carrying a novel KPC variant (KPC-36).
1093/​ofid/​ofz522. Int J Antimicrob Agents. 2020;55: 105816. https://​doi.​org/​10.​
65. Guimarães T, Nouér S, Martins R, Perdigão Neto L, Martins W, 1016/j.​ijant​imicag.​2019.​09.​020.
Narciso Barbosa A, et al. EnterobacterialesCeftazidime–avibac- 78. Galani I, Karaiskos I, Souli M, Papoutsaki V, Galani L, Gkoufa
tam as Salvage Therapy for Infections Caused by Coresistant to A, et al. Klebsiella pneumoniaeOutbreak of KPC-2-producing
Carbapenems and Polymyxins. Antimicrob Agents Chemother. endowed with ceftazidime–avibactam resistance mediated
2019;63. https://​doi.​org/​10.​1128/​aac.​00528-​19. through a VEB-1-mutant (VEB-25), Greece, September to
66. Onorato L, Di Caprio G, Signoriello S, Coppola N. Efficacy of October 2019. Euro surveillance : bulletin Europeen sur les
ceftazidime/avibactam in monotherapy or combination therapy maladies transmissibles = European communicable disease
against carbapenem-resistant Gram-negative bacteria: a meta- bulletin. 2020;25. https://​doi.​org/​10.​2807/​1560-​7917.​Es.​2020.​
analysis. Int J Antimicrob Agents. 2019;54:735–40. https://​doi.​ 25.3.​20000​28.
org/​10.​1016/j.​ijant​imicag.​2019.​08.​025. 79. Galani I, Karaiskos I, Angelidis E, Papoutsaki V, Galani L,
67. Iannaccone M, Boattini M, Bianco G, Corcione S, Cavallo R, Souli M, et al. Emergence of ceftazidime–avibactam resist-
Costa C. EnterobacteralesCeftazidime–avibactam susceptible ance through distinct genomic adaptations in KPC-2-producing
to resistant KPC-producing bloodstream infections: an observa- Klebsiella pneumoniae of sequence type 39 during treatment.
tional study. J Chemotherapy (Florence, Italy). 2020;32:160–2. Euro J Clin Microbiol Infect Dis. 2021;40(1):219–24. https://​
https://​doi.​org/​10.​1080/​11200​09x.​2019.​17093​63. doi.​org/​10.​1007/​s10096-​020-​04000-9.
68. Shields R, Chen L, Cheng S, Chavda K, Press E, Snyder A, 80. Cano Á, Guzmán-Puche J, García-Gutiérrez M, Castón J, Gra-
et al. blaEmergence of Ceftazidime–avibactam Resistance Due cia-Ahufinger I, Pérez-Nadales E, et al. Use of carbapenems in
to Plasmid-Borne Mutations during Treatment of Carbapenem- the combined treatment of emerging ceftazidime/avibactam-
Resistant Klebsiella pneumoniae Infections. Antimicrob Agents resistant and carbapenem-susceptible KPC-producing Kleb-
Chemother. 2017;61. https://​doi.​org/​10.​1128/​aac.​02097-​16. siella pneumoniae infections: Report of a case and review of
69. Shields R, Nguyen M, Press E, Chen L, Kreiswirth B, Clancy the literature. J Global Antimicrobial Resistance. 2020;22:9–
C. Klebsiella pneumonia emergence of ceftazidime–avibactam 12. https://​doi.​org/​10.​1016/j.​jgar.​2019.​11.​007.
resistance and restoration of carbapenem susceptibility in car- 81. Guzmán-Puche J, Pérez-Nadales E, Pérez-Vázquez M, Causse
bapenemase-producing: a case report and review of literature. M, Gracia-Ahufinger I, Mendez-Natera A, et al. In vivo selec-
Open forum infectious diseases. 2017;4:ofx101. https://​doi.​org/​ tion of KPC-94 and KPC-95 in Klebsiella pneumoniae isolates
10.​1093/​ofid/​ofx101. from patients treated with ceftazidime/avibactam. Int J Antimi-
70. Giddins M, Macesic N, Annavajhala M, Stump S, Khan S, crob Agents. 2022;59: 106524. https://​doi.​org/​10.​1016/j.​ijant​
McConville T, et al. blaSuccessive emergence of ceftazidime– imicag.​2022.​106524.
avibactam resistance through distinct genomic adaptations in 82. Li X, Quan J, Ke H, Wu W, Feng Y, Yu Y, et al. Klebsiella
-harboring klebsiella pneumoniae sequence type 307 isolates. pneumonia emergence of a KPC variant conferring resistance
Antimicrob Agents Chemother. 2018;62. https://d​ oi.o​ rg/1​ 0.1​ 128/​ to ceftazidime–avibactam in a widespread ST11 carbapenem-
aac.​02101-​17. resistant clone in China. Front Microbiol. 2021;12: 724272.
71. Castanheira M, Arends S, Davis A, Woosley L, Bhalodi A, https://​doi.​org/​10.​3389/​fmicb.​2021.​724272.
MacVane S. Citrobacter freundiiAnalyses of a Ceftazidime– 83. Shen S, Shi Q, Han R, Guo Y, Yang Y, Wu S, et al. blaIsola-
avibactam-Resistant Isolate Carrying Reveals a Heterogenous tion of a Ceftazidime–avibactam-Resistant -Positive Klebsiella
Population and Reversible Genotype. mSphere. 2018;3. https://​ pneumoniae Clinical Isolate. Microbiology spectrum. 2022;10:
doi.​org/​10.​1128/​mSphe​re.​00408-​18. e0184021. https://​doi.​org/​10.​1128/​spect​r um.​01840-​21.
72. Athans V, Neuner E, Hassouna H, Richter S, Keller G, Castan- 84. Räisänen K, Koivula I, Ilmavirta H, Puranen S, Kallonen T,
heira M, et al. Klebsiella pneumoniaeMeropenem–vaborbac- Lyytikäinen O, et  al. Klebsiella pneumoniaeEmergence of
tam as Salvage Therapy for Ceftazidime–avibactam-Resistant ceftazidime–avibactam-resistant during treatment, Finland,

13

1422 S. Zhen et al.

December 2018. Euro surveillance : bulletin Europeen sur les Avibactam-Resistant subsp. Isolates in a Tertiary Italian Hospi-
maladies transmissibles = European communicable disease tal: Identification of a New Mutation of the Carbapenemase Type
bulletin. 2019;24. https://​doi.​org/​10.​2807/​1560-​7917.​Es.​2019.​ 3 (KPC-3) Gene Conferring Ceftazidime/Avibactam Resistance.
24.​19.​19002​56. Microorganisms. 2021;9. https://​doi.​org/​10.​3390/​micro​organ​
85. Göttig S, Frank D, Mungo E, Nolte A, Hogardt M, Besier isms9​112356.
S, et al. Emergence of ceftazidime/avibactam resistance in 98. Sheu C, Chang Y, Lin S, Chen Y, Hsueh P. Enterobacteriaceae
KPC-3-producing Klebsiella pneumoniae in vivo. J Antimi- infections caused by carbapenem-resistant: an update on thera-
crob Chemother. 2019;74:3211–6. https://​doi.​org/​10.​1093/​jac/​ peutic options. Front Microbiol. 2019;10:80. https://​doi.​org/​10.​
dkz330. 3389/​fmicb.​2019.​00080.
86. Mueller L, Masseron A, Prod’Hom G, Galperine T, Greub G, 99. Humphries RM, Yang S, Hemarajata P, Ward KW, Hindler
Poirel L, et al. Phenotypic, biochemical and genetic analysis of JA, Miller SA, et  al. First report of ceftazidime–avibactam
KPC-41, a KPC-3 variant conferring resistance to ceftazidime– resistance in a KPC-3-expressing <span class=&quot;named-
avibactam and exhibiting reduced carbapenemase activity. Anti- content genus-species&quot; id=&quot;named-content-
microb Agents Chemother. 2019. https://​doi.​org/​10.​1128/​aac.​ 1&quot;>Klebsiella pneumoniae</span> Isolate. Antimicrob
01111-​19. Agents Chemother. 2015;59(10):6605. https://​doi.​org/​10.​1128/​
87. Wang Y, Wang J, Wang R, Cai Y. Resistance to ceftazidime– AAC.​01165-​15.
avibactam and underlying mechanisms. J Global Antimicrobial 100. Gaibani P, Re M, Campoli C, Viale P, Ambretti S. Bloodstream
Resistance. 2020;22:18–27. https://​doi.​org/​10.​1016/j.​jgar.​2019.​ infection caused by KPC-producing Klebsiella pneumoniae
12.​009. resistant to ceftazidime/avibactam: epidemiology and genomic
88. Galani I, Karaiskos I, Giamarellou H. Klebsiella pneumoniae- characterization. Clinical microbiology and infection : the official
Multidrug-resistant : mechanisms of resistance including updated publication of the European Society of Clinical Microbiology
data for novel β-lactam-β-lactamase inhibitor combinations. and Infectious Diseases. 2020;26:516.e1-.e4. https://​doi.​org/​10.​
Expert Rev Anti Infect Ther. 2021;19:1457–68. https://​doi.​org/​ 1016/j.​cmi.​2019.​11.​011.
10.​1080/​14787​210.​2021.​19246​74. 101. Corbella L, Boán J, San-Juan R, Fernández-Ruiz M, Carretero
89. Winkler M, Papp-Wallace K, Bonomo R. Activity of ceftazidime/ O, Lora D, et al. Effectiveness of ceftazidime–avibactam for the
avibactam against isogenic strains of Escherichia coli containing treatment of infections due to Pseudomonas aeruginosa. Int J
KPC and SHV β-lactamases with single amino acid substitu- Antimicrob Agents. 2022;59: 106517. https://​doi.​org/​10.​1016/j.​
tions in the Ω-loop. J Antimicrob Chemother. 2015;70:2279–86. ijant​imicag.​2021.​106517.
https://​doi.​org/​10.​1093/​jac/​dkv094. 102. Chen J, Liang Q, Chen X, Wu J, Wu Y, Teng G, et al. Pseu-
90. Both A, Büttner H, Huang J, Perbandt M, Belmar Campos C, domonas aeruginosa ceftazidime/avibactam versus polymyxin B
Christner M, et al. Emergence of ceftazidime/avibactam non- in the challenge of carbapenem-resistant infection. Infect Drug
susceptibility in an MDR Klebsiella pneumoniae isolate. J Anti- Resistance. 2022;15:655–67. https://​doi.​org/​10.​2147/​idr.​S3509​
microb Chemother. 2017;72:2483–8. https://d​ oi.o​ rg/1​ 0.1​ 093/j​ ac/​ 76.
dkx179. 103. Vena A, Giacobbe D, Castaldo N, Cattelan A, Mussini C, Luzzati
91. Livermore D, Mushtaq S, Doumith M, Jamrozy D, Nichols W, R, et al. EnterobacteralesClinical Experience with Ceftazidime–
Woodford N. Selection of mutants with resistance or dimin- avibactam for the Treatment of Infections due to Multidrug-
ished susceptibility to ceftazidime/avibactam from ESBL- and Resistant Gram-Negative Bacteria Other than Carbapenem-
AmpC-producing Enterobacteriaceae. J Antimicrob Chemother. Resistant. Antibiotics (Basel, Switzerland). 2020;9. https://​doi.​
2018;73:3336–45. https://​doi.​org/​10.​1093/​jac/​dky363. org/​10.​3390/​antib​iotic​s9020​071.
92. Porres-Osante N, Dupont H, Torres C, Ammenouche N, de 104. Rodríguez-Núñez O, Ripa M, Morata L, de la Calle C, Cardozo
Champs C, Mammeri H. Avibactam activity against extended- C, Fehér C, et al. Evaluation of ceftazidime/avibactam for serious
spectrum AmpC β-lactamases. J Antimicrob Chemother. infections due to multidrug-resistant and extensively drug-resist-
2014;69:1715–6. https://​doi.​org/​10.​1093/​jac/​dku002. ant Pseudomonas aeruginosa. J Global Antimicrobial Resistance.
93. Fröhlich C, Sørum V, Thomassen A, Johnsen P, Leiros H, Samu- 2018;15:136–9. https://​doi.​org/​10.​1016/j.​jgar.​2018.​07.​010.
elsen Ø. OXA-48-Mediated Ceftazidime–avibactam Resistance 105. Santevecchi B, Smith T, MacVane S. Clinical experience with
Is Associated with Evolutionary Trade-Offs. mSphere. 2019;4. ceftazidime/avibactam for treatment of antibiotic-resistant organ-
https://​doi.​org/​10.​1128/​mSphe​re.​00024-​19. isms other than Klebsiella pneumoniae. Int J Antimicrob Agents.
94. Nelson K, Hemarajata P, Sun D, Rubio-Aparicio D, Tsivkovski 2018;51:629–35. https://​doi.​org/​10.​1016/j.​ijant​imicag.​2018.​01.​
R, Yang S, et al. Resistance to Ceftazidime–avibactam Is Due to 016.
Transposition of KPC in a Porin-Deficient Strain of Klebsiella 106. Meschiari M, Franconi I, Bacca E, Bianco V, Orlando G, Cuomo
pneumoniae with Increased Efflux Activity. Antimicrob Agents G, et al. Ceftazidime/avibactam and ceftolozane/tazobactam for
Chemother. 2017;61. https://​doi.​org/​10.​1128/​aac.​00989-​17. the treatment of extensively drug-resistant Pseudomonas aerugi-
95. Shields R, Clancy C, Hao B, Chen L, Press E, Iovine N, et al. nosa post-neurosurgical infections: three cases and a review of
Effects of Klebsiella pneumoniae carbapenemase subtypes, the literature. Infection. 2021;49:549–53. https://d​ oi.o​ rg/1​ 0.1​ 007/​
extended-spectrum β-lactamases, and porin mutations on the s15010-​020-​01539-9.
in  vitro activity of ceftazidime–avibactam against carbape- 107. Almangour T, Alsubaie S, Ghonem L, Almohaini H, Mohammed
nem-resistant K. pneumoniae. Antimicrob Agents Chemother. Bakheet H, Altweijri I. Ceftazidime–avibactam for the treatment
2015;59:5793–7. https://​doi.​org/​10.​1128/​aac.​00548-​15. of multidrug-resistant Pseudomonas aeruginosa central nervous
96. Zhang Y, Kashikar A, Brown C, Denys G, Bush K. In VitroUnu- system infection in pediatric patient: a case report. Pediatr Infect
sual Escherichia coli PBP 3 Insertion Sequence Identified from a Dis J. 2021. https://​doi.​org/​10.​1097/​inf.​00000​00000​003439.
Collection of Carbapenem-Resistant Enterobacteriaceae Tested 108. Gatti M, Virgili G, Cojutti P, Gaibani P, Conti M, Sturiale C,
with a Combination of Ceftazidime-, Ceftaroline-, or Aztreonam- et  al. Real-Time Optimization of Pharmacodynamic Target
Avibactam. Antimicrob Agents Chemother. 2017;61. https://​doi.​ Attainment at Infection Site during Treatment of Post-Neurosur-
org/​10.​1128/​aac.​00389-​17. gical Ventriculitis Caused by Carbapenem-Resistant Gram Nega-
97. Fontana C, Favaro M, Campogiani L, Malagnino V, Minelli tives with Ceftazidime–avibactam-Based Regimens: A Report of
S, Bossa M, et  al. Klebsiella pneumoniaeCeftazidime/

13
Update of clinical application in ceftazidime–avibactam for multidrug‑resistant Gram‑negative… 1423

Two Cases. Microorganisms. 2022;10. https://​doi.​org/​10.​3390/​ ceftazidime–avibactam resistance upon treatment. Antimicrob
micro​organ​isms1​00101​54. Agents Chemother. 2019. https://​doi.​org/​10.​1128/​aac.​01637-​19.
109. Ruedas-López A, Alonso-García I, Lasarte-Monterrubio C, 113. Arca-Suárez J, Lasarte-Monterrubio C, Rodiño-Janeiro B, Cabot
Guijarro-Sánchez P, Gato E, Vázquez-Ucha J, et al. Selection of G, Vázquez-Ucha J, Rodríguez-Iglesias M, et  al. Molecular
AmpC β-Lactamase variants and metallo-β-lactamases leading mechanisms driving the in vivo development of OXA-10-me-
to ceftolozane/tazobactam and ceftazidime/avibactam resistance diated resistance to ceftolozane/tazobactam and ceftazidime/
during treatment of MDR/XDR pseudomonas aeruginosa infec- avibactam during treatment of XDR Pseudomonas aeruginosa
tions. Antimicrob Agents Chemother. 2022;66(2): e0206721. infections. J Antimicrob Chemother. 2021;76:91–100. https://​
https://​doi.​org/​10.​1128/​aac.​02067-​21. doi.​org/​10.​1093/​jac/​dkaa3​96.
110. Fraile-Ribot P, Fernández J, Gomis-Font M, Forcelledo L, 114. Horcajada J, Montero M, Oliver A, Sorlí L, Luque S, Gómez-
Mulet X, López-Causapé C, et al. In Vivo Evolution of GES Zorrilla S, et al. Epidemiology and Treatment of Multidrug-
β-lactamases driven by ceftazidime/avibactam treatment of pseu- Resistant and Extensively Drug-Resistant Pseudomonas aerugi-
domonas aeruginosa infections. Antimicrob Agents Chemother. nosa Infections. Clin Microbiol Rev. 2019;32. https://​doi.​org/​10.​
2021;65: e0098621. https://​doi.​org/​10.​1128/​aac.​00986-​21. 1128/​cmr.​00031-​19.
111. Fraile-Ribot P, Mulet X, Cabot G, Del Barrio-Tofiño E, Juan 115. Tu Y, Wang D, Zhu Y, Li J, Jiang Y, Wu W, et al. Emergence
C, Pérez J, et al. In Vivo Emergence of Resistance to Novel of a KPC-90 Variant that Confers Resistance to Ceftazidime–
Cephalosporin-β-Lactamase Inhibitor Combinations through avibactam in an ST463 Carbapenem-Resistant Pseudomonas
the Duplication of Amino Acid D149 from OXA-2 β-Lactamase aeruginosa Strain. Microbiology spectrum. 2022;10: e0186921.
(OXA-539) in Sequence Type 235 Pseudomonas aeruginosa. https://​doi.​org/​10.​1128/​spect​rum.​01869-​21.
Antimicrob Agents Chemother. 2017;61. https://d​ oi.o​ rg/1​ 0.1​ 128/​ 116. Zhu Y, Chen J, Shen H, Chen Z, Yang Q, Zhu J, et al. Emergence
aac.​01117-​17. of Ceftazidime- and Avibactam-Resistant Klebsiella pneumoniae
112. Boulant T, Jousset A, Bonnin R, Barrail-Tran A, Borgel A, Oue- Carbapenemase-Producing Pseudomonas aeruginosa in China.
slati S, et al. Pseudomonas aeruginosaA 2.5-years within-patient mSystems. 2021;6:e0078721. https://d​ oi.o​ rg/1​ 0.1​ 128/m
​ Syste​ ms.​
evolution of a with acquisition of ceftolozane-tazobactam and 00787-​21.

13

You might also like