Next Article in Journal
Chronic Pain and Emotional Stroop: A Systematic Review
Previous Article in Journal
The Impaired Elasticity of Large Arteries in Systemic Sclerosis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Durlobactam in the Treatment of Multidrug-Resistant Acinetobacter baumannii Infections: A Systematic Review

by
Guido Granata
1,
Fabrizio Taglietti
1,
Francesco Schiavone
2 and
Nicola Petrosillo
3,*
1
Clinical and Research Department for Infectious Diseases, National Institute for Infectious Diseases L. Spallanzani, IRCCS, 00149 Rome, Italy
2
Divers and Raiders Group Command “Teseo Tesei” COMSUBIN, Medical Service, Italian Navy, 19025 Portovenere, Italy
3
Head, Infection Prevention and Control–Infectious Disease Service, Foundation University Hospital Campus Bio-Medico, 00128 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(12), 3258; https://doi.org/10.3390/jcm11123258
Submission received: 3 May 2022 / Revised: 24 May 2022 / Accepted: 31 May 2022 / Published: 7 June 2022
(This article belongs to the Section Epidemiology & Public Health)

Abstract

:
A. baumannii is a frequent cause of difficult-to-treat healthcare-associated infections. The use of a novel beta-lactamase inhibitor, durlobactam, has been proposed against multidrug-resistant A. baumannii. A systematic review of studies assessing the efficacy and safety of durlobactam in the treatment of multidrug-resistant A. baumannii infections was carried out. The study protocol was pre-registered on PROSPERO (CRD42022311723). Published articles on durlobactam were identified through computerized literature searches with the search terms “durlobactam” and “ETX2514” using PubMed. PubMed was searched until 15 February 2022. Articles providing data on the main characteristics of durlobactam and on the efficacy and safety of durlobactam in the treatment of A. baumannii infections were included in this systematic review. Attempt was made to obtain information about unpublished studies. English language restriction was applied. The risk of bias in the included studies was not assessed. Both quantitative and qualitative information were summarized by means of textual descriptions. Thirty studies on durlobactam were identified, published from June 2017 to November 2020. Sixteen studies met the inclusion criteria. Durlobactam is effective against A. baumannii when used in combination with sulbactam. Future clinical trials are needed to confirm the possibility to treat infections caused by multidrug-resistant A. baumannii with this combination.

1. Introduction

Worldwide, the rise of multidrug-resistant (MDR) bacteria is an increasing threat to human health [1,2]. Among the most worrisome MDR bacteria, the World Health Organization (WHO) recently recognized Acinetobacter baumannii as a critical pathogen, frequently causing healthcare-associated infections (www.who.int accessed on 1 May 2022).
A. baumannii is a ubiquitous, non-fermenting, rod-shaped Gram-negative coccobacillus. It can be considered an opportunistic pathogen, because predominantly affects immune-compromised and critically ill patients [3,4]. In this particularly frail population, A. baumannii can cause ventilator-associated pneumonia and bloodstream infections, with an overall reported mortality up to 40% [5,6]. A. baumannii possesses the pernicious, innate ability to evade the commonly used antibiotic therapy.
Generally, A. baumannii resistance mechanisms of intrinsic and acquired antibiotic resistance are categorized into three groups. First, resistance can be achieved by increasing efflux of the antibiotic from the bacteria and thus preventing access to the target, i.e., overexpression of drug efflux pumps. Second, A. baumannii can protect its antibiotic target through genetic mutations or post-translational modifications, i.e., mutations in antibiotic binding targets via genetic insertion sequences. Third, antibiotics can be directly inactivated by hydrolysis, i.e., enzymatic inactivation by beta-lactamases. To consider, multiple different resistance mechanisms are often present at the same time in A. baumannii [7,8,9].
As for other bacteria, MDR A. baumannii can be defined as an organism showing acquired non-susceptibility to at least one agent in three or more antimicrobial categories [10].
Unfortunately, thus far, the clinical trials on the treatment of MDR A. baumannii infections have not provided with conclusive evidence in favor of one antibiotic over another. Antibiotic selection relies upon interpretation of in vitro efficacy, host factors, pharmacokinetic (PK) and pharmacodynamic (PD) profiles [11]. Generally, an antimicrobial combination approach is chosen to overcome the multiple mechanisms of resistance and suppress further resistance. However, the real clinical benefit of combination treatments for infections due to MDR A. baumannii remains unclear.
Moreover, the quiver of antimicrobials retaining in vitro activity against MDR A. baumannii strains is generally restricted to few classes, i.e., aminoglycosides, polymyxins, and some tetracyclines. These antimicrobials share important limitations, such as site-specific pharmacokinetics, emergence of resistance and toxicity.
Recently, studies proposed to use a novel compound, durlobactam, against MDR A. baumannii. Durlobactam is a novel beta-lactamase inhibitor, effective against A. baumannii and MDR A. baumannii when used in combination with another beta-lactamase inhibitor with intrinsic antibacterial activity against A. baumannii, i.e., sulbactam [12].
We performed a systematic review of the literature with the main aim to summarize available evidence supporting durlobactam use in the treatment of MDR A. baumannii infections.

2. Materials and Methods

2.1. Search Strategy and Article Identification

The study protocol was pre-registered on PROSPERO (CRD42022311723) (Tables S1 and S2). Published articles (from June 2017 to November 2020) assessing the main characteristics of durlobactam and the efficacy and safety of durlobactam in the treatment of A. baumannii infections were identified through computerized literature searches using MEDLINE (National Library of Medicine Bethesda MD) and by reviewing the references of retrieved articles. PubMed was searched until 15 February 2022. Combinations of the following search terms were applied: ((durlobactam) OR (ETX2514)). Attempt was made to obtain information about unpublished studies. English language restriction was applied. Studies published only in abstract form, correction articles, reviews, case-reports, editorials, guidance articles or guidelines and clinical trial protocols were excluded from further assessment. Reviewed articles were maintained in a master log and any reason for exclusion from analysis was documented in the rejected log.

2.2. Eligibility Criteria

Studies of any design which reported data on durlobactam were eligible for inclusion in this systematic review.

2.3. Study Selection and Data Extraction

Eligibility assessment and extraction of data were performed independently by two investigators. Each investigator was blinded to the other investigator’s data extraction. In case of disagreement between the two reviewers, a third reviewer was consulted. Data from each study were verified for consistency and accuracy, and then entered into a computerized database. Abstracted information included: author, year of publication, country in which the study was conducted; study design, start and end date of study, health-care setting, sample size; criteria for the diagnosis of bacterial infection, if applicable; proportion of animals or patients receiving antibiotics, if applicable; minimum inhibitory concentration; pharmacokinetics and pharmacodynamics data; animals or patients’ outcome data. For the syntheses, studies were grouped in two groups. Group I: in vitro studies on general characteristics and pharmacokinetic of durlobactam and on the activity of durlobactam against A. baumannii isolates. Group II: clinical studies on the efficacy of durlobactam against infections due to A. baumannii.

2.4. Data Synthesis

Both quantitative and qualitative information were summarized by means of textual descriptions.

2.5. Assessment of Bias

A formal assessment for risk of bias was deemed to have limited utility given the lack of an appropriate assessment tool. Although a risk-of-bias tool has been developed for systematic reviews, many aspects of the tool are not directly relevant to our research question.

3. Results

3.1. Studies Description

Figure 1 shows the selection process of studies included in the systematic review. Through a PubMed search with the search terms “durlobactam” and “ETX2514”, we identified 29 studies published from June 2017 to November 2020. Among the 29, five studies that represented review articles, four studies that did not report data on durlobactam, one study representing a “correction article”, one representing an “expert opinion article”, and two representing case reports were excluded. Of the 16 studies included in this systematic review (Figure 1) [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28], six were clinical trials: four phase I trials [16,17,19,27], one phase II clinical trial [18] and one ongoing phase III clinical trial [28].
Of the 16 studies included in the systematic review, in vitro studies and phase I and phase II studies produce data on general characteristics, pharmacokinetic and pharmacodynamic of durlobactam; seven studies assess the in vitro activity of durlobactam against A. baumannii from patients; one was an ongoing phase III trial study on the efficacy of durlobactam against A. baumannii. The included studies were separated in two different tables with a summary description of their characteristics: Table 1, in vitro studies and Table 2, Phase I, phase II or phase III studies.

3.2. Durlobactam Characteristics

3.2.1. Durlobactam Molecular Structure and Pharmacodynamics

Durlobactam, formerly known as ETX2514, is a novel beta-lactamase inhibitor belonging to the diazabicyclooctanone, boronic acid and pyridine-2-carboxylic acid classes {[(2S,5R)-2-carbamoyl-3-methyl-7-oxo-1,6-diazabicyclo [3.2.1]oct-3-en-6-yl] hydrogen sulfate} [13]. Durlobactam molecular structure differs from other beta-lactamases for its endocyclic double bond and its methyl substituent (Figure 2 and Figure 3). Durlobactam is a polar compound, therefore it is able to penetrate into Gram-negative cells through outer membrane porins, i.e., OmpA [26]. Once into the bacterial cell, durlobactam is carbamoylated on its active, nucleophile serine site. This covalent bond is reversible because the sulfated amine is able to recyclize onto the carbamate [15]. These molecular characteristics confer a favorable binding kinetic and extend durlobactam inhibitory effect to a broad range of beta-lactamases, including Amber class A, C and D beta-lactamases [15]. Durlobactam is able to bind penicillin-binding proteins in a rapid and reversible manner, resulting in an efficient and long-lasting inhibition [15].
Durlobactam potently inhibits clinically relevant Amber class A, C, and D beta-lactamases. More precisely, durlobactam recyclizes and dissociates intact from beta-lactams belonging to class A and C, including AmpC, CTX-M-15, P99, SHV-5, and TEM-1 [15]. Even if durlobactam does not completely dissociates intact from other classes A and D beta-lactams, such as KPC-2, OXA-10, OXA-23, OXA-24, OXA-48, it retains in vitro inhibition of these beta-lactamases. Importantly, durlobactam does not inhibits class B metallo-beta-lactamases [15].
However, the sole durlobactam has minimal antibacterial activity against A. baumannii, mainly due to its ability to bind penicillin-binding proteins (PBP) 2 [13,20,21,22,23,24,25].

3.2.2. Durlobactam Pharmacokinetics

Durlobactam is suitable for intravenous administration, with stability for more than 6 h at room temperature and a water solubility higher than 200 mg/mL [13].
As others beta-lactam inhibitors, durlobactam shows a linear pharmacokinetics, allowing prompt dose adjustments when required. The durlobactam estimated volume of distribution at steady state can be described by a two-compartment model and it is influenced by patient body weight [13,16,17,18,19]. The systemic clearance of durlobactam does not change after single and multiple doses (0.25–2.0 g) [16,17,18,19]. The infusion of durlobactam, alone or in combination with sulbactam, is generally safe and well tolerated [16,17,18,19,27].
The pharmacokinetics and pharmacodynamics of durlobactam in combination with sulbactam have been studied in a phase I trial enrolling 30 healthy adults (ClinicalTrials registration no. NCT03303924) [17].
In this trial, 1 g of durlobactam with 1 g of sulbactam were infused as 3 h intravenous infusion every 6 h, for three consecutive doses. At these regimens, the durlobactam/sulbactam combination was safe and generally well tolerated. In this study the concentration of durlobactam and sulbactam were determined in plasma and epithelial lining fluid. Generally, the total plasma concentrations of each agent resulted higher than those observed in the epithelial lining fluid. The times of the observed maximum plasma concentration (Tmax) were similar for durlobactam and sulbactam, with arithmetic means of 2.62 and 2.56 h, respectively. The means of the minimum plasma concentrations (C min) of durlobactam at 6 h after the first, second, and third infusions were 5.71 ± 1.77, 6.63 ± 1.97, and 5.79 ± 2.08 mg/L, respectively [17].
Of note, following the third infusion, durlobactam showed values of area under the concentration-time curves from 0 to 6 h (AUC0-6) of 109.05 ± 23.44 mg/h/L, the half-life of 1.40 ± 0.18 h and a volume of distribution at steady state of 16.7 ± 3.0 L [17].
Moreover, regarding durlobactam distribution to the epithelial fluid, the AUC0–6 based on mean epithelial lining fluid concentrations resulted 40.1 mg/h/L. The ratio of epithelial lining fluid to total plasma durlobactam concentrations based on the mean AUC0–6 values was 0.37. These results support the use of the durlobactam/sulbactam combination in the treatment of lower respiratory tract bacterial infections [17].
A subsequent, phase II clinical trial performed on 80 patients with complicated urinary tract infection confirmed these findings. In this study, patients were randomized to receive imipenem plus the combination of durlobactam/sulbactam at a regimen of 1 plus 1 g, infused over 3 h every 6 h for 7 days (n: 53), or imipenem plus placebo (n: 27) [18]. The durlobactam/sulbactam combination was generally well tolerated, with 37.7% patients reporting drug-related moderate adverse events such as headache, diarrhea, nausea, and phlebitis. One patient had self-limiting urticaria on day 3 of administration. In one patient, renal function decrease was observed on day 3 of administration. In this phase II trial, durlobactam and sulbactam mean elimination half-lives were of 2.2 and 1.6 h, respectively. The mean steady-state clearance and volume of distribution of durlobactam were 10.3 L/h and 31.6 L, respectively. These values were similar to the mean clearance and volume of distribution estimates for sulbactam (13.4 L/h and 36.0 L, respectively) [18]. On these findings, the authors proposed a durlobactam/sulbactam dose of 1 g of each component to be administered every 6 h via a 3 h infusion to achieve the optimal concentrations [18].
To consider, although the urinary excretion of durlobactam over 48 h is only 66% of the systemic clearance, durlobactam exposure doubled in patients with renal impairment with creatinine clearance lower than 30 mL/min/1.73 m2 [21]. Therefore, changes in durlobactam pharmacokinetics should be considered in patients with severely compromised renal function and hemodialysis should be considered as a factor that can remove nearly half a dose of durlobactam [19].

3.3. In Vitro Studies on the Activity of Durlobactam/Sulbactam against A. baumannii

Durlobactam and Sulbactam Synergistic Bactericidal Activity

The sole durlobactam possesses minimal antibacterial activity against A. baumannii, mainly due to durlobactam ability to bind A. baumannii PBPs [13].
Durlobactam showed a synergistic bactericidal activity in combination with beta-lactam antibiotics [14,20,21,22,23,24,25]. Durlobactam improves the activity of several beta-lactam partners tested against A. baumannii, but the most potent combination has been observed with another beta-lactamase inhibitor, sulbactam [14,20,21,22,23,24,25].
Sulbactam is a well-known beta-lactamase inhibitor which also possesses antibacterial activity against A. baumannii, due to its selective binding to the PBP 1, 2, and 3 [14,20,21,22,23,24,25].
However, in the last decades, MDR A. baumannii strains emerged with a reduced expression of PBP2 and produced beta-lactamases able to degrade sulbactam, such as TEM-1, ADC-30, AmpC and a number of OXAs. Unfortunately, these resistance mechanisms narrowed the clinical utility of the sole sulbactam in MDR A. baumannii infections [23,25].
Therefore, considering that the sole durlobactam does not have significant activity against A. baumannii, its use in combination with sulbactam has been proposed. Exploiting inhibition of beta-lactamases by durlobactam, sulbactam is free to exert its intrinsic A. baumannii bactericidal activity by binding PBPs [23,25].
Sulbactam alone is weakly active against MDR A. baumannii clinical isolates, but when combined with durlobactam, minimal inhibitory concentration (MIC) 90 drops to 16-fold, to 4 mg/L [14,20,21,22,23,24,25]. The durlobactam/sulbactam combination showed activity against Ambler class A, C and D beta-lactamases, possessing a potential utility for the treatment of infections due to MDR A. baumannii [14,20,21,22,23,24,25].
To date, the frequency of A. baumannii spontaneous resistance to the durlobactam/sulbactam combination seems low, even if resistance has been recently reported due to the metallo-beta-lactamase NDM−1 or PBP3 substitutions [20].
In a recent study, the in vitro antibacterial activities of the durlobactam/sulbactam combination were assessed by broth microdilution against 1722 A. baumannii clinical isolates, collected across the five continents in 2016 and 2017 [14]. Over 50% of these samples resulted resistant to carbapenems. Against this strain collection, durlobactam/sulbactam showed MIC50 and MIC90 values of 1 and 2 g/mL, respectively. These MICs were lower than that observed for the sole sulbactam, with a MIC50 of 8 g/mL and a MIC90 of 64 g/mL. This level of activity was found to be consistent across regions, sources of infection, and subsets of resistance phenotypes, including MDR isolates. Of note, in this study colistin was the only traditional antimicrobial with activity similar to durlobactam/sulbactam. Moreover, genome sequencing of the 39 specimens (2.3%) with a durlobactam/sulbactam MIC of 4 g/mL confirmed that these strains encoded either the metallo-beta-lactamase NDM-1, not inhibited by durlobactam, or single amino acid substitutions near the active site of PBP3, i.e., the primary target of sulbactam [14].
Similar results were reported in a study on 246 patients infected by A. baumannii collected between 2012 and 2016 from 94 hospitals in 37 different countries. Antimicrobial susceptibility testing was performed by broth microdilution. The observed durlobactam/sulbactam MIC50 and MIC90 values were 0.25 and 0.5 mg/L, respectively. Sulbactam MIC was lowered 16-fold to 64-fold by the addition of durlobactam. Conversely, the addition of imipenem to the durlobactam/sulbactam combination did not improve activity, with only nine isolates showing higher MIC. These samples encoded the metallo-beta-lactamase NDM-1 [21].
In an in vitro study on 982 samples of A. baumannii infection in China between 2016 and 2018, the observed MIC90 of durlobactam/sulbactam was 2 mg/L [22]. In this study, 84.6% of the collected strains was imipenem resistant. The authors of this study propose a concentration of 4 mg/L as a reasonable preliminary cut-off for evaluation of durlobactam/sulbactam susceptibility in A. baumannii.
Moreover, a recent study used whole-genome sequencing and molecular characterization to relate the presence of carbapenemase genes in 28 A. baumannii clinical isolates from India. The authors found that 93% of the collected samples expressed carbapenemases, including OXA-23, OXA-58 and NDM genes, with over a third expressing dual carbapenemase genes. The presence of these carbapenemase genes resulted in sulbactam resistance (MIC: 16–256 mg/L) in all of the studied isolates [23].
The authors then assessed the efficacy of durlobactam against these strains through in silico intermolecular interaction analysis. Several nonsynonymous single nucleotide polymorphisms were identified in PBP2 and PBP3 sequences, but minimal variations were recorded in the protein backbone dynamics in active-site motifs of wild-type and mutants, which correlated with negligible binding energy fluctuations for the PBP2-durlobactam complex. The authors suggested that the stable interaction profiles of durlobactam with carbapenemases can possibly restore sulbactam activity against both wild type and PBP mutant examinee [23].
Finally, a recent study evaluated the antimicrobial activity of durlobactam/sulbactam against a collection of 112 Brazilian MDR A. baumannii patients [24]. The in vitro activity of durlobactam/sulbactam was evaluated by the broth microdilution method using durlobactam at a fixed concentration of 4 mg/L. In this study, the samples presented a variety of beta-lactamases encoding genes, including several OXAs. Despite the high resistance rates to most antimicrobial agents tested, the authors reported in vitro activity of durlobactam/sulbactam, with MIC90 values of 4 mg/L [24].

3.4. In Vivo Studies on the Efficacy of Durlobactam/Sulbactam to Treat A. baumannii Infections

A study on the efficacy of durlobactam/sulbactam against MDR A. baumannii was performed in the animal model. In thigh and lung murine infection models, durlobactam/sulbactam showed a dose dependent reduction in A. baumannii counts. Bactericidal activity of durlobactam/sulbactam greater than one-log kill was achieved when sulbactam concentrations exceeded the combination MIC of 0.5 mg/L. In this study, no bactericidal activity was observed when sulbactam was administered alone, at the dosage of 15 mg/kg every three hours. The addition of durlobactam to sulbactam increased its activity in a dose-dependent manner [25].
Thus far, in human durlobactam/sulbactam combination has been studied in multiple phase I trials [16,17,19], one phase II trial [18], and in an ongoing phase III trial (NCT03894046) [28].
A phase II trial evaluated the tolerability and pharmacokinetic of durlobactam/sulbactam in patients with complicated urinary tract infections, including acute pyelonephritis. In this study all patients received background therapy with imipenem, in addition to either durlobactam/sulbactam or placebo. The microbiological intent-to-treat population were similar in the two groups, 36 (76.6%) and 17 (81.0%) patients in the durlobactam/sulbactam and in the placebo group, respectively [18].
Currently, a phase III trial, i.e., the ATTACK trial, is evaluating the efficacy and safety of the durlobactam/sulbactam combination in patients with bloodstream infections or hospital-associated bacterial pneumonia due to A. baumannii-calcoaceticus complex (NCT03894046) [28]. The first part of this multinational, pathogen-targeted, randomized, active-controlled, comparator-controlled trial compared two treatment arms: durlobactam/sulbactam (1 g/1 g qid) plus imipenem/cilastin (1 g/1 g qid) versus colistin (2.5 mg/kg bid) plus imipenem/cilastin (1 g/1 g qid). This trial concluded the patients’ recruitment, with a total of 207 enrolled patients, and recently an update on the study results has been released [28].
Approximately 95% of baseline examinees tested were carbapenem resistant. Durlobactam/sulbactam achieved statistical non-inferiority to colistin in the 28-day all-cause mortality efficacy endpoint. Mortality analyses favored durlobactam/sulbactam versus colistin in microbiologically modified intent-to-treat population. Durlobactam/sulbactam mortality was 19.0% compared to 32.3% in the colistin arm (treatment difference of −13.2%; 95% CI: −30.0, 3.5) [28].
At test of cure, there was a statistically significant difference in clinical response favoring durlobactam/sulbactam over colistin (61.9% versus 40.3%, respectively; 95% CI: 2.9, 40.3). Moreover, durlobactam/sulbactam met the primary safety objective of the trial, achieving statistically significant reduction in nephrotoxicity [28].

4. Discussion

Globally, severe infections due to MDR A. baumannii give rise to high mortality rates and remain a great challenge for clinicians [6,7].
Among the reasons to explain this increased mortality, there are the potential toxicity and the suboptimal pharmacokinetics and efficacy of the currently available therapeutic approaches [29,30].
During the last decades, polymyxins represented the most used antimicrobial options against MDR A. baumannii. Polymyxins have been considered as a “last resort” to fight MDR infections, often representing the only antimicrobial to achieve adequate serum levels and MICs. Therefore, the reports of colistin-resistant A. baumannii isolates raises concern, considering the further limitations of antimicrobial options and the high mortality rate associated with these infections [4].
Unfortunately, newer beta-lactams and beta-lactamase inhibitor combinations such as ceftolozane–tazobactam, ceftazidime–avibactam, imipenem–relebactam and meropenem–vaborbactam are clinically ineffective against A. baumannii [31].
Among the most recently developed antimicrobials, cefiderocol and eravacycline may be considered as salvage therapy against MDR A. baumannii [32,33]. However, cefiderocol and eravacycline recently showed disappointing clinical outcomes data against MDR A. baumannii [12,32,33,34]. Regarding cefiderocol, it showed in vitro activity against MDR A. baumannii, but its in vivo efficacy is variable [34].
In summary, the in vitro studies demonstrated that durlobactam and sulbactam permeates the A. baumannii outer membrane protein A (OmpA) [26]. After penetrating the bacterial cell, durlobactam recyclizes and dissociates intact from several beta-lactamases [15]. Against A. baumannii isolates, durlobactam/sulbactam showed MIC50 values ranging between 0.5 and 1 mg/mL and MIC90 values from 2 to 4 mg/mL [14,20,21,22].
The clinical studies demonstrated that durlobactam is safe and generally well tolerated when administered in combination with sulbactam [16,27]. Durlobactam showed a half-life of 1.40 ± 0.18 h and a mean steady-state clearance and volume of distribution of 10.3 L/h and 31.6 L, respectively [17,18]. The predominant clearance mechanism is through renal excretion [16]. Decreasing renal function increases peak plasma concentration and AUC in a generally linear manner and hemodialysis is effective at removing both durlobactam and sulbactam from plasma [19]. In an ongoing phase III clinical trial comparing durlobactam/sulbactam and colistin for the treatment of carbapenem-resistant A. baumannii infection, durlobactam/sulbactam met the primary efficacy endpoint of all-cause mortality at 28 days (durlobactam/sulbactam mortality was 19.0% versus 32.3% of the colistin arm, treatment difference of −13.2%; 95% CI: −30.0, 3.5) [28].
In our systematic review, we included 16 studies dealing with the possible use of durlobactam in the treatment of MDR A. baumannii infections. The included studies were extremely heterogeneous, being studies performed in different settings with different designs.
Moreover, we could identify only one phase III clinical trial, providing data on the efficacy of durlobactam and sulbactam in the treatment of infections due to A. baumannii.
The results coming from the first studies evaluating the use of durlobactam against A. baumannii placed great hope in this novel beta-lactam inhibitor. The combination of durlobactam and sulbactam shows excellent in vitro potency against A. baumannii isolates, including MDR isolates that were resistant to carbapenems, aminoglycosides, tetracyclines and polymyxins.
Moreover, durlobactam pharmacokinetic and pharmacodynamic profiles suggest that it may be considered in the treatment of the most common A. baumanni infections, such as blood stream infections and pneumonia.
To consider, among the weaknesses of this novel compound, durlobactam does not inhibit class B beta-lactamases, i.e., NDM-1. This may halt durlobactam future usefulness, although fortunately the most recent surveillance studies report that infection of A. baumannii producing B metallo-beta-lactamases are globally rare [20].
Another factor that may halt the benefit of durlobactam/sulbactam use against infection due to A. baumannii is the potential development of resistance.

5. Conclusions

Certainly, data from randomized clinical trials will be required to confirm the effectiveness of durlobactam/sulbactam to treat infections caused by A. baumannii. In this regard, we are awaiting with interest the definitive results from the ongoing phase III open label clinical trial ATTACK (Acinetobacter Treatment Trial Against Colistin; NCT03894046). In the ATTACK trial, patients were randomized to receive either durlobactam/sulbactam plus imipenem/cilastatin or colistin plus imipenem/cilastatin. Probably, this therapy regimen is more reflective of real-world practice with MDR A. baumannii than monotherapy regimens employed in previous trials. However, this may lead to difficulty in interpreting the stand-alone efficacy of the durlobactam/sulbactam combination.
Positive results from this ongoing trial and from future clinical trials would enable again the possibility to treat infections caused by MDR A. baumannii with this unusual, dual beta-lactamase inhibitor combination.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11123258/s1, Table S1: PRISMA abstract checklist; Table S2: PRISMA checklist.

Author Contributions

Conceptualization, N.P. and G.G.; methodology, G.G.; software, F.S. and G.G.; validation, N.P. and F.T.; formal analysis, F.S. and G.G.; investigation, F.S. and G.G.; resources, G.G. and F.T.; data curation, F.S. and G.G.; writing—original draft preparation, G.G.; writing—review and editing, N.P. and G.G.; visualization, N.P.; supervision, N.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest. The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Sugden, R.; Kelly, R.; Davies, S. Combatting antimicrobial resistance globally. Nat. Microbiol. 2016, 1, 16187. [Google Scholar] [CrossRef]
  2. Sherry, N.; Howden, B. Emerging Gramnegative resistance to last-line antimicrobial agents fosfomycin, colistin and ceftazidime-avibactam—Epidemiology, laboratory detection and treatment implications. Expert Rev. Anti. Infect. Ther. 2018, 16, 289–306. [Google Scholar] [CrossRef] [PubMed]
  3. Bassetti, M.; Labate, L.; Russo, C.; Vena, A.; Giacobbe, D.R. Therapeutic options for difficult-to-treat Acinetobacter baumannii infections: A 2020 perspective. Expert Opin. Pharmacother. 2021, 22, 167–177. [Google Scholar] [CrossRef]
  4. Harding, C.M.; Hennon, S.W.; Feldman, M.F. Uncovering the mechanisms of Acinetobacter baumannii virulence. Nat. Rev. Microbiol. 2018, 16, 91–102. [Google Scholar] [CrossRef]
  5. Giannella, M.; Bussini, L.; Pascale, R.; Bartoletti, M.; Malagrinò, M.; Pancaldi, L.; Toschi, A.; Ferraro, G.; Marconi, L.; Ambretti, S.; et al. Prognostic Utility of the New Definition of Difficult-to-Treat Resistance Among Patients With Gram-Negative Bloodstream Infections. Open Forum Infect. Dis. 2019, 6, ofz505. [Google Scholar] [CrossRef] [PubMed]
  6. Kadri, S.S.; Adjemian, J.; Lai, Y.L.; Spaulding, A.B.; Ricotta, E.; Prevots, D.R.; Palmore, T.N.; Rhee, C.; Klompas, M.; Dekker, J.P.; et al. Difficult-to-Treat Resistance in Gram-negative Bacteremia at 173 US Hospitals: Retrospective Cohort Analysis of Prevalence, Predictors, and Outcome of Resistance to All First-line Agents. Clin. Infect. Dis. 2018, 67, 1803–1814. [Google Scholar] [CrossRef] [Green Version]
  7. Vrancianu, C.O.; Gheorghe, I.; Czobor, I.B.; Chifiriuc, M.C. Antibiotic Resistance Profiles, Molecular Mechanisms and Innovative Treatment Strategies of Acinetobacter baumannii. Microorganisms 2020, 8, 935. [Google Scholar] [CrossRef]
  8. Asif, M.; Alvi, I.A.; Rehman, S.U. Insight into Acinetobacter baumannii: Pathogenesis, global resistance, mechanisms of resistance, treatment options, and alternative modalities. Infect. Drug Resist. 2018, 11, 1249–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Viehman, J.A.; Nguyen, M.H.; Doi, Y. Treatment options for carbapenem-resistant and extensively drug-resistant Acinetobacter baumannii infections. Drugs 2014, 74, 1315–1333. [Google Scholar] [CrossRef] [Green Version]
  10. Magiorakos, A.P.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F.; Kahlmeter, G.; Olsson-Liljequist, B.; et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: An international expert proposal for interim standard definitions for acquired resistance. Clin. Microbiol. Infect. 2012, 18, 268–281. [Google Scholar] [CrossRef] [Green Version]
  11. Abdul-Mutakabbir, J.C.; Griffith, N.C.; Shields, R.K.; Tverdek, F.P.; Escobar, Z.K. Contemporary Perspective on the Treatment of Acinetobacter baumannii Infections: Insights from the Society of Infectious Diseases Pharmacists. Infect. Dis. Ther. 2021, 10, 2177–2202. [Google Scholar] [CrossRef] [PubMed]
  12. Yahav, D.; Giske, C.G.; Grāmatniece, A.; Abodakpi, H.; Tam, V.H.; Leibovici, L. New β-Lactam-β-Lactamase Inhibitor Combinations. Clin. Microbiol. Rev. 2020, 34, e00115-20. [Google Scholar] [CrossRef]
  13. Durand-Réville, T.F.; Guler, S.; Comita-Prevoir, J.; Chen, B.; Bifulco, N.; Huynh, H.; Lahiri, S.; Shapiro, A.B.; McLeod, S.M.; Carter, N.M.; et al. ETX2514 is a broad-spectrum β-lactamase inhibitor for the treatment of drug-resistant Gram-negative bacteria including Acinetobacter baumannii. Nat. Microbiol. 2017, 2, 17104. [Google Scholar] [CrossRef]
  14. McLeod, S.M.; Moussa, S.H.; Hackel, M.A.; Miller, A.A. In Vitro Activity of Sulbactam-Durlobactam against Acinetobacter baumannii-calcoaceticus Complex Isolates Collected Globally in 2016 and 2017. Antimicrob. Agents Chemother. 2020, 64, e02534-19. [Google Scholar] [CrossRef]
  15. Shapiro, A.B.; Gao, N.; Jahić, H.; Carter, N.M.; Chen, A.; Miller, A.A. Reversibility of Covalent, Broad-Spectrum Serine β-Lactamase Inhibition by the Diazabicyclooctenone ETX2514. ACS Infect. Dis. 2017, 3, 833–844. [Google Scholar] [CrossRef] [PubMed]
  16. Lickliter, J.D.; Lawrence, K.; O’Donnell, J.; Isaacs, R. Safety, Pharmacokinetics, and Drug-Drug Interaction Potential of Intravenous Durlobactam, a β-Lactamase Inhibitor, in Healthy Subjects. Antimicrob. Agents Chemother. 2020, 64, e00071-20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Rodvold, K.A.; Gotfried, M.H.; Isaacs, R.D.; O’Donnell, J.P.; Stone, E. Plasma and Intrapulmonary Concentrations of ETX2514 and Sulbactam following Intravenous Administration of ETX2514SUL to Healthy Adult Subjects. Antimicrob. Agents Chemother. 2018, 62, e01089-18. [Google Scholar] [CrossRef] [Green Version]
  18. Sagan, O.; Yakubsevitch, R.; Yanev, K.; Fomkin, R.; Stone, E.; Hines, D.; O’Donnell, J.; Miller, A.; Isaacs, R.; Srinivasan, S. Pharmacokinetics and Tolerability of Intravenous Sulbactam-Durlobactam with Imipenem-Cilastatin in Hospitalized Adults with Complicated Urinary Tract Infections, Including Acute Pyelonephritis. Antimicrob. Agents Chemother. 2020, 64, e01506-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. O’Donnell, J.; Preston, R.A.; Mamikonyan, G.; Stone, E.; Isaacs, R. Pharmacokinetics, Safety, and Tolerability of Intravenous Durlobactam and Sulbactam in Subjects with Renal Impairment and Healthy Matched Control Subjects. Antimicrob. Agents Chemother. 2019, 63, e00794-19. [Google Scholar] [CrossRef] [Green Version]
  20. McLeod, S.M.; Shapiro, A.B.; Moussa, S.H.; Johnstone, M.; McLaughlin, R.E.; De Jonge, B.L.M.; Miller, A.A. Frequency and Mechanism of Spontaneous Resistance to Sulbactam Combined with the Novel β-Lactamase Inhibitor ETX2514 in Clinical Isolates of Acinetobacter baumannii. Antimicrob. Agents Chemother. 2018, 62, e01576-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Seifert, H.; Müller, C.; Stefanik, D.; Higgins, P.G.; Miller, A.; Kresken, M. In vitro activity of sulbactam/durlobactam against global isolates of carbapenem-resistant Acinetobacter baumannii. J. Antimicrob. Chemother. 2020, 75, 2616–2621. [Google Scholar] [CrossRef]
  22. Yang, Q.; Xu, Y.; Jia, P.; Zhu, Y.; Zhang, J.; Zhang, G.; Deng, J.; Hackel, M.; Bradford, P.A.; Reinhart, H. In vitro activity of sulbactam/durlobactam against clinical isolates of Acinetobacter baumannii collected in China. J. Antimicrob. Chemother. 2020, 75, 1833–1839. [Google Scholar] [CrossRef]
  23. Naha, A.; Vijayakumar, S.; Lal, B.; Shankar, B.A.; Chandran, S.; Ramaiah, S.; Veeraraghavan, B.; Anbarasu, A. Genome sequencing and molecular characterisation of XDR Acinetobacter baumannii reveal complexities in resistance: Novel combination of sulbactam-durlobactam holds promise for therapeutic intervention. J. Cell. Biochem. 2021, 122, 1946–1957. [Google Scholar] [CrossRef]
  24. Nodari, C.S.; Santos, F.F.; Kurihara, M.N.L.; Valiatti, T.B.; Cayô, R.; Gales, A.C. In vitro activity of sulbactam/durlobactam against extensively drug-resistant Acinetobacter baumannii isolates belonging to South American major clones. J. Glob. Antimicrob. Resist. 2021, 25, 363–366. [Google Scholar] [CrossRef]
  25. Barnes, M.D.; Kumar, V.; Bethel, C.R.; Moussa, S.H.; O’Donnell, J.; Rutter, J.D.; Good, C.E.; Hujer, K.M.; Hujer, A.M.; Marshall, S.H.; et al. Targeting Multidrug-Resistant Acinetobacter spp.: Sulbactam and the Diazabicyclooctenone β-Lactamase Inhibitor ETX2514 as a Novel Therapeutic Agent. mBio 2019, 10, e00159-19. [Google Scholar] [CrossRef] [Green Version]
  26. Iyer, R.; Moussa, S.H.; Durand-Réville, T.F.; Tommasi, R.; Miller, A. Acinetobacter baumannii OmpA Is a Selective Antibiotic Permeant Porin. ACS Infect. Dis. 2018, 4, 373–381. [Google Scholar] [CrossRef]
  27. O’Donnell, J.; Maloney, K.; Steidler, M.; Morrison, R.; Isaacs, R. A randomized, double-blind, placebo- and positive-controlled crossover study of the effects of durlobactam on cardiac repolarization in healthy subjects. Clin. Transl. Sci. 2021, 14, 1423–1430. [Google Scholar] [CrossRef]
  28. ATTACK Trial. Available online: https://investors.entasistx.com/news-releases/news-release-details/entasis-therapeutics-announces-positive-topline-results (accessed on 1 May 2022).
  29. Tsuji, B.T.; Pogue, J.M.; Zavascki, A.P.; Paul, M.; Daikos, G.L.; Forrest, A.; Giacobbe, D.R.; Viscoli, C.; Giamarellou, H.; Karaiskos, I.; et al. International Consensus Guidelines for the Optimal Use of the Polymyxins: Endorsed by the American College of Clinical Pharmacy (ACCP), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), Infectious Diseases Society of America (IDSA), International Society for Anti-infective Pharmacology (ISAP), Society of Critical Care Medicine (SCCM), and Society of Infectious Diseases Pharmacists (SIDP). Pharmacotherapy 2019, 39, 10–39. [Google Scholar]
  30. Panidis, D.; Markantonis, S.L.; Boutzouka, E.; Karatzas, S.; Baltopoulos, G. Penetration of gentamicin into the alveolar lining fluid of critically ill patients with ventilator-associated pneumonia. Chest 2005, 128, 545–552. [Google Scholar] [CrossRef] [Green Version]
  31. Tooke, C.L.; Hinchliffe, P.; Bragginton, E.C.; Colenso, C.K.; Hirvonen, V.H.A.; Takebayashi, Y.; Spencer, J. β-Lactamases and β-Lactamase Inhibitors in the 21st Century. J. Mol. Biol. 2019, 431, 3472–3500. [Google Scholar] [CrossRef]
  32. Portsmouth, S.; Van Veenhuyzen, D.; Echols, R.; Machida, M.; Ferreira, J.C.A.; Ariyasu, M.; Tenke, P.; Nagata, T.D. Cefiderocol versus imipenem-cilastatin for the treatment of complicated urinary tract infections caused by Gram-negative uropathogens: A phase 2, randomised, double-blind, non-inferiority trial. Lancet Infect. Dis. 2018, 18, 1319–1328. [Google Scholar] [CrossRef]
  33. Solomkin, J.; Evans, D.; Slepavicius, A.; Lee, P.; Marsh, A.; Tsai, L.; Sutcliffe, J.A.; Horn, P. Assessing the Efficacy and Safety of Eravacycline vs Ertapenem in Complicated Intra-abdominal Infections in the Investigating Gram-Negative Infections Treated With Eravacycline (IGNITE 1) Trial: A Randomized Clinical Trial. JAMA Surg. 2017, 152, 224–232. [Google Scholar] [CrossRef] [PubMed]
  34. Bassetti, M.; Echols, R.; Matsunaga, Y.; Ariyasu, M.; Doi, Y.; Ferrer, R.; Lodise, T.P.; Naas, T.; Niki, Y.; Paterson, D.L.; et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): A randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect. Dis. 2021, 21, 226–240. [Google Scholar] [CrossRef]
Figure 1. Flowchart depicting the selection process of studies included in the systematic review.
Figure 1. Flowchart depicting the selection process of studies included in the systematic review.
Jcm 11 03258 g001
Figure 2. Durlobactam chemical structure.
Figure 2. Durlobactam chemical structure.
Jcm 11 03258 g002
Figure 3. X-ray crystal structure of durlobactam in covalent complex with OXA-24/40 at 2.0 Å resolution (PDB: 6MPQ). Reproduced from Barnes MD et al. (2019) with permission from the authors.
Figure 3. X-ray crystal structure of durlobactam in covalent complex with OXA-24/40 at 2.0 Å resolution (PDB: 6MPQ). Reproduced from Barnes MD et al. (2019) with permission from the authors.
Jcm 11 03258 g003
Table 1. Summary description of in vitro studies on durlobactam included in the systematic review.
Table 1. Summary description of in vitro studies on durlobactam included in the systematic review.
Author &
Country
YearStudy AimStudy Design
& Setting
MethodsStudy Results
Durand-Réville TF et al. [13] US2017To modify the
diazabicyclooctanone
scaffold shared by
traditional beta-lactamase inhibitors to extend its
spectrum of activity to
include a broad range of class D, A and C
beta-lactamases
In vitro study LaboratoryReverse phase chromatographiesDurlobactam was
discovered. A novel broad-spectrum serine beta-lactamase inhibitor to be combined with a beta-lactam to treat
patients infected by Gram-negative bacteria
Iyer R et al. [26] US2018To study the permeation characteristics of the A. baumannii outer membrane porin “OmpA” of substrates including durlobactamIn vitro study LaboratoryA specific whole-cell approach called titrable outer membrane permeability assay system was used to characterize the structure porin-permeation relationships. Antibacterial assays used a standard MIC format in Mueller-Hinton cation-adjusted broth. Contribution of OmpA to bacterial fitness was evaluated using a murine thigh model of infectionDurlobactam and
sulbactam are substrates of OmpA, with a potential for structure-porin-
permeation relationships
Shapiro AB et al. [15] US2017To assess the reversibility of durlobactam acylation of a set of beta-lactamasesIn vitro study Laboratory“Jump Dilution” to measure beta-lactamases off-rate
constants; Mass spectrometry to assess durlobactam
degradation
Durlobactam recyclized and dissociated intact from beta-lactamases AmpC, CTX-M-15, P99, SHV-5 and TEM-1
Author &
Country
YearStudy aimSettingAntimicrobial susceptibility testingBacterial
isolates
Infection sourceStudy result and minimum inhibitory
concentration
McLeod SM
et al. [14] US
2020To report the in vitro activity of durlobactam/
sulbactam and comparator
antibiotics against clinical isolates of A. baumanni-
calcoaceticus complex
Isolates
collected in 2016 and 2017 from 209 medical centers in 31 different countries
In-house broth microdilution panels1722 Acinetobacter baumannii- calcoaceticus complex 1420 A. baumannii isolatesbloodstream (13.9%),
intra-
abdominal (3.8%),
respiratory tract (61.2%),
urinary tract (18.3%), skin and soft tissue (0.8%), other (2.0%)
Durlobactam/sulbactam had a MIC50/MIC90 of 1 and 4 mg/L, respectively, compared to a MIC50/MIC90 of 16/64 mg/L for sulbactam alone
McLeod SM
et al. [20] US
2018To determine spontaneous resistance to sulbactam in the presence of 4 mg/L
durlobactam
Pharmaceutical industry
laboratory
Susceptibility testing was performed in cation-adjusted Mueller–Hinton broth4 A. baumannii clinical isolates-Durlobactam/sulbactam had MICs between 0.5 and 1 mg/L. The frequency of resistance to durlobactam/sulbactam (4xMIC) was lower than 9.0 × 1010
Seifert H et al. [21] US, Germany2020To evaluate the activity of durlobactam/sulbactam against global isolates of carbapenem- resistant A. baumanniiIsolates collected between 2012 and 2016 from 94 hospitals in 37 different countriesBroth microdilution246 carbapenem-resistant A. baumannii isolatesThe isolates were collected from various body sitesDurlobactam/sulbactam MIC50 and MIC90 values were 0.25 and 0.5 mg/L, respectively
Yang Q et al.
[22] China
2020To determine the in vitro activity of durlobactam/sulbactam against A. baumannii isolatesA. baumannii clinical isolates were collected from 22 sites across China between 2016 and 2018Frozen
microbroth
dilution panels
982 A. baumannii clinical isolates. (831 (84.6%) were imipenem resistant)Lower respiratory tract (715 isolates, 72.8%), intra- abdominal (170 isolates, 17.3%), urinary tract (59 isolates, 6.0%), skin and soft tissue (35 isolates, 3.6%) and blood (3 isolates, 0.3%)Sulbactam/durlobactam was equally active against A. baumannii isolates from all infection types: the MIC90 was 2 mg/L for isolates from lower respiratory tract, intraabdominal and skin infections and 1 mg/L for urinary tract isolates. The MIC90 of sulbactam/durlobactam was 0.5 and 2 mg/L for carbapenem-susceptible and -resistant A. baumannii isolates, respectively
Nodari CS et al. [24] Brazil2021To evaluate the antimicrobial activity of durlobactam/sulbactam against a collection of MDR A. baumannii isolatesIsolates collected between 2000 and 2019 in BrazilBroth microdilution method using durlobactam at a fixed concentration of 4 mg/L112 MDR A. baumannii
clinical isolates
-Durlobactam/sulbactam MIC90 values of 4 mg/L
Barnes MD et al. [25] US2019To test the susceptibility of A. baumannii isolates to durlobactam/sulbactam and characterize the ability of durlobactam to inhibit class C and class D beta lactamases Acinetobacter spp.Isolates collected in at four medical centers in USStrains were phenotypically characterized using Mueller–Hinton agar dilution98 A. baumannii clinical isolates. (43 were carbapenem resistant)-The MIC90 of the sole sulbactam was 32 mg/L, in comparison to a durlobactam/sulbactam MIC90 of 2 mg/L
Naha et al.
[23] India
2021To evaluate the efficacy of the durlobactam/sulbactam against clinical isolates of A. baumanniiIsolates collected between 2018 and 2019 in Indian hospitalKirby–Bauer disc-diffusion method and broth micro-dilution. The efficacy of durlobactam was assessed through in silico intermolecular interaction analysis28 A. baumannii clinical isolatesThe 28 clinical strains were isolated from blood (n: 21) and sputum
(n: 7)
93% of isolates expressed carbapenemases. Presence of carbapenemase genes resulted in sulbactam resistance (MIC: 16–256 mg/L) in all isolates. The intermolecular interactions of durlobactam and sulbactam with their respective targets displayed strong binding affinities against the strains of MDR A. baumannii
Table 2. Summary description of phase I, II and III clinical trials on durlobactam included in the systematic review.
Table 2. Summary description of phase I, II and III clinical trials on durlobactam included in the systematic review.
Author,
Year
&Country
Study
Population
Study DesignStudy AimSettingMethodsStudy Results
Lickliter JD et al. [16] 2020
US,
Australia
124Randomized, double-blind, placebo-
controlled
phase I
clinical trial
To evaluate the safety and pharmacokinetics of durlobactam,
durlobactam/sulbactam and imipenem-
cilastatin in healthy subjects
A single
clinical site in Australia,
between 2016 and 2017
4-part study.
Part A was a single-
ascending-dose escalation phase. Part B was a
multiple-ascending-dose escalation phase. In parts
C and D, the drug–drug
interaction potential and the safety of
durlobactam/sulbactam was investigated after
single and multiple doses
On a total of 124
subjects, durlobactam was generally safe and well tolerated when it was administered either alone or in combination with sulbactam or imipenem–cilastatin. Renal excretion was the predominant clearance mechanism
Rodvold KA et al. [17] 2018
US
30Phase I,
multiple-dose open-label
pharmacokinetic study in healthy adults
To determine and
compare plasma, epithelial lining fluid and alveolar
macrophage
concentrations of
durlobactam and
sulbactam following
intravenous
administration
A single
private
facility in US, during 2017
Liquid chromatography-tandem mass spectrometry following repeated dosing of 1 g of durlobactam and 1 g of sulbactam every 6 h, for a total
of 3 doses. A
bronchoalveolar lavage was performed once in each subject
In 30 healthy adults
subjects,
durlobactam/sulbactam was safe and tolerated. Following the third
infusion, durlobactam showed values of AUC 0–6 of 109.05 ± 23.44 mg/h/L, the half-life of 1.40 ± 0.18 h and a volume of distribution at steady state of 16.7 ± 3.0 L. Durlobactam
AUC 0–6 based on mean epithelial lining fluid concentrations
resulted 40.1 mg/h/L
O’Donnel J et al.
[27] 2021
US
32Placebo-
controlled,
single-infusion, phase I
clinical trial
To evaluate the effect of a single
supratherapeutic dose of durlobactam on the heart rate corrected
QT interval in healthy
volunteers
Private
clinical
pharmacology center
32 healthy volunteers were randomized to 1 of 6
sequences that included a single infusion of
durlobactam 4 g, a single infusion of placebo, and a single infusion of placebo plus a single oral dose of moxifloxacin
400 mg given open-label at the end of the infusions.
In each treatment period, Holter electrocardiogram measurements were
obtained
No significant change was observed with
durlobactam in
comparison to placebo. A concentration-QT analysis demonstrated no significant effect of durlobactam on
electrocardiogram
parameters, including QT interval prolongation
O’Donnell J et al.
[19] 2019
US
34Phase I
open-label, non-randomized study
To evaluate the
effects of various
degrees of renal
impairment, including subjects with end-stage renal disease on
hemodialysis, on the pharmacokinetics and tolerability of
durlobactam and
sulbactam
Three clinical sites in the United States between 2017 and 2018Study included 8 patients with normal renal
function, 26 patients with renal impairment. For healthy subjects and those with mild or moderate
renal impairment, single
1 g dose each of
durlobactam and
sulbactam via 3 h
infusion was administered, and for severe renal impairment, 500 mg doses were administered. For subjects on hemodialysis, 500 mg doses each of
durlobactam and sulbactam were administered post-hemodialysis and pre-hemodialysis, with a
1-week washout between doses
Renal impairment had no effect of the safety profile of durlobactam and sulbactam.
Decreasing renal
function increased peak plasma concentration and AUC to
durlobactam and
sulbactam in a generally linear manner.
Durlobactam exposure doubled in patients with renal impairment with creatinine
clearance lower than
30 mL/min/1.73 m2.
Hemodialysis was
effective at removing both durlobactam and sulbactam
from plasma
Sagan O et al.
[18] 2020
Ukraine, Belarus, Bulgaria, Russia, US
80Phase II
double-blind, randomized, placebo-
controlled trial
To evaluate the
tolerability and
pharmacokinetic of durlobactam/sulbactam in patients with
complicated urinary tract infections
20 clinical sites in
Belarus,
Bulgaria,
Russia, and Ukraine
between
January 2018 and May 2018
All the included patients received background
therapy with imipenem,
in addition to either
durlobactam/sulbactam
or placebo
The mean steady-state clearance and VD of
durlobactam were 10.3 L/h and 31.6
liters, respectively. The microbiological
intent-to-treat
population were similar in the two groups, 36 (76.6%) and 17 (81.0%) patients in the
durlobactam/sulbactam and in the placebo group, respectively
ATTACK trial
[28] 2021
US
207Open label,
randomized phase III
clinical trial
To evaluate the efficacy and safety of
durlobactam/sulbactam in the treatment of
patients with infections caused by
A. Baumannii-
calcoaceticus Complex
17 countries in the world, 95 clinical sites between 2019 and 20212-part study, with Part A being the randomized, controlled portion of the study in patients with
A. baumannii hospital-
acquired bacterial
pneumonia or bacteremia. Part B is the single-group portion of the study and includes A. baumannii
infections that are resistant to colistin. Part A contemplated two treatment arms: durlobactam/sulbactam
(1 g/1 g qid) plus imipenem/cilastin (1 g/1 g qid) Versus colistin
(2.5 mg/kg bid) plus imipenem/cilastin
(1 g/1 g qid)
This clinical trial is
ongoing. Recruitment phase ended with 207 participants.
Durlobactam/sulbactam met the primary
efficacy endpoint of 28-day all-cause mortality compared to colistin in the carbapenem-
resistant Acinetobacter microbiologically
modified intent-to-treat population
(125 patients).
Durlobactam/sulbactam mortality was 19.0% (12/63) compared to 32.3% (20/62) in the
colistin arm (treatment difference of −13.2%; 95% CI: −30.0, 3.5).
AUC: area under the concentration-time curve; AUC 0–6: area under the concentration–time curve from 0 to 6 h; VD: volume of distribution.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Granata, G.; Taglietti, F.; Schiavone, F.; Petrosillo, N. Durlobactam in the Treatment of Multidrug-Resistant Acinetobacter baumannii Infections: A Systematic Review. J. Clin. Med. 2022, 11, 3258. https://doi.org/10.3390/jcm11123258

AMA Style

Granata G, Taglietti F, Schiavone F, Petrosillo N. Durlobactam in the Treatment of Multidrug-Resistant Acinetobacter baumannii Infections: A Systematic Review. Journal of Clinical Medicine. 2022; 11(12):3258. https://doi.org/10.3390/jcm11123258

Chicago/Turabian Style

Granata, Guido, Fabrizio Taglietti, Francesco Schiavone, and Nicola Petrosillo. 2022. "Durlobactam in the Treatment of Multidrug-Resistant Acinetobacter baumannii Infections: A Systematic Review" Journal of Clinical Medicine 11, no. 12: 3258. https://doi.org/10.3390/jcm11123258

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop