Next Article in Journal
Newly Diagnosed Crohn’s Disease Patients in India and Israel Display Distinct Presentations and Serological Markers: Insights from Prospective Cohorts
Previous Article in Journal
Total Hip Arthroplasty Patients with Distinct Postoperative Fibrinolytic Phenotypes Require Different Antifibrinolytic Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacokinetics, Pharmacodynamics, and Dosing Considerations of Novel β-Lactams and β-Lactam/β-Lactamase Inhibitors in Critically Ill Adult Patients: Focus on Obesity, Augmented Renal Clearance, Renal Replacement Therapies, and Extracorporeal Membrane Oxygenation

1
Department of Clinical Pharmacy, Critical Care, Hamad Medical Corporation, Doha 3050, Qatar
2
Department of Pharmacy, Infectious Diseases, Hamad Medical Corporation, Doha 3050, Qatar
3
School of Medicine, Jordan University of Science and Technology, Irbid 3030, Jordan
4
Clinical Pharmacy and Practice, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
5
Department of Anesthesiology, Intensive Care and Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
6
Dipartimento di Scienze Biotecnologiche di Base Cliniche Intensivologiche e Perioperatorie, Universita’ Cattolica del Sacro Cuore, 00168 Rome, Italy
7
Department of Medicine, Critical Care Services, Hamad Medical Corporation, P.O. Box 305, Doha 3050, Qatar
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(23), 6898; https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11236898
Submission received: 25 September 2022 / Revised: 15 November 2022 / Accepted: 18 November 2022 / Published: 22 November 2022
(This article belongs to the Section Pharmacology)

Abstract

:
Objective: Dose optimization of novel β-lactam antibiotics (NBLA) has become necessary given the increased prevalence of multidrug-resistant infections in intensive care units coupled with the limited number of available treatment options. Unfortunately, recommended dose regimens of NBLA based on PK/PD indices are not well-defined for critically ill patients presenting with special situations (i.e., obesity, extracorporeal membrane oxygenation (ECMO), augmented renal clearance (ARC), and renal replacement therapies (RRT)). This review aimed to discuss and summarize the available literature on the PK/PD attained indices of NBLA among critically ill patients with special circumstances. Data Sources: PubMed, MEDLINE, Scopus, Google Scholar, and Embase databases were searched for studies published between January 2011 and May 2022. Study selection and data extraction: Articles relevant to NBLA (i.e., ceftolozane/tazobactam, ceftazidime/avibactam, cefiderocol, ceftobiprole, imipenem/relebactam, and meropenem/vaborbactam) were selected. The MeSH terms of “obesity”, “augmented renal clearance”, “renal replacement therapy”, “extracorporeal membrane oxygenation”, “pharmacokinetic”, “pharmacodynamic” “critically ill”, and “intensive care” were used for identification of articles. The search was limited to adult humans’ studies that were published in English. A narrative synthesis of included studies was then conducted accordingly. Data synthesis: Available evidence surrounding the use of NBLA among critically ill patients presenting with special situations was limited by the small sample size of the included studies coupled with high heterogeneity. The PK/PD target attainments of NBLA were reported to be minimally affected by obesity and/or ECMO, whereas the effect of renal functionality (in the form of either ARC or RRT) was more substantial. Conclusion: Critically ill patients presenting with special circumstances might be at risk of altered NBLA pharmacokinetics, particularly in the settings of ARC and RRT. More robust, well-designed trials are still required to define effective dose regimens able to attain therapeutic PK/PD indices of NBLA when utilized in those special scenarios, and thus aid in improving the patients’ outcomes.

1. Background

The incidence of deaths owing to sepsis/septic shock in noncardiac patients admitted to intensive care units (ICUs) continues to increase, despite advances in management and personalized treatments Despite advances in management and personalized treatments, mortality rate of infected patients was found to be more than double of that noninfected patients admitted to intensive care units (ICUs) [1,2]. Sepsis-causing multi-drug-resistant (MDR) pathogens, including Pseudomonasaeruginosa (PsA), Enterobacterspecies, methicillin-resistant Staphylococcus aureus, and Enterococcus faecium, have become a remarkable burden, especially for severely ill patients [3,4]. Effective therapy for these patients relies not only on early diagnosis and timely antimicrobial administration, but also on dose-regimen optimization to achieve optimal pharmacokinetic (PK)/pharmacodynamic (PD) targets associated with maximal efficacy.
The interest in the use of novel β-lactam antibiotics (NBLA), which include both novel β-lactam and novel β-lactam/β-lactamase inhibitors (BLBLI) (namely ceftolozane/tazobactam (C/T), ceftazidime/avibactam (CAZ/AVI), cefiderocol, ceftobiprole, imipenem/relebactam (IMI/REL), and meropenem/vaborbactam (MEV)) as alternatives against MDR pathogens among critically ill patients has recently increased. Recommended dosing regimens of antimicrobials are often based on PK studies in healthy volunteers. However, critically ill patients are known to present with various physiological/pathological changes that result in substantial alterations of achieved drug concentrations, along with other key PK parameters [5,6]. Moreover, extracorporeal support (including renal replacement therapies (RRT) and extracorporeal membrane oxygenation (ECMO)) may sometimes be required, further contributing to PK-related variabilities [7,8,9]. Previous researchers utilizing the recommended package insert doses of conventional β-lactams in such heterogeneous populations revealed the risk of both under-/over-exposures depending on the underlying pathology and clinical condition [10,11,12]. Despite growing use in critically ill patients, limited evidence exists describing such variabilities for NBLA. Recently, the adequacy of the current monograph-recommended dosing of three different NBLA in ICU patients was investigated using Monte Carlo simulation [13]. While standard dosing resulted in adequate attainment of a PD index of 40–60% of free drug concentration above MIC (%ƒT > MIC) for CAZ/AVI, C/T, and MEV, such dosing did not attain more aggressive targets (i.e., 100%ƒT > 1–4 × MIC). Although some reviews had recently described altered PK/PD for various NBLA during critical illness [14,15], sub-populations that warrant special considerations including obesity, augmented renal clearance (ARC), RRT modalities, and ECMO were commonly excluded or not comprehensively addressed [14,15,16,17,18]. Hence, this narrative review seeks to [1] summarize the evidence for the effects of common ICU circumstances (including ECMO, ARC, RRT, and obesity) on the PK of NBLA, and [2] evaluate whether the utilized dosing regimens were adequate in achieving the required PK/PD indices.

2. Methods

PubMed, MEDLINE, Scopus, Google Scholar, and Embase databases were searched for studies published between January 2011 and May 2022 (to focus on NBLA approved during the last decade). Searched keywords included C/T, CAZ/AVI, cefiderocol, ceftobiprole, IMI/REL, and MEV. The following terms were used in combination with the keywords listed above: “obesity”, “augmented renal clearance”, “renal replacement therapy”, “extracorporeal membrane oxygenation”, “pharmacokinetic”, “pharmacodynamic”, “critically ill”, and “intensive care”. The search was limited to adult humans and articles published in English. Two authors (A. B. and R. E.) independently performed the systematic search. Disagreements were resolved by referring to a third author (D. B.). The inclusion of articles in this review was guided by the Population, Intervention, Comparator, Outcome (PICO) framework outlined in detail in Table 1. No additional analyses of the risk of bias were performed since the intention was purely descriptive narration.

3. Results

The initial search yielded 637 articles. After removing duplicates and applying limitations stated above, 64 full-text articles were obtained. Of these, 27 fulfilled the PICO outlined in Table 1 and thus were included in this review. A detailed overview of the selection process is illustrated in Figure 1.

3.1. Altered PK and Associated PD Targets

Physiologic alterations encountered in critically ill populations have been implicated in influencing drugs’ PK, specifically the two primary PK parameters: volume of distribution (Vd) and clearance (CL).
Endothelial dysfunction, capillary leakage, and fluid resuscitation, along with other factors result in increased Vd of hydrophilic antimicrobials. Clearance, on the other hand, is often determined by the properties of the drugs, with hydrophilic antimicrobials being principally cleared through the renal pathway. Both extremes of renal clearance (i.e., ARC/renal failure) are encountered in ICU and can thus have profound effects on the elimination of hydrophilic renally-eliminated antimicrobials. Hypoalbuminemia, as an acute-phase reactant, is commonly seen in critically ill patients and can potentially further impact the PK of antimicrobials, especially the highly protein-bounded. The increased free/unbound concentrations results in increased Vd and can be coupled with decreased overall exposure of the renally-cleared antimicrobials, as more unbound fraction is gaining access to the nephrons for elimination [19,20,21,22]. A detailed description of different sources of PK variabilities encountered during critical-illness is beyond the scope of this paper, and readers are referred to the literature for further details [23,24,25].
Given their physicochemical and PK properties, namely hydrophilicity and renal CL, NBLA are therefore considered susceptible to exposure discrepancies when used among critically ill patients [26,27,28].
Pharmacodynamically, like conventional β-lactams, the %ƒT > MIC has been described as the optimal PK/PD index associated with the efficacy of NBLA. However, the optimal percentage to target is still controversial [29]. Preclinical studies of conventional β-lactams suggested that approximately 1–2 log10 reductions of colony-forming units (CFU) can be achieved with <100%ƒT > MIC, depending on the used antimicrobial (i.e., 40%, 50–60%, and 50–70%ƒT > MIC for carbapenems, penicillins, and cephalosporins respectively) [30]. Although these animal/in-vitro derived targets were replicated in human studies, promising outcomes were not always consistent and some suggested higher than 40–70%ƒT > MIC might sometimes be required for favorable outcomes, especially among severely sick patients [30,31]. For instance, the Defining Antibiotic Levels in ICU(DALI) trial described significant exposure variabilities of conventional β-lactams when used in ICU patients, with one-fifth of the included cohort failing to achieve the most conservative PD index of 50%ƒT > MIC. On the contrary, higher odds of favorable clinical outcomes were seen amongst patients achieving 50%ƒT > MIC and 100%ƒT > MIC targets (OR 1.02, 1.56 respectively; p < 0.03) [32]. Likewise, McKinnon et al., compared the PD of ceftazidime and cefepime and observed significantly higher rates of bacteriological eradication and clinical cure among patients achieving 100%ƒT > MIC as opposed to <100%ƒT > MIC [33]. Similar conclusion was recently reported with meropenem and piperacillin-tazobactam, with faster infection resolution being observed among patients attaining 100%ƒT > MIC target [34].
Although the utilization of conventional PD thresholds (i.e., 40–60%ƒT > MIC) could be argued for NBLA, especially when considering the concentrations of BLIs, for the most part, BLIs by their own are inactive against invading pathogens [30]. The PD targets of such inhibitors (i.e., threshold of concentration (CT) for avibactam/tazobactam and 24 h area under the free concentration–time curve (fAUC24/MIC) for vaborbactam/relebactam) reflect the required concentrations to restore β-lactam activity, thus leaving PD targets of β-lactam backbone consistent with the PD targets described above for the severely sick population [29,35].
More aggressive indices (i.e., 100%ƒT > 4–8 × MIC) were recently advocated among critically ill patients (especially empirically upon initiation when the pathogen/MIC are still unknown) to improve the likelihood of favorable outcomes and prevent resistance development, a detrimental consequence frequently encountered in the ICU population [34,36,37,38,39,40,41,42]. However, given the limited available data, coupled with safety concerns of increased adverse outcomes/toxicities associated with such higher trough concentrations, this target might be more appropriately tailored to individualized situations only [34,36,39,43,44,45].
Hence, in this review, we stated the PK/PD indices that were targeted by the authors of each included study, and then compared the adequacy of the used regimen in attaining the aggressive target of 100%ƒT > MIC.
The leaflet-derived PK parameters along with conventional murine/in vitro-derived PD targets of NBLA included in this review are summarized in Supplementary Materials (Table S1) for reference. Table 2 summarizes the included studies in this review, along with their utilized dosage regimens and attained PD indices.

3.2. Obesity

Antimicrobial dosing for critically ill obese patients is challenging. Different pathophysiologic alterations seen in obesity can result in additive effects of altered PK including increased cardiac output, lean/fat masses, kidney size, and renal blood flow [39,46]. For hydrophilic antimicrobials, increased lean mass, coupled with increased renal CL translate to more PK variations. Different PK studies of conventional β-lactams confirmed altered PK in obese/morbidly-obese patients compared to non-obese population [44,47,48]. However, whether the effect of altered PK is significant enough to warrant dosage adjustment is still debatable [48,49].
Cojutti and colleagues [50] described the effect of obesity on PK/PD of ceftobiprole when used as an add-on to daptomycin against methicillin-resistant Staphylococcus epidermidis bacteremia in a morbidly obese critically ill patient (BMI 51.2 kg/m2). The utilization of standard dosing (0.5 g Q8h) as extended infusion (E.I) over 4 h (vs. recommended 2 h) resulted in 100%ƒT > MIC≤2 mg/L attainment. However, more frequent administrations (Q6h) coupled with E.I were required to attain a more aggressive index (100%ƒT > 3 × MIC) and resulted in a favorable clinical outcome. Unfortunately, only maximum and minimum concentrations (Cmax, Cmin respectively) were reported in this case report, and thus the effects of obesity on other PK parameters could not be determined. Similarly, C/T use at the manufacturer’s recommended regimen resulted in an adequate exposure (100%ƒT > MIC) when used to treat ventilator-associated pneumonia in a morbidly obese patient, with higher targets (100%ƒT > 4 × MIC) being achieved using the continuous infusion (C.I) technique [51]. In both cases, the dose required to attain the conventional PD index (i.e., 30–50% and 40%ƒT > MIC for ceftobiprole and C/T respectively) was not investigated. No studies were found describing the effect of obesity on PK of CAZ/AVI, cefiderocol, IMI/REL, and MEV in critically ill patients.
In summary, limited data exist to guide dosing of NBLA in critically ill morbidly/obese patients, and interpretation of antimicrobial exposure is thus difficult, given the variabilities that exist in such populations. More research is still required, and therapeutic drug monitoring (TDM) might be warranted to guide therapy across such groups.

3.3. Extra-Corporeal Membrane Oxygenation (ECMO)

ECMO has been believed to have various effects on PK of antimicrobials, including altered protein binding, increased Vd (secondary to fluid boluses, transfusion requirements, drug sequestration into ECMO circuits, etc.) and altered CL, with lipophilic highly protein-bound drugs being mostly affected [7,52]. These observations were mainly extrapolated from old neonatal PK studies [53]. Nevertheless, despite the hydrophilicity and limited protein-binding of NBLA, the additive effects of critical-illness and ECMO might correlate with altered PK, and hence result in exposure variabilities.
The effects of ECMO on NBLA’s PK/PD were documented in four case reports/series: two with C/T [54,55] and two for cefiderocol [56,57].
Arena et al. utilized C/T in treating persistent PsA pneumonia in a patient requiring venoarterial ECMO (VA-ECMO) [54]. The standard dosage (3 g Q8h) was considered sufficient for both the conventional target and the aggressive target of 100%ƒT > MIC. Notably, the authors observed lower Cmax and Cmin during the last 2 days of therapy and attributed it to increased Vd (positive fluid balance) and/or enhanced renal CL.
In the other case, C/T was used as a part of chemoprophylaxis in a cystic fibrosis patient requiring ECMO post-lung transplantation [55]. RRT via continuous venovenous hemodiafiltration (CVVHDF) was also required for this patient, owing to acute kidney injury (AKI). When administered as an unadjusted regimen (i.e., regular manufacturer dosing irrespective of AKI/RRT modality), 100%ƒT > 4 × MIC was attained for ceftolozane. It was concluded that ECMO did not require dosing modifications; rather, given the incidence of AKI, reduced dosing might have been adequate.
Comparable with C/T, minimal effects on the PK of cefiderocol among two ECMO case-series [56,57] with adequate concentrations (i.e., 100%ƒT > MIC) were achieved using the regular manufacturer’s regimen.
In summary, based on the available human data, ECMO seemed to have minimal effects on NBLA. However, given the scarcity of reports, the use of CVVHDF in one patient, and keeping in mind that most reports were utilizing VA-ECMO [which might result in different physiological effects as opposed to venovenous ECMO (VV-ECMO)], more data are still required to confirm the observation. Additionally, no studies were found reporting the effect of ECMO on the PK of CAZ/AVI, ceftobiprole, IMI/REL, or MEV.

3.4. Augmented Renal Clearance (ARC)

ARC is a phenomenon frequently encountered across the ICU population secondary to fluid resuscitation, coupled with enhanced cardiac output, leading to amplified renal perfusion. ARC (defined as creatinine clearance (CrCL) > 130 mL/min/1.73 m2) has been linked to lower plasma drug concentrations of renally cleared antimicrobials and worse clinical outcomes [58,59,60]. Given the hydrophilicity and predominant renal clearance of NBLA, ARC may increase the risk of therapeutic failure and/or drug resistance. A total of six studies were retrieved describing the effects of ARC on PK of C/T [61,62], CAZ/AVI [26], IMI/REL [63] and ceftobiprole [50,64].
Sime et al. analyzed the PK of C/T using data from 12 ICU patients receiving either 1.5 g or 3 g Q8h regimens and performed Monte Carlo simulations to predict optimal regimens for critically ill patients with preserved kidney functionality, including those with ARC [61]. When utilized empirically (i.e., aiming for 40%ƒT > MIC≤64 mg/L to cover PsA), a 1.5 g-regimen was found inadequate among the ARC cohort, and a 3 g regimen was required. Nevertheless, when a more aggressive index was targeted (i.e., 100%ƒT > MIC≤64 mg/L), the 3 g intermittent regimen was not satisfactory, and 1.5 g loading over 30 min followed by 4.5 g C.I (over 24 h) was suggested instead. In contrast, directed therapy against MIC≤4 mg/L was adequately achieved with 1.5 g and 3 g intermittent regimens when aiming for 40% and 100%ƒT > MIC≤4 mg/L respectively. Remarkably, the suggested empiric regimens were selected, aiming for 100%ƒT > 4 × MIC≤16 mg/L, which is not routinely targeted in clinical practice nor representative given the sensitivity breakpoint of ceftolozane (≤4, 2 mg/L for PsA and Enterobacterales, respectively [65]).
In early 2021, the results of a phase I PK study of C/T among critically ill patients with confirmed ARC (using the 8 h urine collection method) were published [62]. The mean Vd was 1.5-fold higher in critically ill patients with ARC with a resultant lower Cmax than that of retrospective healthy cohorts. A single dose of 3 g was considered sufficient to achieve 40%ƒT > MIC≤4 mg/L and almost half of the patients were able to attain 100%ƒT > MIC≤4 mg/L for ceftolozane with the same dose. It is noteworthy that, this was a single-dose PK assessment and thus might not reflect effects of accumulation/repeated administrations.
The PK of ceftobiprole use in ARC was investigated in a multicenter, open-label, and non-randomized trial [64]. The systemic CL of ceftobiprole in patients with CrCL > 150 mL/min was two-fold higher than that in patients with normal/slightly decreased CrCL. As part of the study protocol, patients received 1 g Q8h as E.I over 4 h (as opposed to 0.5 g over 2 h, according to the manufacturer’s dosing). This resulted in attaining 100%ƒT > MIC≤4 mg/L. When extrapolated to the manufacturer’s recommended dosing of 0.5 g, only E.I over 4 h allowed the attainment of 100%ƒT > MIC [66]. Likewise, administration of 0.5 g by Cojutti et al. [50] was sufficient to attain 100%ƒT > MIC≤2 mg/L when E.I over 4 h was utilized.
Serum levels of CAZ/AVI following administration of the recommended dose (2.5 g Q8h over 2 h)were used in a phase 4 study to simulate effects of CrCL on CAZ/AVI’s PK among critically ill patients [26]. Despite a two-fold increase in Vd of CAZ compared to healthy volunteers, the probability of target attainment(PTA) was successfully increased in patients with ARC when aiming for 50%ƒT > MIC≤16 mg/Lof CAZ. Higher targets (i.e., 100%ƒT > MIC≤16 mg/L) were not investigated, and thus the adequacy of CAZ/AVI regular dosing among ARC patients when aiming for aggressive targets is yet to be defined.
The effects of ARC on IMI/REL’s PK were recently presented by Fratoni and colleagues [63]. Compared to healthy volunteers, the researchers observed an increased CL of IMI/REL. However, regular dosing (1.25 g over 30 min) was found adequate in providing IMI exposures > 40%ƒT > MIC (range: 40–90%). It is worth noting that this was a single-dose PK study, thus repeated administrations’ effects were not investigated, nor was an aggressive target (100%ƒT > MIC) achieved by any of the included cohorts.
Cefiderocol is the only NBLA that has a manufacturer recommendation for ARC. Such a recommendation was initially based on simulations using healthy volunteers’ concentrations, as opposed to critically ill patients [67,68]. However, using plasma concentrations of patients with ARC including patients from APEKS-NP (with 70% of the cefiderocol group being in ICU at time of randomization) and CREDIBLE-CR (with more than 50% of the cefiderocol group being in ICU at time of randomization), Kawaguchi and colleagues reported that adequacy of such a regimen (i.e., 2g Q6h each administered as E.I over 3 h), with its ability to attain 100%fT > MIC for MIC ≤ 4 mg/L [69,70,71]. Similarly, although MEVs’ PKs were not described for an ARC setting, extrapolation from meropenem use among ICU patients with ARC might be considered. Different reports have highlighted reduced meropenem concentrations in such settings, coupled with required regimens’ modifications to attain a PD target [60,72,73,74]. Moreover, keeping in mind the predominant renal CL of vaborbactam, studies are therefore required to characterize ARC effects on MEV in terms of not only altered PK/required dosage adjustments, but also retained efficacy.
In summary, ARC appeared to result in a disturbed PK of NBLA. Modified dosing regimens (including higher dosing, extended/or C.I) coupled with TDM might be warranted, especially when aggressive targets (i.e., 100%fT > MIC or higher) are required.

3.5. Renal Replacement Therapy

AKI is commonly encountered in ICU patients, resulting in the accumulation of renally cleared drugs, including NBLA, and can result in potential toxicities. Nevertheless, many of these patients require extracorporeal renal support (i.e., RRT) in form of either prolonged intermittent renal replacement therapy (PIRRT) or continuous renal replacement therapy (CRRT), based on the clinical scenario [75]. The differences in the used modalities (e.g., effluent flow rate, filter type, renal replacement method, etc.), patient’s residual kidney functionality, and the disturbed PK owing to critical illness may render the dosing of NBLA in such settings challenging [14,15].

3.5.1. Prolonged Intermittent Renal Replacement Therapy (PIRRT)

A case report described effects of PIRRT on C/T’s PK when used in a critically ill patient with MDR PsA (MIC: 4 mg/L) [76]. The patient received a loading dose of 0.75 g C/T (over 1.5 h), followed by 0.15 g Q8 h and 0.75 g Q12 h on non-PIRRT and PIRRT days respectively. Even though higher amounts of ceftolozane and tazobactam were removed during PIRRT than non-PIRRT days (>20× difference in overall CL), the administration of the aforementioned regimen during and immediately after PIRRT replenished the lost amount, and maintained concentrations above MIC for the entire therapy duration (both 40%, 100%ƒT > MIC for ceftolozane).
The effects of PIRRT on CAZ/AVI, ceftobiprole, cefiderocol, IMI/REL, and MEV when used among ICU patients were not reported.
In summary, little data exist regarding the effects of PIRRT on the PK of NBLA. Until more data are available, therapies might better be guided by TDM to ensure adequacy.

3.5.2. Continuous Renal Replacement Therapy (CRRT)

Gatti and Pea have recently described the effects of various CRRT modalities on PK of NBLA [14]. Relevant studies provided in reviews by Gatti and Pea [14,15], along with several other studies, are highlighted in Table 2. In summary, NBLA’s PK is influenced by CRRT type (continuous venovenous hemofiltration (CVVH), continuous venovenous hemodialysis (CVVHD), and CVVHDF), flow rates, and dilutional fluids. The PK is further affected by residual kidney functionality, which can impact its ability to attain the desired PD index. The results from different reported studies (highlighted in Table 2) suggest that higher doses and/or longer infusions might be necessary in cases of residual kidney function (as compared to anuric states), or when higher PK/PD indices are targeted, especially with highly-resistant pathogens. For a more detailed discussion on the effects of CRRT, readers are advised to refer to Gatti and Pea’s review [14,15].
Based on the limited available evidence, Table 3 briefly provides our suggested initial regimens of different NBLA based on PK/PD targets (i.e., in-vitro derived vs. 100%ƒT > MIC targets) when used among critically ill patients with special scenarios. However, given that most of the reported data were based on case reports/series, those recommendations might be considered as initial dosing regimens. TDM might still be warranted especially among unstudied scenarios or when other PK/PD indices are targeted (e.g., 100%fT > 4–8 × MIC).
Table 2. Novel β-lactam antibiotics utilized dosing regimens and PK/PD target attained of in the included studies.
Table 2. Novel β-lactam antibiotics utilized dosing regimens and PK/PD target attained of in the included studies.
Reference Study
Design
(# of Patients)
Source of
Infection
Pathogen/
MIC (mg/L)
PK/PD Target Aimed by InvestigatorsDose AdministeredPatient(s) Creatinine Clearance (mL/min)/Urine Output (mL/Day)Studied Scenario(s)PK/PD Target Achieved with Given Regimen Clinical Outcome
ARCRRTECMOObesity
Ceftolozane/Tazobactam
Kuti et al. [77]Case
Report (1)
VAPP. aeruginosa
MIC: 0.75/4
100%ƒT > MIC3 g Q8h (over 1 h)<80 mL/day CVVHDF 100%ƒT > MICClinical cure
Bremmer
et al. [78]
Case
Report (1)
BSI, VAP, OsteomyelitisP. aeruginosa
MIC: 2
100%ƒT > MIC3 g Q8h (over 1 h)<50 mL/day CVVHDF 100%ƒT > MIC≤8 mg/LClinical cure *
Carbonell
et al. [79]
Case
Report (1)
CRBSI P. aeruginosa
MIC: NR
100%ƒT > 4 × MIC≤4 mg/L3 g Q8h (over 3 h)NR CVVHDF (+oXiris filter)
+ MARS
100%ƒT > 4 × MIC≤4 mg/LClinical failure
Aguilar et al. [80]Case
Report (1)
cIAI NR100%ƒT > MIC≤8 mg/L3 g Q8h (over 1 h)0 mL/day CVVHD 100%ƒT > MIC≤8 mg/LClinical cure
Oliver et al. [81]Case
Report (1)
OsteomyelitisP. aeruginosa
MIC: 1.5
100%ƒT > MIC1.5 g Q8h (over 4 h)NR CVVH 100%ƒT > 8 × MIC≤4 mg/LClinical cure
Mahmoud et al. [51]Case
Report (1)
VAPP. aeruginosa
MIC: 2/4
100%ƒT > MIC
100%ƒT > 4 × MIC≤2 mg/L
3 g Q8h (over 1 h); then changed to 9 g/24 h (as C.I)0 mL/day CVVHDF BMI 54.5  kg/m2100%ƒT > MIC and
100%ƒT > 4 × MIC≤2 mg/L
NR
Sime et al. [82]PK population
Study (6)
Unknown (n = 1)
Lung (n = 1)
BSI (n = 2)
BSI + Lung (n = 2)
Polymicrobial including
P. aeruginosa,
S. maltophilia,
S. marcescens,
K. pneumoniae and others; MIC: NR
Ceftolozane: 40%ƒT > MIC≤4 mg/L
Simulations done for 60%, 100%ƒT > MIC
Tazobactam:
Simulations done for 20%ƒT > CT:1 mg/L 50%ƒT > CT:2 mg/L100%ƒT > CT:4 mg/L
1.5 g Q8h (over 1 h)NR CVVHDF Empiric therapy (first 24 hr; covering EUCASTP. aeruginosa sensitivity breakpoint of 4 mg/L):
- 40%ƒT > MIC: 0.75 g Q8h
- 100%ƒT > MIC
1.5 g Q8h (over 1 h) or 3 g Q8h (over 1 h) or 3 g LD then 0.75 g Q8h (over 1 h)
- 100%ƒT > MIC (assuming empirically coverage of up to MIC≤64 mg/L): 3 g LD plus 9 g/24 h C.I

Targeted therapy (after 24 h and known MIC≤4 mg/L):
- 40%ƒT > MIC: 0.75 g q8h (over 1 h) (lower dosing can be theoretically possible)
- 100%ƒT > MIC: 0.75g Q8 (over 1 h)
(Note: 0.75 g Q8h achieved adequate Tazobactam targets of 20%ƒT > CT:1 mg/Land 50%ƒT > CT:2 mg/L
NA
Rawlins et al. [76]Case
Report (1)
Osteomyelitis P. aeruginosa
MIC: 4
42%ƒT > MICOff dialysis days: LD 0.75 g (over 1.5 h), then 0.15 g Q8h
Dialysis days: 0.75 g Q12h
NR PIRRT 100%ƒT > MICNR
Sime et al. [61]PK population
Study (12)
BSI (n = 2), CNS abscess (n = 3), CIAI (n = 3), UTI (n = 1), Pneumonia (n = 9), Vascular access (n = 1)Multiple organisms
MIC: NR
Ceftolozane:
Simulations done for 40%, 60%, 100%ƒT > MIC
Tazobactam:
Simulations done for 20%ƒT > CT:1 mg/L
1.5 g Q8h (over 1 h) and 3 g Q8h (over 1 h)Median: 107 mL/min/1.73 m2
IQR: 74–145 mL/min/1.73 m2
Simulated for Crcl > 140 and >180 mL/min/1.73 m2 Empiric therapy (MIC unknown; covering emprically for P. aeruginosa with MIC up to 64):
- 40%ƒT > MIC≤64 mg/L: 3 g Q8h (over 1 h)
-100%ƒT > MIC64 mg/L:
1.5 g LD then 4.5 g/24 h C.I
Targeted therapy (Known MIC≤4 mg/L):
-40%ƒT > MIC4 mg/L: 1.5-g q8h (over 1 h)
-100%ƒT > MIC4 mg/L: 3 g q8h or 1.5 g LD then 4.5 g/24 h C.I
100%ƒT > 4–5 × MIC4 mg/L: 3 g LD then 9 g/24 h C.I
(Note: all doses achieved adequate Tazobactam targets of 20%ƒT > CT:1 mg/L
NA
Nicolau et al. [62]Phase 1, prospective study (11)NRNRCeftolozane:
39%ƒT > MIC≤4 mg/L
Tazobactam:
20%ƒT > CT:1 mg/L
3 g (single dose; over 1 h)>130 mL/minCrCL > 130 mL/min Ceftolozane:
86%ƒT > MIC≤4 mg/L
(did not achieve 100%ƒT > MIC≤4 mg/L)
Tazobactam:
55%ƒT > CT:1 mg/L
NA
Arena et al. [54]Case
Report (1)
Nosocomial pneumonia P. aeruginosa
MIC: 4
Ceftolozane 60%ƒT > MIC3 g Q8h (over 1 h)Day 1: ~90 mL/min,
Day 4: ~150 mL/min
VA-ECMO Ceftolozane
100%ƒT > MIC and 100%ƒT > 3.9 × MIC
Tazobactam
100%ƒT > CT:1 mg/L
Clinical cure
Argudo et al. [55]Case
Report (1)
Post lung transplant prophylaxis (History of CF with XDR-PA colonization)P. aeruginosa
MIC: 1.5
100%ƒT > MIC3 g Q8h (over 1 h)0 mL/day CVVHDFVA-ECMO 100%ƒT > MIC and 100%ƒT > 25 × MIC≤1.5 mg/LNR
Ceftazidime/Avibactam
Stein et al. [26]Phase 4 PK analysis (10)Pneumonia (n = 9)
Urosepsis (n = 1)
Different Enterobacteriales MIC: NR CAZ: 50%ƒT > MIC
AVI:
50%ƒT > CT:1 mg/L
2.5 g Q8h (over 2 h) Mean: 103 mL/min
(range: 47–190 mL/min)
2 patients had ARC; simulations done for CrCL 130–190 mL/min 50%ƒT > MIC≤16 mg/L (based on Monte Carlo simulations)
(100%ƒT > MIC was not investigated)
NA
Wenzler et al. [83]Case
Report (1)
BSI P. aeruginosa
MIC: 6
CAZ: 100%ƒT > MIC≤6 mg/L
AVI:
100%ƒT > CT:1 mg/L
1.25 g Q8h (over 2 h)NR CVVH CAZ:
100%ƒT > MIC
100%ƒT > 4 × MIC
AVI:
100% fT > CT:7 mg/L
Death
Soukup et al. [84]Case
Report (1)
Pneumonia P. aeruginosa
MIC: 8
CAZ:100%ƒT > 4 × MICfor MIC≤
AVI:
100%ƒT > CT:1 mg/L
2.5 g Q8h (over 2 h)<100 mL/day CVVHDF CAZ:
100%ƒT > 4 × MIC
AVI:
100% fT > CT:1 mg/L
Clinical cure
Kline et al. [85]Case series (6)NRNRCAZ:
100%ƒT > 1 and 4 × MIC≤8 mg/L
AVI:
100%ƒT > CT:1and2.5 mg/L
2.5 g Q8h (Infusion duration NR)NR CVVHDF CAZ:
- 90% of patients achieved 100%ƒT > MIC,
- 55% achieved 100%ƒT > 4MIC
AVI:
- 100% of pts achieved
100%ƒT > CT:1 mg/L
- 80% of pts achieved 100%ƒT > CT:2.5 mg/L
NR
Zhang et al. [86]Case report (1) Pneumonia K. pneumoniaeCAZ:
100%ƒT > 4 × MIC≤8 mg/L
2.5 g Q12h (over 2 h)30 mL/day CVVHD 100%ƒT > 4 × MIC≤8 mg/LDeath *
Cefiderocol
Kobic et al. [87]Case report (1)Pneumonia and BSI P. aeruginosa
MIC: 4
82%ƒT > MIC≤4 mg/L2 g Q8h (over 3 h)0 mL/day CVVHDF >90%ƒT > MIC4–8 mg/L for anuric, residual CrCL 11 and residual CrCL 27 mL/minClinical cure
1.5 g Q12h (over 3 h) extrapolated>82%ƒT > MIC≤4 mg/L for anuric, or residual CrCL 11 and residual CrCL 27 mL/min

97%ƒT > MIC≤8 mg/Lfor anuric,
81%fT > MIC≤8 mg/Lfor residual CrCL 11,
65%fT > MIC≤8 mg/Lfor residual CrCL = 27 mL/min
König et al. [56]Case series (5)Pneumonia and/or BSI P. aeruginosa
A. baumanii
MIC: 0.1250.5
75%ƒT > MIC<2 mg/L2 g Q8h (over 3 h, 4 cases)

1 g Q8h (over 3 h, 1 case)
-Pt 1: 10 mL/min
-Pt 2: Day 1: 67 mL/min, Day 7: 22 mL/min
-Pt 3: >80 mL/min
-Pt 4: 28 mL/min
-Pt 5: NR
CVVHD (3 cases) VA-ECMO (2 cases) - 100%ƒT > MIC≤2 mg/L
- 100%ƒT > 4 × MIC≤2 mg/L
3 patients with microbiological cure, 2 patients died
Fratoni et al. [27]Case report (1)Bacteremia and pneumonia ESBL—Escherichia coli bacteremia
MIC: NR
S. maltophilia pneumonia
MIC: 0.125
100%ƒT > MIC2 g Q12h
(over 3 h)
0 mL/min CVVHDF
Note: Pt had 20% protein binding as opposed to 58% of package insert
100%ƒT > MIC≤16 mg/L for protein binding of 20% and no residual kidney function

>92%ƒT > MIC≤4 mg/L for all scenarios of anuria, residual CrCL of 15–30 mL/min and protein binding of 20–58%
Clinical cure *
Wenzler et al. [88]Population PK (9)nosocomial pneumonia (n = 3) and carbapenem-resistant Gram-negative infection (n = 6) 75%ƒT > MIC0.25–16 mg/LPatient received: 1 g q12h (over 3 h) for CVVH
1.5 g q12h (over 3 h) for CVVHD and CVVHDF
Modeling based on assumption of minimal residual kidney functionality CVVH
CVVHD
CVVHDF
>90% PTA of 75%ƒT > MIC≤4 mg/L at effluent flow rates from 0.5 to 5 L/h
Simulated regimens required (each infused over 3 h):
- 1.5 g every 12 h for effluent flow rate ≤ 2 L/h
- 2 g every 12 h for effluent flow rate 2.1–3 L/h
- 1.5 g every 8 h for effluent flow rate 3.1–4 L/h
- 2 g every 8 h for effluent flow rate ≥4.1 L/h
NR
Gatti et al. [57]Case series (5/13 with special scenario)Pneumonia and/or BSIXDR-Acinetobacter baumannii
MIC: 0.5–1
Optimal if fCmin/MIC ≥ 4
(100%ƒT > 4 × MIC)

Quasi optimal if fCmin/MIC 1–4
(100%ƒT > 1–4 × MIC)
2 g q8h (over 3 h) for 4 pts
[1 case received 1.5 g q8h (over 3 h)]
NR CVVHDF (2 cases) ECMO (4 cases) All patients achieved 100% fT > MIC (fCmin/MIC > 1)

(3/5 achieved target of fCmin/MIC > 4 (i.e., 100%ƒT > 4 × MIC))
3/5 documented microbiological eradication
Ceftobiprole
Torres et al. [64,66]Multi-center, open-labelled non-RCT (31) NRNRƒT > MIC≤4 mg/L(in hours)CrCL > 80 mL/min: 1 g Q8h (over 4 h)>80 mL/minCrCL > 150 mL/min were included
(6 patients)
- CrCL 80–150 mL/min: 13.2 h
- CrCL > 150 mL/min: 10.8 h
(=100%ƒT > MIC≤4 mg/L
NR
Extrapolated dosing of 0.5 g Q8h (over 4 h)
100%ƒT > MIC≤4 mg/L
(PTA: 100% if CrCL 80–150 mL/min vs. ~90% if CrCL > 150 mL/min)
Cojutti et al. [50]Case report (1)BSI Methicillin Resistant Staphylococcus epidermidis (MRSE)
MIC: 2
Cmin/MIC: 1–4 (i.e., 100%fT > 1–4 × MIC)0.5 g Q8h (over 4 h)>120 mL/minCrCL > 120 mL/min BMI
51.2 kg/m2
Cmin/MIC 2.85
(=100%ƒT > 2.4 × MIC≤2 mg/L
Clinical cure
Cmin/MIC: 1–4 (i.e., 100%fT > 1–4 × MIC)
Aiming for higher end of the range)
0.5 g Q6h (over 4 h)Cmin/MIC 3.19
(=100%ƒT > 2.7 × MIC≤2 mg/L)
Cojutti et al. [89]Case report (1)Pneumonia Empirically covering Methicillin-resistant Staphylococcus aureus (MRSA)
MIC: NR
Cmin/MIC: 1–40.25 g Q12h (over 2 h)10 mL/min/1.73 m2 CVVHDF Cmin/MIC 2.12

100%ƒT > 1.8 MIC≤2 mg/L
Clinical cure *
Imipenem/Relebactam
Fratoni et al. [63]Prospective PK analysis (5)NRNR
MIC: 2
- IMI 30%fT > MIC)
- REL (fAUC:MIC 18)
For MIC ≤ 2 mg/L
1.25 g once (over 30 min) > 130 mL/minCrCL > 130 mL/min - IMI 40–90%ƒT > MIC≤2 mg/L
- REL fAUC:MIC ranged 22.6–59.0
Meropenem/Vaborbactam
Kufel et al. [90]Case report (1)Periprosthetic hip joint infectionK. pneumoniae
MIC: 0.094/8
100%ƒT > MIC2 g Q8h (over 3 h) 0 mL/day CVVHD 100%ƒT > MIC (for MIC 4/8 mg/L of Meropenem/vaborbactam respectively) Clinical failure
MIC: minimum inhibitory concentration, ARC: augmented renal clearance, RRT: renal replacement therapy, ECMO: extracorporeal membrane oxygenation, BSI: bloodstream infection, CNS: central nervous system, VAP: ventilator-associated pneumonia, cIAI: complicated intra-abdominal infection, CRBSI: catheter-related bloodstream infection, CVVH: continuous veno-venous hemofiltration, CVVHD: continuous venovenous hemodialysis, CVVHDF: continuous venovenous hemodiafiltration, PIRRT: prolonged intermittent renal replacement therapy, BMI: body mass index, VA-ECMO: venoarterial extracorporeal membrane oxygenation, NR: not reported, NA: not applicable, MARS: molecular adsorbent-recirculating-system, C.I: continuous infusion, LD: loading dose, CF: cystic fibrosis, XDR: extensively drug-resistant, ESBL: Extended Spectrum B-Lactamase, Pt: patient, AUC: area under the concentration time curve, %ƒT > MIC: percentage of free drug concentration above MIC, Cmin: minimal concentration, PK/PD: pharmacokinetic/pharmacodynamic, CrCL: creatinine clearance, Qxh: every x hours, IMI/REL: imipenem/relebactam, CAZ/AVI: ceftazidime/avibactam. * Patient death was considered non-infection related. Although ARC is commonly defined as >130 mL/min/1.73 m2, different studies used different cut-offs ranging from >120 to >160 mL/min/1.73 m2. Thus, this study was referenced as ARC for the sake of completeness.

4. Discussion

This review highlights the scarcity of evidence regarding optimal dose-regimens for NBLA in some of the special scenarios encountered among ICU patients. Variations within the studied settings and PK/PD-used targets across the included studies limited the overall generalizability of the findings. This is in line with earlier assessments that highlighted the lack of adequate information when conventional β-lactams were utilized among similar special scenarios [21,44]. Given the altered physiology/pathology of critically ill patients, standard dose regimens may be insufficient for NBLA and may increase the risk of therapeutic failure and/or resistance development. Such conclusion can be inferred from the pool of evidence pertaining to conventional β-lactams, where different antimicrobials were reported to be associated with altered PK when utilized among critically ill patients, and had suboptimal exposures when utilized at the usual manufacturer recommended dosing. Readers are encouraged to refer to the previous published literature for more in-depth information [17,25,91,92]
Although the “one size fits all” approach has been well-described to be inappropriate across such critically ill population, more studies are still required to properly adopt patient-centered treatment approaches [93]. The importance of achieving PK/PD targets in critically ill patients has recently been emphasized, considering the increasing microbial resistance, limited treatment alternatives, and severity of illnesses [94]. Various factors should be considered when administering such NBLA among critically ill patients with special circumstances, including critical-illness-related PK alterations, the physiochemical properties of the NBLA, the site of infection/MIC of invading pathogen, and the type/modality of extracorporeal support required [14,15]. However, caution should still be practiced when considering these factors. For instance, hydrophilic drugs with limited protein binding were previously reported to be minimally subjected to ECMO-mediated variations [52,95] Thus, one would extrapolate such minimal effects of ECMO to NBLA’s PK given their similar physiochemical properties and support such extrapolation by the reported studies highlighted in the Section 3 above. However, recent ex-vivo ECMO models have shown substantial removal of various NBLAs (e.g., C/T [96] and MEV [97]. Despite the limitations of such ex-vivo models in terms of correlation with clinical settings (i.e., single dosing administrations, no effects of human metabolism/clearance, different oxygenators used, effects of different primed fluids, etc.), they reveal commonly encountered variations seen in practice, and thus highlight the need for more clinical research to clarify such heterogeneity.
Similarly, the effect of obesity on the PK of different antimicrobials has been a topic of debate. Despite being associated with alterations in different physiological parameters, including increased cardiac output and renal blood flow [44,47,48], different studies questioned the clinical significance in terms of required dosage adjustments. For instance, in a population PK study conducted by Alobaid and colleagues [48], obesity was associated with an altered central volume of the distribution of meropenem, yet such alteration did not translate into an altered probability of attaining PD targets. Rather, in their analysis, CrCL was found to be the significant covariate affecting PTA. Similar conclusions were drawn for other conventional β-lactams, too [49,98]. This might be in line with what we found in our review. Among the two reports of morbid obesity included in this review, both patients had altered renal functionalities (ARC in the case of ceftobiprole [50], and anuric requiring CVVHDF in the case of C/T [51]). In both cases, modifications targeted toward the altered renal functionality resulted in adequate exposures of the studied NBLA, without the requirement of further adjustments targeted towards BMI.
Renal functionality has been well documented to derive major changes in the altered PK of different antimicrobials, including β-lactams. ARC has been suggested to be associated with subtherapeutic plasma concentrations of conventional β-lactams, along with increased rates of therapeutic failure when utilized at the standard dosing regimens [59,60,99,100]. Such studies have proposed increased dosing requirements and/or modified dosing administrations (i.e., prolonged infusions) to ensure attaining therapeutic targets. Comparing the results of conventional β-lactams’ modified dosing requirements in the setting of ARC to the studies of NBLA included in this review provide similar conclusions. Based on the limited available evidence from the included studies, modified administrations seemed to be required to achieve required PTA of different NBLAs. On the other extreme, Gatti and Pea [14,15] suggested four main factors to be considered when designing an appropriate regimen in the setting of CRRT. These included PK features/physiochemical properties of the utilized NBLA, RRT modality/setting/filter type, infection site/associated MIC of the pathogen, and critical illness related altered PK. Similar concepts are still applicable to the setting of PIRRT.
Considering the aforementioned variabilities associated with the four included special scenarios, TDM-based dosing coupled with modified dosing administrations (i.e., increased dosing and/or prolonged infusions) might still be warranted. Although a recent review questioned the role of comprehensive β-lactam TDM among critically ill patients, authors acknowledged the role of targeted TDM among special scenarios(including ARC, RRT, ECMO and obesity), where the benefits of preventing over or underexposure would be expected [101]. Nevertheless, keeping in mind that TDM is still not widely available across many centers worldwide, and until more robust data are available, one might consider utilizing actual creatinine clearance (e.g., using 12 h urine collection) to design an initial dosing regimen, especially in the settings of obesity and ECMO, where CrCL seemed to be the main determinant of PTA of NBLAs based on the available evidence included in this review.

Limitations

The findings of this review should be interpreted with caution. Although this is not a systematic review, we sought to include most of the relevant studies reported. Furthermore, most of the retrieved studies included small patient populations and were of low quality (i.e., case reports/series). Finally, remarkable heterogeneity was detected in the targeted %ƒT > MIC across different reports/studies (highlighted in Table 2). Therefore, extrapolating conclusions should be thoughtfully considered when different PK/PD indices are clinically targeted.

5. Conclusions

Alterations in PK are often encountered in critically ill adult patients, and can result in suboptimal %ƒT > MIC attainments and potential therapeutic failures. This can be further complicated by many of the commonly encountered circumstances during critical illness, including obesity, ECMO, and extreme renal functionalities (ARC/RRT). The available literature, although confined, highlights the limitations of current dosing strategies in such settings. Different studies and reports have suggested modified dosing approaches (such as increased dosing and/or prolonged infusions) to optimize NBLA dosing across ICU-encountered scenarios, yet evidence has been limited by small sample sizes and the low quality of the studies. More robust, well-designed, studies are still required to determine the optimal dosing strategies of NBLA for such patient populations, and thus aid in improving clinical outcomes. Until more robust data are available, the use of TDM two guide therapy in such specialized scenarios might still be warranted.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/jcm11236898/s1, Table S1: Pharmacokinetics and murine/in vitro-derived Pharmacodynamic targets of novel β-lactams.

Author Contributions

D.B.: Conceptualization, Methodology, Investigation, formal analysis, Writing—Review & Editing. R.E.: Conceptualization, Methodology, Investigation, formal analysis, Writing—Review & Editing. A.R.B.: Methodology, Investigation, formal analysis. A.A.: Writing—Original Draft, Supervision. G.D.P.: Validation, Supervision, Writing—Review & Editing. A.A.H.: Conceptualization, Validation, Supervision. 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.

Abbreviations

AKIacute kidney injury
ARCaugmented renal clearance
AUCarea under the concentration time curve
BLBLIβ-lactam/β-lactamase inhibitors
BLIβ-lactamase inhibitors
BMIbody mass index
CAZ/AVIceftazidime/avibactam
CFRcumulative fraction of response
CFUcolony-forming units
C.Icontinuous infusion
CLclearance
Cmaxmaximum concentration
Cminminimal concentration
CrCLcreatinine clearance
CRRTcontinuous renal replacement therapy
CTthreshold of concentration
C/Tceftolozane/tazobactam
CVVHcontinuous venovenous hemofiltration
CVVHDcontinuous venovenous hemodialysis
CVVHDFcontinuous venovenous hemodiafiltration
ECMOextracorporeal membrane oxygenation
E.Iextended infusion
ggram
hhour
ICUsintensive care units
IMI/RELimipenem/relebactam
LDloading dose
MDRmultidrug-resistant
MEVmeropenem/vaborbactam
MICminimum inhibitory concentration
MRSEmethicillin-resistant S. epidermidis
PICOpopulation, intervention, comparator, outcome
PIRRTprolonged intermittent renal replacement therapy
PK/PDpharmacokinetic/pharmacodynamic
PsAPseudomonas aeruginosa
PTAprobability of target attainment
Qxhevery x hours
RRTrenal replacement therapy
TDMtherapeutic drug monitoring
VA-ECMOvenoarterial ECMO
Vdvolume of distribution
VV-ECMOvenovenous ECMO
%ƒT > MICpercentage of free drug concentration above MIC

References

  1. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [Green Version]
  2. Vincent, J.L.; Rello, J.; Marshall, J.; Silva, E.; Anzueto, A.; Martin, C.D.; Moreno, R.; Lipman, J.; Gomersall, C.; Sakr, Y.; et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA 2009, 302, 2323–2329. [Google Scholar] [CrossRef] [Green Version]
  3. Cosgrove, S.E. The relationship between antimicrobial resistance and patient outcomes: Mortality, length of hospital stay, and health care costs. Clin. Infect. Dis. 2006, 42 (Suppl. 2), S82–S89. [Google Scholar] [CrossRef] [Green Version]
  4. Sunenshine, R.H.; Wright, M.O.; Maragakis, L.L.; Harris, A.D.; Song, X.; Hebden, J.; Cosgrove, S.E.; Anderso, A.; Carnell, J.; Jernigan, D.B.; et al. Multidrug-resistant Acinetobacter infection mortality rate and length of hospitalization. Emerg. Infect. Dis. 2007, 13, 97–103. [Google Scholar] [CrossRef] [PubMed]
  5. Póvoa, P.; Moniz, P.; Pereira, J.G.; Coelho, L. Optimizing Antimicrobial Drug Dosing in Critically Ill Patients. Microorganisms 2021, 9, 1401. [Google Scholar] [CrossRef] [PubMed]
  6. Roberts, J.A.; Roberts, M.S.; Semark, A.; Udy, A.A.; Kirkpatrick, C.M.; Paterson, D.L.; Roberts, M.J.; Kruger, P.; Lipman, J. Antibiotic dosing in the “at risk” critically ill patient: Linking pathophysiology with pharmacokinetics/pharmacodynamics in sepsis and trauma patients. BMC Anesthesiol. 2011, 11, 3. [Google Scholar] [CrossRef] [Green Version]
  7. Ha, M.A.; Sieg, A.C. Evaluation of Altered Drug Pharmacokinetics in Critically Ill Adults Receiving Extracorporeal Membrane Oxygenation. Pharmacotherapy 2017, 37, 221–235. [Google Scholar] [CrossRef]
  8. Hoff, B.M.; Maker, J.H.; Dager, W.E.; Heintz, B.H. Antibiotic Dosing for Critically Ill Adult Patients Receiving Intermittent Hemodialysis, Prolonged Intermittent Renal Replacement Therapy, and Continuous Renal Replacement Therapy: An Update. Ann. Pharmacother. 2020, 54, 43–55. [Google Scholar] [CrossRef]
  9. Heintz, B.H.; Matzke, G.R.; Dager, W.E. Antimicrobial dosing concepts and recommendations for critically ill adult patients receiving continuous renal replacement therapy or intermittent hemodialysis. Pharmacotherapy 2009, 29, 562–577. [Google Scholar] [CrossRef] [Green Version]
  10. Kollef, M.H.; Sherman, G.; Ward, S.; Fraser, V.J. Inadequate antimicrobial treatment of infections: A risk factor for hospital mortality among critically ill patients. Chest 1999, 115, 462–474. [Google Scholar] [CrossRef] [PubMed]
  11. Epstein, B.J.; Gums, J.G.; Drlica, K. The changing face of antibiotic prescribing: The mutant selection window. Ann. Pharmacother. 2004, 38, 1675–1682. [Google Scholar] [CrossRef] [PubMed]
  12. Olofsson, S.K.; Cars, O. Optimizing drug exposure to minimize selection of antibiotic resistance. Clin. Infect. Dis. 2007, 45 (Suppl. 2), S129–S136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Han, R.; Sun, D.; Li, S.; Chen, J.; Teng, M.; Yang, B.; Dong, Y.; Wang, T. Pharmacokinetic/Pharmacodynamic Adequacy of Novel β-Lactam/β-Lactamase Inhibitors against Gram-Negative Bacterial in Critically Ill Patients. Antibiotics 2021, 10, 993. [Google Scholar] [CrossRef] [PubMed]
  14. Gatti, M.; Pea, F. Pharmacokinetic/pharmacodynamic target attainment in critically ill renal patients on antimicrobial usage: Focus on novel beta-lactams and beta lactams/beta-lactamase inhibitors. Expert Rev. Clin. Pharmacol. 2021, 14, 583–599. [Google Scholar] [CrossRef] [PubMed]
  15. Gatti, M.; Pea, F. Antimicrobial Dose Reduction in Continuous Renal Replacement Therapy: Myth or Real Need? A Practical Approach for Guiding Dose Optimization of Novel Antibiotics. Clin. Pharmacokinet. 2021, 60, 1271–1289. [Google Scholar] [CrossRef]
  16. Karaiskos, I.; Galani, I.; Souli, M.; Giamarellou, H. Novel β-lactam-β-lactamase inhibitor combinations: Expectations for the treatment of carbapenem-resistant Gram-negative pathogens. Expert Opin. Drug Metab. Toxicol. 2019, 15, 133–149. [Google Scholar] [CrossRef]
  17. Maguigan, K.L.; Al-Shaer, M.H.; Peloquin, C.A. Beta-Lactams Dosing in Critically Ill Patients with Gram-Negative Bacterial Infections: A PK/PD Approach. Antibiotics 2021, 10, 1154. [Google Scholar] [CrossRef]
  18. Gorham, J.; Taccone, F.S.; Hites, M. Drug Regimens of Novel Antibiotics in Critically Ill Patients with Varying Renal Functions: A Rapid Review. Antibiotics 2022, 11, 546. [Google Scholar] [CrossRef]
  19. Shah, S.; Barton, G.; Fischer, A. Pharmacokinetic considerations and dosing strategies of antibiotics in the critically ill patient. J. Intensive Care Soc. 2015, 16, 147–153. [Google Scholar] [CrossRef] [Green Version]
  20. Sime, F.B.; Roberts, M.S.; Peake, S.L.; Lipman, J.; Roberts, J.A. Does Beta-lactam Pharmacokinetic Variability in Critically Ill Patients Justify Therapeutic Drug Monitoring? A Systematic Review. Ann. Intensive Care 2012, 2, 35. [Google Scholar] [CrossRef]
  21. Masich, A.M.; Heavner, M.S.; Gonzales, J.P.; Claeys, K.C. Pharmacokinetic/Pharmacodynamic Considerations of Beta-Lactam Antibiotics in Adult Critically Ill Patients. Curr. Infect. Dis. Rep. 2018, 20, 9. [Google Scholar] [CrossRef] [PubMed]
  22. Ulldemolins, M.; Roberts, J.A.; Rello, J.; Paterson, D.L.; Lipman, J. The effects of hypoalbuminaemia on optimizing antibacterial dosing in critically ill patients. Clin. Pharmacokinet. 2011, 50, 99–110. [Google Scholar] [CrossRef] [PubMed]
  23. Heffernan, A.J.; Mohd Sazlly Lim, S.; Lipman, J.; Roberts, J.A. A personalised approach to antibiotic pharmacokinetics and pharmacodynamics in critically ill patients. Anaesth. Crit. Care Pain Med. 2021, 40, 100970. [Google Scholar] [CrossRef] [PubMed]
  24. Roberts, J.A.; Abdul-Aziz, M.H.; Lipman, J.; Mouton, J.W.; Vinks, A.A.; Felton, T.W.; Hope, W.W.; Farkas, A.; Neely, M.N.; Schentag, J.J.; et al. Individualised antibiotic dosing for patients who are critically ill: Challenges and potential solutions. Lancet Infect. Dis. 2014, 14, 498–509. [Google Scholar] [CrossRef] [Green Version]
  25. Gonçalves-Pereira, J.; Póvoa, P. Antibiotics in critically ill patients: A systematic review of the pharmacokinetics of β-lactams. Crit. Care 2011, 15, R206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Stein, G.E.; Smith, C.L.; Scharmen, A.; Kidd, J.M.; Cooper, C.; Kuti, J.; Mitra, S.; Nicolau, D.P.; Havlichek, D.H. Pharmacokinetic and Pharmacodynamic Analysis of Ceftazidime/Avibactam in Critically Ill Patients. Surg. Infect. 2019, 20, 55–61. [Google Scholar] [CrossRef]
  27. Fratoni, A.J.; Kuti, J.L.; Nicolau, D.P. Optimised cefiderocol exposures in a successfully treated critically ill patient with polymicrobial Stenotrophomonas maltophilia bacteraemia and pneumonia receiving continuous venovenous haemodiafiltration. Int. J. Antimicrob.Agents 2021, 58, 106395. [Google Scholar] [CrossRef]
  28. Caro, L.; Nicolau, D.P.; De Waele, J.J.; Kuti, J.L.; Larson, K.B.; Gadzicki, E.; Yu, B.; Zeng, Z.; Adedoyin, A.; Rhee, E.G. Lung penetration, bronchopulmonary pharmacokinetic/pharmacodynamic profile and safety of 3 g of ceftolozane/tazobactam administered to ventilated, critically ill patients with pneumonia. J. Antimicrob. Chemother. 2020, 75, 1546–1553. [Google Scholar] [CrossRef]
  29. Berry, A.V.; Kuti, J.L. Pharmacodynamic Thresholds for Beta-Lactam Antibiotics: A Story of Mouse Versus Man. Front. Pharmacol. 2022, 13, 833189. [Google Scholar] [CrossRef]
  30. Fratoni, A.J.; Nicolau, D.P.; Kuti, J.L. A guide to therapeutic drug monitoring of β-lactam antibiotics. Pharmacotherapy 2021, 41, 220–233. [Google Scholar] [CrossRef]
  31. Barreto, E.F.; Webb, A.J.; Pais, G.M.; Rule, A.D.; Jannetto, P.J.; Scheetz, M.H. Setting the Beta-Lactam Therapeutic Range for Critically Ill Patients: Is There a Floor or Even a Ceiling? Crit. Care Explor. 2021, 3, e0446. [Google Scholar] [CrossRef] [PubMed]
  32. Roberts, J.A.; Paul, S.K.; Akova, M.; Bassetti, M.; De Waele, J.J.; Dimopoulos, G.; Kaukonen, K.M.; Koulenti, D.; Martin, C.; Montravers, P.; et al. DALI: Defining antibiotic levels in intensive care unit patients: Are current β-lactam antibiotic doses sufficient for critically ill patients? Clin. Infect. Dis. 2014, 58, 1072–1083. [Google Scholar] [CrossRef] [PubMed]
  33. McKinnon, P.S.; Paladino, J.A.; Schentag, J.J. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T > MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int. J. Antimicrob. Agents 2008, 31, 345–351. [Google Scholar] [CrossRef]
  34. Scharf, C.; Liebchen, U.; Paal, M.; Taubert, M.; Vogeser, M.; Irlbeck, M.; Zoller, M.; Schroeder, I. The higher the better? Defining the optimal beta-lactam target for critically ill patients to reach infection resolution and improve outcome. J. Intensive Care 2020, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  35. Crass, R.L.; Pai, M.P. Pharmacokinetics and Pharmacodynamics of β-Lactamase Inhibitors. Pharmacotherapy 2019, 39, 182–195. [Google Scholar] [CrossRef]
  36. Sumi, C.D.; Heffernan, A.J.; Lipman, J.; Roberts, J.A.; Sime, F.B. What Antibiotic Exposures Are Required to Suppress the Emergence of Resistance for Gram-Negative Bacteria? A Systematic Review. Clin. Pharmacokinet. 2019, 58, 1407–1443. [Google Scholar] [CrossRef]
  37. Abdul-Aziz, M.; Lipman, J.; Mouton, J.; Hope, W.; Roberts, J. Applying Pharmacokinetic/Pharmacodynamic Principles in Critically Ill Patients: Optimizing Efficacy and Reducing Resistance Development. Semin. Respir. Crit. Care Med. 2015, 36, 136–153. [Google Scholar] [CrossRef] [Green Version]
  38. Tam, V.H.; Schilling, A.N.; Neshat, S.; Poole, K.; Melnick, D.A.; Coyle, E.A. Optimization of meropenem minimum concentration/MIC ratio to suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2005, 49, 4920–4927. [Google Scholar] [CrossRef] [Green Version]
  39. Tam, V.H.; McKinnon, P.S.; Akins, R.L.; Rybak, M.J.; Drusano, G.L. Pharmacodynamics of cefepime in patients with Gram-negative infections. J. Antimicrob. Chemother. 2002, 50, 425–428. [Google Scholar] [CrossRef] [Green Version]
  40. Mouton, J.W.; Vinks, A.A. Pharmacokinetic/Pharmacodynamic Modelling of Antibacterials In Vitro and In Vivo Using Bacterial Growth and Kill Kinetics: The Minimum Inhibitory Concentration versus Stationary Concentration. Clin. Pharmacokinet. 2005, 44, 201–210. [Google Scholar] [CrossRef]
  41. Delattre, I.K.; Taccone, F.S.; Jacobs, F.; Hites, M.; Dugernier, T.; Spapen, H.; Laterre, P.F.; Wallemacq, P.E.; Van Bambeke, F.; Tulkens, P.M. Optimizing β-lactams treatment in critically-ill patients using pharmacokinetics/pharmacodynamics targets: Are first conventional doses effective? Expert Rev. Anti. Infect. Ther. 2017, 15, 677–688. [Google Scholar] [CrossRef] [PubMed]
  42. Tam, V.H.; Chang, K.T.; Zhou, J.; Ledesma, K.R.; Phe, K.; Gao, S.; Van Bambeke, F.; Sánchez-Díaz, A.M.; Zamorano, L.; Oliver, A.; et al. Determining β-lactam exposure threshold to suppress resistance development in Gram-negative bacteria. J. Antimicrob. Chemother. 2017, 72, 1421–1428. [Google Scholar] [CrossRef] [PubMed]
  43. Guilhaumou, R.; Benaboud, S.; Bennis, Y.; Dahyot-Fizelier, C.; Dailly, E.; Gandia, P.; Goutelle, S.; Lefeuvre, S.; Mongardon, N.; Roger, C.; et al. Optimization of the treatment with beta-lactam antibiotics in critically ill patients-guidelines from the French Society of Pharmacology and Therapeutics (Société Française de Pharmacologie et Thérapeutique-SFPT) and the French Society of Anaesthesia and Intensive Care Medicine (Société Française d’Anesthésie et Réanimation-SFAR). Crit. Care 2019, 23, 104. [Google Scholar] [PubMed] [Green Version]
  44. Abdulla, A.; Dijkstra, A.; Hunfeld, N.G.M.; Endeman, H.; Bahmany, S.; Ewoldt, T.M.J.; Muller, A.E.; van Gelder, T.; Gommers, D.; Koch, B.C.P. Failure of target attainment of beta-lactam antibiotics in critically ill patients and associated risk factors: A two-center prospective study (EXPAT). Crit. Care 2020, 24, 558. [Google Scholar] [CrossRef]
  45. Duszynska, W.; Taccone, F.S.; Switala, M.; Hurkacz, M.; Kowalska-Krochmal, B.; Kübler, A. Continuous infusion of piperacillin/tazobactam in ventilator-associated pneumonia: A pilot study on efficacy and costs. Int. J. Antimicrob. Agents 2012, 39, 153–158. [Google Scholar] [CrossRef]
  46. Meng, L.; Mui, E.; Holubar, M.K.; Deresinski, S.C. Comprehensive Guidance for Antibiotic Dosing in Obese Adults. Pharmacotherapy 2017, 37, 1415–1431. [Google Scholar] [CrossRef]
  47. Alobaid, A.S.; Hites, M.; Lipman, J.; Taccone, F.S.; Roberts, J.A. Effect of obesity on the pharmacokinetics of antimicrobials in critically ill patients: A structured review. Int. J. Antimicrob. Agents 2016, 47, 259–268. [Google Scholar] [CrossRef]
  48. Alobaid, A.S.; Wallis, S.C.; Jarrett, P.; Starr, T.; Stuart, J.; Lassig-Smith, M.; Ordóñez Mejia, J.L.; Roberts, M.S.; Lipman, J.; Roberts, J.A. Effect of Obesity on the Population Pharmacokinetics of Meropenem in Critically Ill Patients. Antimicrob. Agents Chemother. 2016, 60, 4577–4584. [Google Scholar] [CrossRef] [Green Version]
  49. Alobaid, A.S.; Wallis, S.C.; Jarrett, P.; Starr, T.; Stuart, J.; Lassig-Smith, M.; Mejia, J.L.; Roberts, M.S.; Roger, C.; Udy, A.A.; et al. Population Pharmacokinetics of Piperacillin in Nonobese, Obese, and Morbidly Obese Critically Ill Patients. Antimicrob. Agents Chemother. 2017, 61, e01276-16. [Google Scholar] [CrossRef] [Green Version]
  50. Cojutti, P.G.; Carnelutti, A.; Mattelig, S.; Sartor, A.; Pea, F. Real-Time Therapeutic Drug Monitoring-Based Pharmacokinetic/Pharmacodynamic Optimization of Complex Antimicrobial Therapy in a Critically Ill Morbidly Obese Patient. Grand Round/A Case Study. Ther. Drug Monit. 2020, 42, 349–352. [Google Scholar] [CrossRef]
  51. Mahmoud, A.; Shah, A.; Nutley, K.; Nicolau, D.P.; Sutherland, C.; Jain, M.; Scheetz, M.H.; Rhodes, N.J. Clinical pharmacokinetics of ceftolozane and tazobactam in an obese patient receiving continuous venovenous haemodiafiltration: A patient case and literature review. J. Glob. Antimicrob. Resist. 2020, 21, 83–85. [Google Scholar] [CrossRef] [PubMed]
  52. Duceppe, M.A.; Kanji, S.; Do, A.T.; Ruo, N.; Cavayas, Y.A.; Albert, M.; Robert-Halabi, M.; Zavalkoff, S.; Dupont, P.; Samoukovic, G.; et al. Pharmacokinetics of Commonly Used Antimicrobials in Critically Ill Adults During Extracorporeal Membrane Oxygenation: A Systematic Review. Drugs 2021, 81, 1307–1329. [Google Scholar] [CrossRef] [PubMed]
  53. Sherwin, J.; Heath, T.; Watt, K. Pharmacokinetics and Dosing of Anti-infective Drugs in Patients on Extracorporeal Membrane Oxygenation: A Review of the Current Literature. Clin. Ther. 2016, 38, 1976–1994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Arena, F.; Marchetti, L.; Henrici De Angelis, L.; Maglioni, E.; Contorni, M.; Cassetta, M.I.; Novelli, A.; Rossolini, G.M. Ceftolozane-Tazobactam Pharmacokinetics during Extracorporeal Membrane Oxygenation in a Lung Transplant Recipient. Antimicrob. Agents Chemother. 2019, 63, e02131-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Argudo, E.; Riera, J.; Luque, S.; Los-Arcos, I.; López-Meseguer, M.; Sandiumenge, A.; Nuvials, X.; Grau, S.; Ferrer, R. Effects of the extracorporeal membrane oxygenation circuit on plasma levels of ceftolozane. Perfusion 2020, 35, 267–270. [Google Scholar] [CrossRef]
  56. König, C.; Both, A.; Rohde, H.; Kluge, S.; Frey, O.R.; Röhr, A.C.; Wichmann, D. Cefiderocol in Critically Ill Patients with Multi-Drug Resistant Pathogens: Real-Life Data on Pharmacokinetics and Microbiological Surveillance. Antibiotics 2021, 10, 649. [Google Scholar] [CrossRef]
  57. Gatti, M.; Bartoletti, M.; Cojutti, P.G.; Gaibani, P.; Conti, M.; Giannella, M.; Viale, P.; Pea, F. A descriptive case series of pharmacokinetic/pharmacodynamic target attainment and microbiological outcome in critically ill patients with documented severe extensively drug-resistant Acinetobacter baumannii bloodstream infection and/or ventilator-associated pneumonia treated with cefiderocol. J. Glob. Antimicrob. Resist. 2021, 27, 294–298. [Google Scholar]
  58. Bilbao-Meseguer, I.; Rodríguez-Gascón, A.; Barrasa, H.; Isla, A.; Solinís, M.Á. Augmented Renal Clearance in Critically Ill Patients: A Systematic Review. Clin. Pharmacokinet. 2018, 57, 1107–1121. [Google Scholar] [CrossRef]
  59. Claus, B.O.M.; Hoste, E.A.; Colpaert, K.; Robays, H.; Decruyenaere, J.; De Waele, J.J. Augmented renal clearance is a common finding with worse clinical outcome in critically ill patients receiving antimicrobial therapy. J. Crit. Care 2013, 28, 695–700. [Google Scholar] [CrossRef]
  60. Udy, A.A.; Varghese, J.M.; Altukroni, M.; Briscoe, S.; McWhinney, B.C.; Ungerer, J.P.; Lipman, J.; Roberts., J.A. Subtherapeutic initial β-lactam concentrations in select critically ill patients: Association between augmented renal clearance and low trough drug concentrations. Chest 2012, 142, 30–39. [Google Scholar] [CrossRef]
  61. Sime, F.B.; Lassig-Smith, M.; Starr, T.; Stuart, J.; Pandey, S.; Parker, S.L.; Wallis, S.C.; Lipman, J.; Roberts, J.A. Population Pharmacokinetics of Unbound Ceftolozane and Tazobactam in Critically Ill Patients without Renal Dysfunction. Antimicrob. Agents Chemother. 2019, 63, e01265-19. [Google Scholar] [CrossRef] [PubMed]
  62. Nicolau, D.P.; De Waele, J.; Kuti, J.L.; Caro, L.; Larson, K.B.; Yu, B.; Gadzicki, E.; Zeng, Z.; Rhee, E.G.; Rizk, M.L. Pharmacokinetics and Pharmacodynamics of Ceftolozane/Tazobactam in Critically Ill Patients With Augmented Renal Clearance. Int. J. Antimicrob. Agents 2021, 57, 106299. [Google Scholar] [CrossRef] [PubMed]
  63. Fratoni, A.J.; Mah, J.W.; Nicolau, D.P.; Kuti, J.L. 1087. Imipenem-Cilastatin-Relebactam (I/R) Pharmacokinetics (PK) in Critically Ill Patients with Augmented Renal Clearance (ARC). Open Forum Infect. Dis. 2021, 8 (Suppl. 1), S635. [Google Scholar] [CrossRef]
  64. Torres, A.; Mouton, J.W.; Pea, F. Pharmacokinetics and Dosing of Ceftobiprole Medocaril for the Treatment of Hospital- and Community-Acquired Pneumonia in Different Patient Populations. Clin. Pharmacokinet. 2016, 55, 1507–1520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Clinical and Laboratory Standards Institute (CLSI). Performance Standards for Antimicrobial Susceptibility Testing, 32nd ed.; [Electronic]; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2022; ISBN 978-1-68440-135-2. [Google Scholar]
  66. Torres, A.; Sanchez-Garcia, M.; Demeyer, I. Pharmacokinetics, safety and tolerability of high-dose ceftobiprole medocaril administered as prolonged infusion in intensive-care-unit (ICU) patients [abstract O199]. In Proceedings of the 25th European Congress of Clinical Microbiology and Infectious Diseases, Copenhagen, Denmark, 25–28 April 2015. [Google Scholar]
  67. Katsube, T.; Wajima, T.; Ishibashi, T.; Arjona Ferreira, J.C.; Echols, R. Pharmacokinetic/Pharmacodynamic Modeling and Simulation of Cefiderocol, a Parenteral Siderophore Cephalosporin, for Dose Adjustment Based on Renal Function. Antimicrob. Agents Chemother. 2017, 61, e01381-16. [Google Scholar] [CrossRef] [Green Version]
  68. Katsube, T.; Echols, R.; Wajima, T. Pharmacokinetic and Pharmacodynamic Profiles of Cefiderocol, a Novel Siderophore Cephalosporin. Clin. Infect. Dis. 2019, 69 (Suppl. 7), S552–S558. [Google Scholar] [CrossRef] [PubMed]
  69. Kawaguchi, N.; Katsube, T.; Echols, R.; Wajima, T. Population Pharmacokinetic and Pharmacokinetic/Pharmacodynamic Analyses of Cefiderocol, a Parenteral Siderophore Cephalosporin, in Patients with Pneumonia, Bloodstream Infection/Sepsis, or Complicated Urinary Tract Infection. Antimicrob. Agents Chemother. 2021, 65, e01437-20. [Google Scholar] [CrossRef]
  70. Wunderink, R.G.; Matsunaga, Y.; Ariyasu, M.; Clevenbergh, P.; Echols, R.; Kaye, K.S.; Kollef, M.; Menon, A.; Pogue, J.M.; Shorr, A.F.; et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of Gram-negative nosocomial pneumonia (APEKS-NP): A randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 2021, 21, 213–225. [Google Scholar] [CrossRef]
  71. Bassetti, M.; Echols, R.; Matsunaga, Y.; Ariyasu, M.; Doi, Y.; Ferrer, R.; Lodisem, 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]
  72. Carlier, M.; Carrette, S.; Roberts, J.A.; Stove, V.; Verstraete, A.; Hoste, E.; Depuydt, P.; Decruyenaere, J.; Lipman, J.; Wallis, S.C.; et al. Meropenem and piperacillin/tazobactam prescribing in critically ill patients: Does augmented renal clearance affect pharmacokinetic/pharmacodynamic target attainment when extended infusions are used? Crit. Care 2013, 17, R84. [Google Scholar] [CrossRef] [Green Version]
  73. Nezarat, F.; Kobarfard, F.; Hassanpour, R.; Pourheidar, E.; Sistanizad, M. Evaluation of Recommended Doses of Meropenem in Patients with Augmented Renal Clearance, a Prospective Observational Study [Internet]. Preprint. July 2020. Available online: https://www.authorea.com/users/346825/articles/472668-evaluation-of-recommended-doses-of-meropenem-in-patients-with-augmented-renal-clearance-a-prospective-observational-study?commit=91ae3715eb2faad6236bf28b6499b0ef0b1edd3c (accessed on 14 August 2022).
  74. Tamatsukuri, T.; Ohbayashi, M.; Kohyama, N.; Kobayashi, Y.; Yamamoto, T.; Fukuda, K.; Nakamura, S.; Miyake, Y.; Dohi, K.; Kogo, M. The exploration of population pharmacokinetic model for meropenem in augmented renal clearance and investigation of optimum setting of dose. J. Infect. Chemother. 2018, 24, 834–840. [Google Scholar] [CrossRef] [PubMed]
  75. De Mendonça, A.; Vincent, J.L.; Suter, P.M.; Moreno, R.; Dearden, N.M.; Antonelli, M.; Takala, J.; Sprung, C.; Cantraine, F. Acute renal failure in the ICU: Risk factors and outcome evaluated by the SOFA score. Intensive Care Med. 2000, 26, 915–921. [Google Scholar] [CrossRef] [PubMed]
  76. Rawlins, M.; Cheng, V.; Raby, E.; Dyer, J.; Regli, A.; Ingram, P.; McWhinney, B.C.; Ungerer, J.P.J.; Roberts, J.A. Pharmacokinetics of Ceftolozane-Tazobactam during Prolonged Intermittent Renal Replacement Therapy. Chemotherapy 2018, 63, 203–206. [Google Scholar] [CrossRef] [PubMed]
  77. Kuti, J.L.; Ghazi, I.M.; Quintiliani, R.; Shore, E.; Nicolau, D.P. Treatment of multidrug-resistant Pseudomonas aeruginosa with ceftolozane/tazobactam in a critically ill patient receiving continuous venovenous haemodiafiltration. Int. J. Antimicrob. Agents 2016, 48, 342–343. [Google Scholar] [CrossRef] [PubMed]
  78. Bremmer, D.N.; Nicolau, D.P.; Burcham, P.; Chunduri, A.; Shidham, G.; Bauer, K.A. Ceftolozane/Tazobactam Pharmacokinetics in a Critically Ill Adult Receiving Continuous Renal Replacement Therapy. Pharmacotherapy 2016, 36, e30–e33. [Google Scholar] [CrossRef]
  79. Carbonell, N.; Aguilar, G.; Ferriols, R.; Huerta, R.; Ferreres, J.; Calabuig, M.; Juan, M.; Ezquer, C.; Colomina, J.; Blasco, M.L. Ceftolozane Pharmacokinetics in a Septic Critically Ill Patient under Different Extracorporeal Replacement Therapies. Antimicrob. Agents Chemother. 2019, 64, e01782-19. [Google Scholar] [CrossRef]
  80. Aguilar, G.; Ferriols, R.; Martínez-Castro, S.; Ezquer, C.; Pastor, E.; Carbonell, J.A.; Alós, M.; Navarro, D. Optimizing ceftolozane-tazobactam dosage in critically ill patients during continuous venovenous hemodiafiltration. Crit. Care 2019, 23, 145. [Google Scholar] [CrossRef] [Green Version]
  81. Oliver, W.D.; Heil, E.L.; Gonzales, J.P.; Mehrotra, S.; Robinett, K.; Saleeb, P.; Nicolau, D.P. Ceftolozane-Tazobactam Pharmacokinetics in a Critically Ill Patient on Continuous Venovenous Hemofiltration. Antimicrob. Agents Chemother. 2015, 60, 1899–1901. [Google Scholar] [CrossRef] [Green Version]
  82. Sime, F.B.; Lassig-Smith, M.; Starr, T.; Stuart, J.; Pandey, S.; Parker, S.L.; Wallis, S.C.; Lipman, J.; Roberts, J.A. A Population Pharmacokinetic Model-Guided Evaluation of Ceftolozane-Tazobactam Dosing in Critically Ill Patients Undergoing Continuous Venovenous Hemodiafiltration. Antimicrob. Agents Chemother. 2019, 64, e01655-19. [Google Scholar] [CrossRef] [Green Version]
  83. Wenzler, E.; Bunnell, K.L.; Bleasdale, S.C.; Benken, S.; Danziger, L.H.; Rodvold, K.A. Pharmacokinetics and Dialytic Clearance of Ceftazidime-Avibactam in a Critically Ill Patient on Continuous Venovenous Hemofiltration. Antimicrob. Agents Chemother. 2017, 61, e00464-17. [Google Scholar] [CrossRef] [Green Version]
  84. Soukup, P.; Faust, A.C.; Edpuganti, V.; Putnam, W.C.; McKinnell, J.A. Steady-State Ceftazidime-Avibactam Serum Concentrations and Dosing Recommendations in a Critically Ill Patient Being Treated for Pseudomonas aeruginosa Pneumonia and Undergoing Continuous Venovenous Hemodiafiltration. Pharmacotherapy 2019, 39, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  85. Kline, E.G.; Nguyen, M.H.T.; McCreary, E.K.; Wildfeuer, B.; Kohl, J.; Hughes, K.L.; Jones, C.E.; Doi, Y.; Shields, R.K. 1298. Population Pharmacokinetics of Ceftazidime-avibactam among Critically-ill Patients with and without Receipt of Continuous Renal Replacement Therapy. Open Forum Infect. Dis. 2020, 7 (Suppl. 1), S663–S664. [Google Scholar] [CrossRef]
  86. Zhang, X.S.; Wang, Y.Z.; Shi, D.W.; Xu, F.M.; Yu, J.H.; Chen, J.; Lin, G.Y.; Zhang, C.H.; Yu, X.B.; Tang, C.R. Efficacy and Pharmacodynamic Target Attainment for Ceftazidime-Avibactam Off-Label Dose Regimens in Patients with Continuous or Intermittent Venovenous Hemodialysis: Two Case Reports. Infect. Dis. Ther. 2022, 11, 2311–2319. [Google Scholar] [CrossRef] [PubMed]
  87. Kobic, E.; Gill, C.M.; Mochon, A.B.; Nicolasora, N.P.; Nicolau, D.P. Cefiderocol Pharmacokinetics in a Patient Receiving Continuous Venovenous Hemodiafiltration. Open Forum Infect. Dis. 2021, 8, ofab252. [Google Scholar] [CrossRef] [PubMed]
  88. Wenzler, E.; Butler, D.; Tan, X.; Katsube, T.; Wajima, T. Pharmacokinetics, Pharmacodynamics, and Dose Optimization of Cefiderocol during Continuous Renal Replacement Therapy. Clin. Pharmacokinet. 2022, 61, 539–552. [Google Scholar] [CrossRef]
  89. Cojutti, P.G.; Merelli, M.; De Stefanis, P.; Fregonese, C.; Lucchese, F.; Bassetti, M.; Pea, F. Disposition of ceftobiprole during continuous venous-venous hemodiafiltration (CVVHDF) in a single critically ill patient. Eur. J. Clin. Pharmacol. 2018, 74, 1671–1672. [Google Scholar] [CrossRef]
  90. Kufel, W.D.; Eranki, A.P.; Paolino, K.M.; Call, A.; Miller, C.D.; Mogle, B.T. In vivo pharmacokinetic analysis of meropenem/vaborbactam during continuous venovenous haemodialysis. J. Antimicrob. Chemother. 2019, 74, 2117–2118. [Google Scholar] [CrossRef]
  91. Boidin, C.; Moshiri, P.; Dahyot-Fizelier, C.; Goutelle, S.; Lefeuvre, S. Pharmacokinetic variability of beta-lactams in critically ill patients: A narrative review. Anaesth. Crit. Care Pain Med. 2020, 39, 87–109. [Google Scholar] [CrossRef]
  92. Crass, R.L.; Williams, P.; Roberts, J.A. The challenge of quantifying and managing pharmacokinetic variability of beta-lactams in the critically ill. Anaesth. Crit. Care Pain Med. 2020, 39, 27–29. [Google Scholar] [CrossRef]
  93. Abdul-Aziz, M.H.; Driver, E.; Lipman, J.; Roberts, J.A. New paradigm for rapid achievement of appropriate therapy in special populations: Coupling antibiotic dose optimization rapid microbiological methods. Expert Opin. Drug Metab. Toxicol. 2018, 14, 693–708. [Google Scholar] [CrossRef]
  94. Abdul-Aziz, M.H.; Alffenaar, J.C.; Bassetti, M.; Bracht, H.; Dimopoulos, G.; Marriott, D.; Neely, M.N.; Paiva, J.A.; Pea, F.; Sjovall, F.; et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: A Position Paper. Intensive Care Med. 2020, 46, 1127–1153. [Google Scholar]
  95. Wildschut, E.D.; Ahsman, M.J.; Allegaert, K.; Mathot, R.A.A.; Tibboel, D. Determinants of drug absorption in different ECMO circuits. Intensive Care Med. 2010, 36, 2109–2116. [Google Scholar] [CrossRef]
  96. Cies, J.J.; Moore, W.S.; Giliam, N.; Low, T.; Enache, A.; Chopra, A. Oxygenator Impact on Ceftolozane and Tazobactam in Extracorporeal Membrane Oxygenation Circuits. Pediatr. Crit. Care Med. 2020, 21, 276–282. [Google Scholar] [CrossRef]
  97. Cies, J.J.; Nikolos, P.; Moore, W.S.; Giliam, N.; Low, T.; Marino, D.; Deacon, J.; Enache, A.; Chopra, A. Oxygenator impact on meropenem/vaborbactam in extracorporeal membrane oxygenation circuits. Perfusion 2021, 37, 729–737. [Google Scholar] [CrossRef]
  98. Chung, E.K.; Cheatham, S.C.; Fleming, M.R.; Healy, D.P.; Kays, M.B. Population Pharmacokinetics and Pharmacodynamics of Meropenem in Nonobese, Obese, and Morbidly Obese Patients. J. Clin. Pharmacol. 2017, 57, 356–368. [Google Scholar] [CrossRef]
  99. Carrie, C.; Bentejac, M.; Cottenceau, V.; Masson, F.; Petit, L.; Cochard, J.F.; Sztark, F. Association between augmented renal clearance and clinical failure of antibiotic treatment in brain-injured patients with ventilator-acquired pneumonia: A preliminary study. Anaesth. Crit. Care Pain Med. 2018, 37, 35–41. [Google Scholar] [CrossRef]
  100. Carrié, C.; Chadefaux, G.; Sauvage, N.; de Courson, H.; Petit, L.; Nouette-Gaulain, K.; Pereira, B.; Biais, M. Increased β-Lactams dosing regimens improve clinical outcome in critically ill patients with augmented renal clearance treated for a first episode of hospital or ventilator-acquired pneumonia: A before and after study. Crit. Care 2019, 23, 379. [Google Scholar] [CrossRef] [Green Version]
  101. Dilworth, T.J.; Schulz, L.T.; Micek, S.T.; Kollef, M.H.; Rose, W.E. β-Lactam Therapeutic Drug Monitoring in Critically Ill Patients: Weighing the Challenges and Opportunities to Assess Clinical Value. Crit. Care Explor. 2022, 4, e0726. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of the study selection. * Three abstracts/posters were indentified through searching and were included in the review since pertinent data regarding PK/PD and target attainments in critically-ill patients were available.
Figure 1. Flow diagram of the study selection. * Three abstracts/posters were indentified through searching and were included in the review since pertinent data regarding PK/PD and target attainments in critically-ill patients were available.
Jcm 11 06898 g001
Table 1. Eligibility criteria for the included studies.
Table 1. Eligibility criteria for the included studies.
Population 1. Critically ill adult patients (ICU admissions)
2. Having any of the special scenarios (i.e., obesity, augmented renal clearance (ARC), extracorporeal interventions for life support (i.e., ECMO, prolonged intermittent renal replacement therapy (PIRRT), or continuous renal replacement therapy (CRRT))
Intervention Receiving novel β-lactam antibiotics of interest (including Ceftolozane/Tazobactam, Ceftazidime/Avibactam, Cefiderocol, Ceftobiprole, Imipenem/Relebactam and Meropenem/Vaborbactam) with reported dose being used
ComparatorNone
Outcomes 1. Reported PK profile of different novel β-lactam antibiotics under different scenarios studied
2. Reported PK/PD target attained with different doses used +/− clinical outcomes associated with use of drug therapy under studied circumstances
Study design PK studies (including phase 1 trials), population pharmacokinetic analyses (PopPK), PD studies, case-reports/case series, or clinical trials if PK/PD characteristics were reported
Table 3. Suggested initial dosing of novel β-lactam antibiotics based on aimed PK/PD targets among critically ill patients with special scenarios.
Table 3. Suggested initial dosing of novel β-lactam antibiotics based on aimed PK/PD targets among critically ill patients with special scenarios.
Novel β-Lactam AntibioticStandard Dose (Normal Kidney Function)Aimed PK/PD Target
(In-Vitro/Murine vs.
100% fT > MIC)
ARCRRTECMOObesity
Ceftolozane/Tazobactam1.5–3 g Q8h
(over 1–3 h)
C: 40% fT > MIC
T: 20% fT > CT:1 mg/L
1.5 g LD then 4.5 g/24 h C.I (if CI is not feasible, can consider 1.5 g Q8h for MIC ≤ 4 mg/L)CVVHDF
3 g LD then 0.75 g Q8h (over 1 h) for MIC ≤ 4 mg/L
NR NR
CVVH/CVVHD/PIRRT
NA
100% fT > MIC1.5 g LD then 4.5 g/24 h C.I
(if CI is not feasible, can consider 3 g Q8h for MIC ≤ 4 mg/L)
CVVHDF
3 g LD then 0.75 g Q8h (over 1 h) for MIC ≤ 4 mg/L
3 g Q8h (over 1 h)3 g Q8h (over 1 h)
PIRRT
Off dialysis days:LD 0.75 g, then 0.15 g Q8h (over 1 h)
Dialysis days:0.75 g Q12h (over 1 h)
CVVH/CVVHD
NA
Ceftazidime/Avibactam2.5 g Q8h
(over 2 h)
CAZ: 50% fT > MIC
AVI: 50% fT > CT:1 mg/L
2.5 g Q8h (over 2 h) for MIC ≤ 16 mg/LCVVH/CVVHD/CVVHDF/PIRRT
NA
NANA
100% fT > MICNRCVVHDF:
2.5 g Q8h (over 2 h) for MIC ≤ 8 mg/L
NANA
CVVH/CVVHD/PIRRT
NA
Cefiderocol2 g Q8h
(over 3 h)
75% fT > MIC2 g Q6h (over 3 h)CVVH/CVVHD/CVVHDF
All doses infused over 3 hr(for MIC ≤ 4 mg/L)
-Effluent flow rate ≤ 2 L/h → 1.5 g Q12h
-Effluent flow rate 2.1–3 L/h → 2 g Q12h
-Effluent flow rate 3.1–4 L/h→ 1.5 g Q8h
-Effluent flow rate ≥ 4.1 L/h→ 2 g Q8h
NR NA
PIRRT
NA
100% fT > MICNACVVHDF
2 g Q8h (over 3 h) for MIC ≤ 1 mg/L
CVVH/CVVHD/PIRRT
NR
2 g Q8h (over 3 h) for MIC ≤ 2 mg/LNA
Ceftobiprole0.5 g Q8h
(over 2 h)
25–40% fT > MICNR CVVH/CVVHD/CVVHDF/PIRRT
NA
NANR
100% fT > MIC0.5 g Q8h (increase infusion rate to over 4 h) for MIC ≤ 4 mg/LCVVHDF:
0.25 g Q12h (over 2 h) for MIC ≤ 2 mg/L
NA0.5 g Q8h (Increase infusion rate to over 4 h) for MIC ≤ 2 mg/L
CVVH/CVVHD/PIRRT
NA
Imipenem/Relebactam1.25 g Q6
(over 0.5 h)
IMI: 40% fT > MIC
REL: fAUC/MIC = 7.5
1.25 g Q6h (over 0.5 h) for MIC ≤ 2 mg/LCVVH/CVVHD/CVVHDF/PIRRT
NA
NANA
100% fT > MICNRNANA
Meropenem/Vaborbactam2–4 g Q8
(over 3 h)
ME: 45% fT > MIC
V: fAUC/MIC≥ 18–24
NACVVH/CVVHD/CVVHDF/PIRRT
NR
NANA
100% fT > MICNACVVHD
2 g Q8h (over 3 h) for MIC ≤ 4 mg/L
NANA
CVVH/CVVHDF/PIRRT
NA
IMI/REL: imipenem/relebactam, CAZ/AVI: ceftazidime/avibactam, C/T: ceftolozane/tazobactam, MEV: meropenem/vaborbactam, LD: loading dose, MIC: minimum inhibitory concentration, ARC: augmented renal clearance, MIC: minimum inhibitory concentration, RRT: renal replacement therapy, ECMO: extracorporeal membrane oxygenation, CVVH: continuous veno-venous hemofiltration, CVVHD: continuous venovenous hemodialysis, CVVHDF: continuous venovenous hemodiafiltration, PIRRT: prolonged intermittent renal replacement therapy, NR: not reported as a specific target of the included studies, NA: not available/no studies found, C.I: continuous infusion. ‡ Dosing adaptation from 100%fT > MIC target can be considered. However, the effects of higher exposures or the need of decreased dosing requirements have not be studied/reported for this conventional PK/PD index among critically ill patients presenting with this special scenario.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bakdach, D.; Elajez, R.; Bakdach, A.R.; Awaisu, A.; De Pascale, G.; Ait Hssain, A. Pharmacokinetics, Pharmacodynamics, and Dosing Considerations of Novel β-Lactams and β-Lactam/β-Lactamase Inhibitors in Critically Ill Adult Patients: Focus on Obesity, Augmented Renal Clearance, Renal Replacement Therapies, and Extracorporeal Membrane Oxygenation. J. Clin. Med. 2022, 11, 6898. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11236898

AMA Style

Bakdach D, Elajez R, Bakdach AR, Awaisu A, De Pascale G, Ait Hssain A. Pharmacokinetics, Pharmacodynamics, and Dosing Considerations of Novel β-Lactams and β-Lactam/β-Lactamase Inhibitors in Critically Ill Adult Patients: Focus on Obesity, Augmented Renal Clearance, Renal Replacement Therapies, and Extracorporeal Membrane Oxygenation. Journal of Clinical Medicine. 2022; 11(23):6898. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11236898

Chicago/Turabian Style

Bakdach, Dana, Reem Elajez, Abdul Rahman Bakdach, Ahmed Awaisu, Gennaro De Pascale, and Ali Ait Hssain. 2022. "Pharmacokinetics, Pharmacodynamics, and Dosing Considerations of Novel β-Lactams and β-Lactam/β-Lactamase Inhibitors in Critically Ill Adult Patients: Focus on Obesity, Augmented Renal Clearance, Renal Replacement Therapies, and Extracorporeal Membrane Oxygenation" Journal of Clinical Medicine 11, no. 23: 6898. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm11236898

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