1
|
Suya S, Nasomsong W, Santimaleeworagun W, Juntanawiwat P, Chatreewonanakul T, Saelim W. Monte Carlo simulation for dosage optimization of the best available therapy for bloodstream infections secondary to carbapenemase-producing Klebsiella pneumoniae in critically ill patients. J Glob Antimicrob Resist 2024; 39:257-265. [PMID: 39547573 DOI: 10.1016/j.jgar.2024.10.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/20/2024] [Accepted: 10/27/2024] [Indexed: 11/17/2024] Open
Abstract
OBJECTIVE We aimed to use Monte Carlo simulation, based on pharmacokinetic/pharmacodynamic targets, to investigate and determine the optimal dosage of the available combination therapies for carbapenem-resistant Klebsiella pneumoniae (CRKP) in critically ill patients. METHODS We collected CRKP clinical isolates from Phramongkutklao Hospital between October 2020 and June 2022. A molecular study of resistant genes was performed using polymerase chain reaction. Broth microdilution checkerboards were used to evaluate the mono- and synergistic antibiotic activities. Monte Carlo simulation was used to determine the optimal antibiotic regimens, based on the probability of target attainment (PTA) and cumulative fraction of response. RESULTS The 54 CRKP isolates were resistant to tigecycline (100%), colistin (75.9%), amikacin (70.4%), and gentamicin (63.0%). The most common carbapenemase genotype was blaoxacillinases (OXA)-48-like (42.6%), followed by blaNew Delhi metallo beta-lactamase (NDM) (29.6%) and coexistence of blaOXA-48-like and blaNDM (22.2%). Based on the PTA, synergistic and additive activities against CRKP isolates were observed with appropriate dosages of tigecycline-colistin (67.9%), tigecycline-gentamicin (62.2%), and tigecycline-amikacin (51.4%). CONCLUSIONS Tigecycline-colistin was the best available combination therapy for critically ill patients with CRKP, especially NDM. When used in combination with tigecycline, a colistin creatinine clearance of <90 mL/min can raise the cumulative fraction of response target and less nephrotoxicity.
Collapse
Affiliation(s)
- Sujareenoot Suya
- The College of Pharmacotherapy of Thailand, Nonthaburi, Thailand
| | - Worapong Nasomsong
- Department of Internal Medicine, Division of Infectious Diseases, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Wichai Santimaleeworagun
- Department of Pharmaceutical Care, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Pharmaceutical Initiative for Resistant Bacteria and Infectious Diseases Working Group [PIRBIG], Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Piraporn Juntanawiwat
- Department of Clinical Pathology, Division of Microbiology, Phramongkutklao Hospital, Bangkok, Thailand
| | | | - Weerayuth Saelim
- Department of Pharmaceutical Care, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Pharmaceutical Initiative for Resistant Bacteria and Infectious Diseases Working Group [PIRBIG], Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
| |
Collapse
|
2
|
Duong A, Marsot A. Nlmixr2 Versus NONMEM: An Evaluation of Maximum A Posteriori Bayesian Estimates Following External Evaluation of Gentamicin and Tobramycin Population Pharmacokinetic Models. Clin Pharmacol Drug Dev 2024; 13:739-747. [PMID: 38465725 DOI: 10.1002/cpdd.1395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
The objective of this project is to compare the results of the same study carried out on NONMEM and nlmixr2. This analysis consists of evaluating previously published population pharmacokinetic models of gentamicin and tobramycin in our population of interest with sparse concentrations. A literature review was performed to determine the gentamicin and tobramycin models in critically ill adult patients. In parallel, gentamicin and tobramycin dosing data, information on the treatment, the patient, and the bacteria were collected retrospectively in 2 Quebec establishments. The external evaluations were previously performed using NONMEM Version 7.5. Model equations were rewritten with R, and external evaluations were performed using nlmixr2. Predictive performance was assessed based on the estimation of bias and imprecision of the prediction error for maximum a posteriori (MAP) Bayesian PK parameter and observed concentrations. Comparison between nlmixr2 and NONMEM was performed on 4 gentamicin and 3 tobramycin population pharmacokinetic models. Compared to NONMEM, for gentamicin and tobramycin clearance and central volume of distribution, nlmixr2 produced individual pharmacokinetic parameters with bias values ranging from -32.5% to 5.67% and imprecision values ranging from 6.33% to 32.5%. Despite these differences, population bias and imprecision for sparse concentrations were low and ranged from 0% to 5.3% and 0.2% to 6.5%, respectively. The external evaluations performed with both software packages resulted in the same interpretation in terms of population predictive performance for all 7 models. Nlmxir2 showed comparable predictive performance with NONMEM with sparse concentrations that are, at most, sampled twice within a single dose administration (peak and trough).
Collapse
Affiliation(s)
- Alexandre Duong
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Montréal, QC, Canada
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Amélie Marsot
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Montréal, QC, Canada
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
- Centre de Recherche CHU Sainte-Justine, Montréal, QC, Canada
| |
Collapse
|
3
|
Liu F, Aulin LBS, Manson ML, Krekels EHJ, van Hasselt JGC. Unraveling the Effects of Acute Inflammation on Pharmacokinetics: A Model-Based Analysis Focusing on Renal Glomerular Filtration Rate and Cytochrome P450 3A4-Mediated Metabolism. Eur J Drug Metab Pharmacokinet 2023; 48:623-631. [PMID: 37715056 PMCID: PMC10624742 DOI: 10.1007/s13318-023-00852-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND AND OBJECTIVES: Acute inflammation caused by infections or sepsis can impact pharmacokinetics. We used a model-based analysis to evaluate the effect of acute inflammation as represented by interleukin-6 (IL-6) levels on drug clearance, focusing on renal glomerular filtration rate (GFR) and cytochrome P450 3A4 (CYP3A4)-mediated metabolism. METHODS A physiologically based model incorporating renal and hepatic drug clearance was implemented. Functions correlating IL-6 levels with GFR and in vitro CYP3A4 activity were derived and incorporated into the modeling framework. We then simulated treatment scenarios for hypothetical drugs by varying the IL-6 levels, the contribution of renal and hepatic drug clearance, and protein binding. The relative change in observed area under the concentration-time curve (AUC) was computed for these scenarios. RESULTS Inflammation showed opposite effects on drug exposure for drugs eliminated via the liver and kidney, with the effect of inflammation being inversely proportional to the extraction ratio (ER). For renally cleared drugs, the relative decrease in AUC was close to 30% during severe inflammation. For CYP3A4 substrates, the relative increase in AUC could exceed 50% for low-ER drugs. Finally, the impact of inflammation-induced changes in drug clearance is smaller for drugs with a larger unbound fraction. CONCLUSION This analysis demonstrates differences in the impact of inflammation on drug clearance for different drug types. The effects of inflammation status on pharmacokinetics may explain the inter-individual variability in pharmacokinetics in critically ill patients. The proposed model-based analysis may be used to further evaluate the effect of inflammation, i.e., by incorporating the effect of inflammation on other drug-metabolizing enzymes or physiological processes.
Collapse
Affiliation(s)
- Feiyan Liu
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Linda B S Aulin
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elke H J Krekels
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - J G Coen van Hasselt
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
4
|
Tilanus A, Drusano G. Inoculum-Based Dosing: A Novel Concept for Combining Time with Concentration-Dependent Antibiotics to Optimize Clinical and Microbiological Outcomes in Severe Gram Negative Sepsis. Antibiotics (Basel) 2023; 12:1581. [PMID: 37998783 PMCID: PMC10668771 DOI: 10.3390/antibiotics12111581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Certain classes of antibiotics show "concentration dependent" antimicrobial activity; higher concentrations result in increased bacterial killing rates, in contrast to "time dependent antibiotics", which show antimicrobial activity that depends on the time that antibiotic concentrations remain above the MIC. Aminoglycosides and fluoroquinolones are still widely used concentration-dependent antibiotics. These antibiotics are not hydrolyzed by beta-lactamases and are less sensitive to the inoculum effect, which can be defined as an increased MIC for the antibiotic in the presence of a relatively higher bacterial load (inoculum). In addition, they possess a relatively long Post-Antibiotic Effect (PAE), which can be defined as the absence of bacterial growth when antibiotic concentrations fall below the MIC. These characteristics make them interesting complementary antibiotics in the management of Multi-Drug Resistant (MDR) bacteria and/or (neutropenic) patients with severe sepsis. Global surveillance studies have shown that up to 90% of MDR Gram-negative bacteria still remain susceptible to aminoglycosides, depending on the susceptibility breakpoint (e.g., CLSI or EUCAST) being applied. This percentage is notably lower for fluoroquinolones but depends on the region, type of organism, and mechanism of resistance involved. Daily (high-dose) dosing of aminoglycosides for less than one week has been associated with significantly less nephro/oto toxicity and improved target attainment. Furthermore, higher-than-conventional dosing of fluoroquinolones has been linked to improved clinical outcomes. Beta-lactam antibiotics are the recommended backbone of therapy for severe sepsis. Since these antibiotics are time-dependent, the addition of a second concentration-dependent antibiotic could serve to quickly lower the bacterial inoculum, create PAE, and reduce Penicillin-Binding Protein (PBP) expression. Inadequate antibiotic levels at the site of infection, especially in the presence of high inoculum infections, have been shown to be important risk factors for inadequate resistance suppression and therapeutic failure. Therefore, in the early phase of severe sepsis, effort should be made to optimize the dose and quickly lower the inoculum. In this article, the authors propose a novel concept of "Inoculum Based Dosing" in which the decision for antibiotic dosing regimens and/or combination therapy is not only based on the PK parameters of the patient, but also on the presumed inoculum size. Once the inoculum has been lowered, indirectly reflected by clinical improvement, treatment simplification should be considered to further treat the infection.
Collapse
Affiliation(s)
- Alwin Tilanus
- Department of Infectious Diseases, Clinica Los Nogales, Calle 95 # 23-61, Bogota 110221, Colombia
| | - George Drusano
- Institute for Therapeutic Innovation, University of Florida, 6550 Sanger Road, Orlando, FL 32827, USA;
| |
Collapse
|
5
|
Khromov T, Dihazi GH, Brockmeyer P, Fischer A, Streit F. 24/7 Therapeutic Drug Monitoring of Beta-Lactam Antibiotics with CLAM-2000. Antibiotics (Basel) 2023; 12:1526. [PMID: 37887227 PMCID: PMC10604791 DOI: 10.3390/antibiotics12101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND The aim of this study was to evaluate the CLAM-2000 automated preanalytical sample preparation module with integrated liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) as a method for 24/7 therapeutic drug monitoring (TDM) of beta-lactam antibiotics in routine clinical diagnostics. METHODS Method validation was performed using quality control samples. Method comparison was performed with routine samples from patients treated with beta-lactam antibiotics. RESULTS The determination of piperacillin, meropenem, ceftazidime, flucloxacillin, and cefotaxime was performed using D5-piperacillin and D6-meropenem as internal standards. The linearity of the method was within the therapeutic range of beta-lactam antibiotics. The imprecision and accuracy data obtained from quality control samples were within 15%, and the imprecision of patient samples on the instrument was less than the 5% coefficient of variation (CV). Internal standards stored in the instrument at 9 °C for at least one week were stable, which facilitated reagent use and storage. CONCLUSION The CLAM-2000 (Shimadzu, Kyoto, Japan) provides reproducible results as an established routine instrument and is a useful tool for 24/7 TDM of beta-lactam antibiotics in routine clinical diagnostics.
Collapse
Affiliation(s)
- Tatjana Khromov
- Department of Clinical Chemistry, University Medical Center Goettingen, Robert-Koch Str. 40, D-37075 Goettingen, Germany; (G.H.D.); (A.F.); (F.S.)
| | - Gry Helene Dihazi
- Department of Clinical Chemistry, University Medical Center Goettingen, Robert-Koch Str. 40, D-37075 Goettingen, Germany; (G.H.D.); (A.F.); (F.S.)
| | - Phillipp Brockmeyer
- Department of Oral and Maxillofacial Surgery, University Medical Center Goettingen, Robert-Koch Str. 40, D-37075 Goettingen, Germany;
| | - Andreas Fischer
- Department of Clinical Chemistry, University Medical Center Goettingen, Robert-Koch Str. 40, D-37075 Goettingen, Germany; (G.H.D.); (A.F.); (F.S.)
| | - Frank Streit
- Department of Clinical Chemistry, University Medical Center Goettingen, Robert-Koch Str. 40, D-37075 Goettingen, Germany; (G.H.D.); (A.F.); (F.S.)
| |
Collapse
|
6
|
Abbasi MY, Chaijamorn W, Wiwattanawongsa K, Charoensareerat T, Doungngern T. Recommendations of Gentamicin Dose Based on Different Pharmacokinetic/Pharmacodynamic Targets for Intensive Care Adult Patients: A Redefining Approach. Clin Pharmacol 2023; 15:67-76. [PMID: 37427084 PMCID: PMC10329437 DOI: 10.2147/cpaa.s417298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Background In addition to the maximum plasma concentration (Cmax) to the minimum inhibitory concentration (MIC) ratio, the 24-hour area under the concentration-time curve (AUC24h) to MIC has recently been suggested as pharmacokinetic/pharmacodynamic (PK/PD) targets for efficacy and safety in once-daily dosing of gentamicin (ODDG) in critically ill patients. Purpose This study aimed to predict the optimal effective dose and risk of nephrotoxicity for gentamicin in critically ill patients for two different PK/PD targets within the first 3 days of infection. Methods The gathered pharmacokinetic and demographic data in critically ill patients from 21 previously published studies were used to build a one-compartment pharmacokinetic model. The Monte Carlo Simulation (MCS) method was conducted with the use of gentamicin once-daily dosing ranging from 5-10 mg/kg. The percentage target attainment (PTA) for efficacy, Cmax/MIC ~8-10 and AUC24h/MIC ≥110 targets, were studied. The AUC24h >700 mg⋅h/L and Cmin >2 mg/L were used to predict the risk of nephrotoxicity. Results Gentamicin 7 mg/kg/day could achieve both efficacy targets for more than 90% when the MIC was <0.5 mg/L. When the MIC increased to 1 mg/L, gentamicin 8 mg/kg/day could reach the PK/PD and safety targets. However, for pathogens with MIC ≥2 mg/L, no studied gentamicin doses were sufficient to reach the efficacy target. The risk of nephrotoxicity using AUC24h >700 mg⋅h/L was small, but the risk was greater when applying a Cmin target >2 mg/L. Conclusion Considering both targets of Cmax/MIC ~8-10 and AUC24h/MIC ≥110, an initial gentamicin dose of 8 mg/kg/day should be recommended in critically ill patients for pathogens with MIC of ≤1 mg/L. Clinical validation of our results is essential.
Collapse
Affiliation(s)
- Mohammad Yaseen Abbasi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Prince Songkla University, Hat Yai, Songkhla, 90110Thailand
| | - Weerachai Chaijamorn
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathum Wan, Bangkok, 10330, Thailand
| | - Kamonthip Wiwattanawongsa
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Prince Songkla University, Hat Yai, Songkhla, 90110Thailand
| | | | - Thitima Doungngern
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Prince Songkla University, Hat Yai, Songkhla, 90110Thailand
| |
Collapse
|
7
|
Hashiguchi Y, Matsumoto N, Oda K, Jono H, Saito H. Population Pharmacokinetics and AUC-Guided Dosing of Tobramycin in the Treatment of Infections Caused by Glucose-Nonfermenting Gram-Negative Bacteria. Clin Ther 2023:S0149-2918(23)00128-5. [PMID: 37120413 DOI: 10.1016/j.clinthera.2023.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/06/2023] [Accepted: 03/27/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE Tobramycin (TOB) exhibits variable pharmacokinetic properties due to the clinical condition of patients. This study aimed to investigate the AUC-guided dosing of TOB based on population pharmacokinetic analysis in the treatment of infections caused by Pseudomonas aeruginosa, Acinetobacter baumannii, and Stenotrophomonas maltophilia. METHODS This retrospective study was conducted between January 2010 and December 2020 after obtaining approval from our institutional review board. For 53 patients who received therapeutic drug monitoring of TOB, a population pharmacokinetic model was developed with covariates of estimated glomerular filtration rate using serum creatinine (eGFRcre) on clearance (CL) and weight on both CL and Vd in exponential error modeling (CL = 2.84 × [weight/70] × eGFRcre0.568, interindividual variability [IIV] = 31.1%; Vd = 26.3 × [weight/70], IIV = 20.2%; residual variability = 28.8%). FINDINGS The final regression model for predicting 30-day mortality was developed with risk factors of AUC during a 24-hour period after the first dose to MIC ratio (odds ratio [OR] = 0.996; 95% CI, 0.968-1.003) and serum albumin (OR = 0.137; 95% CI, 0.022-0.632). The final regression model for predicting acute kidney injury was developed with the risk factors of C-reactive protein (OR = 1.136; 95% CI, 1.040-1.266) and AUC during a 72-hour period after the first dose (OR = 1.004; 95% CI, 1.000-1.001). A dose of 8 or 15 mg/kg was beneficial for achievement of AUC during a 24-hour period after the first dose/MIC >80 and trough concentration <1 µg/mL in patients with preserved kidney function and TOB CL >4.47 L/h/70 kg in the events of MIC of 1 or 2 µg/mL, respectively. We propose that the first dose of 15, 11, 10, 8, and 7 mg/kg for eGFRcre >90, 60 to 89, 45 to 59, 30 to 44, and 15 to 29 mL/min/1.73 m2 be followed by therapeutic drug monitoring at peak and 24 hours after the first dose. IMPLICATIONS This study suggests that TOB use encourages the replacement of trough- and peak-targeted dosing with AUC-guided dosing.
Collapse
Affiliation(s)
- Yumi Hashiguchi
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan
| | - Naoya Matsumoto
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazutaka Oda
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan; Department of Infection Control, Kumamoto University Hospital, Kumamoto, Japan.
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan; Department of Infection Control, Kumamoto University Hospital, Kumamoto, Japan
| | - Hideyuki Saito
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan; Department of Infection Control, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
8
|
Duong A, Simard C, Williamson D, Marsot A. Model Re-Estimation: An Alternative for Poor Predictive Performance during External Evaluations? Example of Gentamicin in Critically Ill Patients. Pharmaceutics 2022; 14:pharmaceutics14071426. [PMID: 35890322 PMCID: PMC9315759 DOI: 10.3390/pharmaceutics14071426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/03/2022] [Accepted: 07/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background: An external evaluation is crucial before clinical applications; however, only a few gentamicin population pharmacokinetic (PopPK) models for critically ill patients included it in the model development. In this study, we aimed to evaluate gentamicin PopPK models developed for critically ill patients. Methods: The evaluated models were selected following a literature review on aminoglycoside PopPK models for critically ill patients. The data of patients were retrospectively collected from two Quebec hospitals, the external evaluation and model re-estimation were performed with NONMEM® (v7.5) and the population bias and imprecisions were estimated. Dosing regimens were simulated using the best performing model. Results: From the datasets of 39 and 48 patients from the two Quebec hospitals, none of the evaluated models presented acceptable values for bias and imprecision. Following model re-estimations, all models showed an acceptable predictive performance. An a priori dosing nomogram was developed with the best performing re-estimated model and was consistent based on recommended dosing regimens. Conclusion: Due to the poor predictive performance during the external evaluations, the latter must be prioritized during model development. Model re-estimation may be an alternative to developing a new model, especially when most known models display similar covariates.
Collapse
Affiliation(s)
- Alexandre Duong
- Faculté de Pharmacie, Université de Montréal, Montreal, QC H3T 1J4, Canada; (D.W.); (A.M.)
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence:
| | - Chantale Simard
- Institut Universitaire de Cardiologie et Pneumologie de Québec, Quebec, QC G1V 4G5, Canada;
- Faculté de Pharmacie, Université Laval, Quebec, QC G1V 0A6, Canada
| | - David Williamson
- Faculté de Pharmacie, Université de Montréal, Montreal, QC H3T 1J4, Canada; (D.W.); (A.M.)
- Hôpital Sacré-Cœur de Montréal, Université de Montréal, Montreal, QC H4J 1C5, Canada
| | - Amélie Marsot
- Faculté de Pharmacie, Université de Montréal, Montreal, QC H3T 1J4, Canada; (D.W.); (A.M.)
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Centre de Recherche, CHU Sainte Justine, Montreal, QC H3T 1C5, Canada
| |
Collapse
|
9
|
Hodiamont CJ, van den Broek AK, de Vroom SL, Prins JM, Mathôt RAA, van Hest RM. Clinical Pharmacokinetics of Gentamicin in Various Patient Populations and Consequences for Optimal Dosing for Gram-Negative Infections: An Updated Review. Clin Pharmacokinet 2022; 61:1075-1094. [PMID: 35754071 PMCID: PMC9349143 DOI: 10.1007/s40262-022-01143-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2022] [Indexed: 11/04/2022]
Abstract
Gentamicin is an aminoglycoside antibiotic with a small therapeutic window that is currently used primarily as part of short-term empirical combination therapy. Gentamicin dosing schemes still need refinement, especially for subpopulations where pharmacokinetics can differ from pharmacokinetics in the general adult population: obese patients, critically ill patients, paediatric patients, neonates, elderly patients and patients on dialysis. This review summarizes the clinical pharmacokinetics of gentamicin in these patient populations and the consequences for optimal dosing of gentamicin for infections caused by Gram-negative bacteria, highlighting new insights from the last 10 years. In this period, several new population pharmacokinetic studies have focused on these subpopulations, providing insights into the typical values of the most relevant pharmacokinetic parameters, the variability of these parameters and possible explanations for this variability, although unexplained variability often remains high. Both dosing schemes and pharmacokinetic/pharmacodynamic (PK/PD) targets varied widely between these studies. A gentamicin starting dose of 7 mg/kg based on total body weight (or on adjusted body weight in obese patients) appears to be the optimal strategy for increasing the probability of target attainment (PTA) after the first administration for the most commonly used PK/PD targets in adults and children older than 1 month, including critically ill patients. However, evidence that increasing the PTA results in higher efficacy is lacking; no studies were identified that show a correlation between estimated or predicted PK/PD target attainment and clinical success. Although it is unclear if performing therapeutic drug monitoring (TDM) for optimization of the PTA is of clinical value, it is recommended in patients with highly variable pharmacokinetics, including patients from all subpopulations that are critically ill (such as elderly, children and neonates) and patients on intermittent haemodialysis. In addition, TDM for optimization of the dosing interval, targeting a trough concentration of at least < 2 mg/L but preferably < 0.5–1 mg/L, has proven to reduce nephrotoxicity and is therefore recommended in all patients receiving more than one dose of gentamicin. The usefulness of the daily area under the plasma concentration–time curve for predicting nephrotoxicity should be further investigated. Additionally, more research is needed on the optimal PK/PD targets for efficacy in the clinical situations in which gentamicin is currently used, that is, as monotherapy for urinary tract infections or as part of short-term combination therapy.
Collapse
Affiliation(s)
- Caspar J Hodiamont
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Annemieke K van den Broek
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Suzanne L de Vroom
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan M Prins
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ron A A Mathôt
- Hospital Pharmacy and Clinical Pharmacology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Reinier M van Hest
- Hospital Pharmacy and Clinical Pharmacology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
10
|
He S, Cheng Z, Xie F. PK/PD guided gentamicin dosing in critically ill patients: a revisit of the Hartford nomogram. Int J Antimicrob Agents 2022; 59:106600. [DOI: 10.1016/j.ijantimicag.2022.106600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/29/2022] [Accepted: 04/28/2022] [Indexed: 11/05/2022]
|
11
|
The Role of Surface Enhanced Raman Scattering for Therapeutic Drug Monitoring of Antimicrobial Agents. CHEMOSENSORS 2022. [DOI: 10.3390/chemosensors10040128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The rapid quantification of antimicrobial agents is important for therapeutic drug monitoring (TDM), enabling personalized dosing schemes in critically ill patients. Highly sophisticated TDM technology is becoming available, but its implementation in hospitals is still limited. Among the various proposed techniques, surface-enhanced Raman scattering (SERS) stands out as one of the more interesting due to its extremely high sensitivity, rapidity, and fingerprinting capabilities. Here, we present a comprehensive review of various SERS-based novel approaches applied for direct and indirect detection and quantification of antibiotic, antifungal, and antituberculosis drugs in different matrices, particularly focusing on the challenges for successful exploitation of this technique in the development of assays for point-of-care tests.
Collapse
|
12
|
He S, Cheng Z, Xie F. Population Pharmacokinetics and Dosing Optimization of Gentamicin in Critically Ill Patients Undergoing Continuous Renal Replacement Therapy. Drug Des Devel Ther 2022; 16:13-22. [PMID: 35023902 PMCID: PMC8747548 DOI: 10.2147/dddt.s343385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose Appropriate gentamicin dosing in continuous renal replacement therapy (CRRT) patients remains undefined. This study aimed to develop a population pharmacokinetic (PK) model of gentamicin in CRRT patients and to infer the optimal dosing regimen for gentamicin. Methods Fourteen CRRT patients dosed with gentamicin were included to establish a population PK model to characterize the variabilities and influential covariates of gentamicin. The pharmacokinetic/pharmacodynamic (PK/PD) target attainment and risk of toxicity for different combinations of gentamicin regimens (3–7 mg/kg q24h) and CRRT effluent doses (30–50 mL/h/kg) were evaluated by Monte Carlo simulation. The probability of target attainment (PTA) was determined for the PK/PD indices of the ratio of drug peak concentration/minimum inhibitory concentration (Cmax/MIC > 10) and the ratio of area under the drug concentration–time curve/MIC over 24 h (AUC0-24h/MIC > 100), and the risk of toxicity was estimated by drug trough concentration thresholds (1 and 2 mg/L). Results A one-compartment model adequately described the PK characteristics of gentamicin. Covariates including body weight, age, gender, and CRRT modality did not influence the PK parameters of gentamicin based on our dataset. All studied gentamicin regimens failed to achieve satisfactory PTAs for pathogens with an MIC ≥2 mg/L. A good balance of PK/PD target attainment and risk of toxicity (>2 mg/L) was achieved under 7 mg/kg gentamicin q24h and 40 mL/kg/h CRRT dose for an MIC ≤1 mg/L. CRRT dose intensity had a significant impact on the target attainment of AUC0-24h/MIC >100 and risk of toxicity. Conclusion A combination of 7 mg/kg gentamicin q24h and 40 mL/kg/h CRRT dose might be considered as a starting treatment option for CRRT patients, and drug monitoring is required to manage toxicity.
Collapse
Affiliation(s)
- Sha He
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, People's Republic of China
| | - Zeneng Cheng
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, People's Republic of China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, People's Republic of China
| |
Collapse
|
13
|
Antimicrobial Activity Profiles and Potential Antimicrobial Regimens against Carbapenem-Resistant Enterobacterales Isolated from Multi-Centers in Western Thailand. Antibiotics (Basel) 2022; 11:antibiotics11030355. [PMID: 35326818 PMCID: PMC8944502 DOI: 10.3390/antibiotics11030355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
The spread of carbapenem-resistant Enterobacterales (CRE) constitutes a global health burden. Antimicrobial susceptibility and types of carbapenemase differ by geographic region. This study aimed to (1) examine the minimum inhibitory concentrations (MICs) and antibiotic resistance genes and (2) investigate antibiotic dosing regimens against CRE using Monte Carlo simulation. Clinical carbapenem-resistant Klebsiella pneumoniae (CRKP), Escherichia coli (CREC), and Enterobacter cloacae (CREclo) isolates were collected from various hospitals in western Thailand. Broth microdilution was performed, and the types of carbapenemase and mcr-1 genes were detected using polymerase chain reaction (PCR). Monte Carlo simulation was used to establish optimal antimicrobial dosing regimens meeting the criterion of a cumulative fraction of response (CFR) >90%. A total of 150 CRE isolates from 12 hospitals were included. The proportion of CRKP (76%) was greater than that of CREC (22%) and CREclo (2%). Regional hospitals reported higher rates of resistance than general hospitals. Most isolates were resistant to aztreonam and ceftazidime/avibactam, whereas they were highly susceptible to aminoglycosides. Most carbapenemases were NDM (47.33%), OXA-48 (43.33%) and NDM plus OXA-48 (6.67%); five OXA-48 positive isolates carried mcr-1 genes. Currently, high-dose tigecycline is the only optimal regimen against CRE isolates. Further extensive research on antibiotic synergism or new antibiotics should be conducted.
Collapse
|
14
|
Risk Factors Associated with Suboptimal Tobramycin Levels in the Medical Intensive Care Unit. Eur J Drug Metab Pharmacokinet 2022; 47:271-278. [PMID: 35029839 DOI: 10.1007/s13318-021-00749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND Optimal aminoglycoside dosing in critically ill patients represents a challenge for practitioners, especially in the medical intensive care unit (MICU). MICU patients exhibit altered pharmacokinetics due to pathophysiological changes the body undergoes in critical illness, leading to possible treatment failure. The literature surrounding optimal dosing and therapeutic drug monitoring strategies of aminoglycosides in MICU patients is scarce and conflicting. Additionally, only a few studies have investigated risk factors for suboptimal pharmacokinetic target obtainment. Currently, no definitive risk factors have been identified to predict suboptimal aminoglycoside target obtainment in MICU patients. OBJECTIVE The objective of this study was to determine risk factors for suboptimal pharmacokinetic target obtainment in patients receiving tobramycin in the MICU. METHODS This single-center, retrospective cohort study included patients 18-89 years old who received at least one 7 mg/kg tobramycin dose in the MICU from January, 1 2015 to September, 30 2020. Patients also had to have at least two detectable drug levels obtained at least one half-life apart following the first tobramycin dose. The primary outcome was to determine the incidence of optimal pharmacokinetic target obtainment, defined as a tobramycin maximum concentration (Cmax) ≥ 10 mcg/ml, and to identify the risk factors for suboptimal (Cmax < 10 mcg/mL) pharmacokinetic target obtainment, in MICU patients. Secondary outcomes were compared between suboptimal and optimal target obtainment in patients with culture confirmed gram-negative infection susceptible to tobramycin. These secondary outcomes included all-cause in-hospital mortality, ICU length of stay (LOS), hospital LOS, and vasopressor duration in those with shock. RESULTS A total of 230 patients were included in this retrospective study. For the primary outcome, 187 (81.3%) patients achieved optimal target obtainment. Through multivariate logistic regression, female sex and serum albumin < 2.5 g/dL were identified as independent risk factors for suboptimal target obtainment; [OR = 2.14; 95% CI (1.05-4.37), p = 0.037], [OR = 2.50; 95% CI (1.21-5.19), p = 0.014], respectively. Fifty-four (23%) patients had culture-confirmed gram-negative infections susceptible to tobramycin and were included in the subgroup analysis. Of these 54 patients, 11 (20.4%) did not achieve optimal target concentrations. In patients with culture-confirmed gram-negative infection, there was no difference between patients with optimal target obtainment and suboptimal target obtainment in ICU LOS, hospital LOS, all-cause mortality, or vasopressor duration in those with shock. CONCLUSIONS Among patients receiving their first dose of tobramycin in the MICU, 81.3% obtained an optimal serum concentration. Female sex and serum albumin < 2.5 g/dL were identified as risk factors for suboptimal target obtainment; however, further research is warranted to assess the utility of using these two covariates as risk factors for more aggressive dosing in critically ill MICU patients.
Collapse
|
15
|
Smith NM, Chan A, Wilkinson LA, Chua HC, Nguyen TD, de Souza H, Shah AP, D'Argenio DZ, Mergenhagen KA. Open-source maximum a posteriori-bayesian dosing AdDS to current therapeutic drug monitoring: Adapting to the era of individualized therapy. Pharmacotherapy 2021; 41:953-963. [PMID: 34618919 DOI: 10.1002/phar.2631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 11/07/2022]
Abstract
Recent updates in the therapeutic drug monitoring (TDM) guidelines for vancomycin have rekindled interest in maximum a posteriori-Bayesian (MAP-Bayesian) estimation of patient-specific pharmacokinetic parameters. To create a versatile infrastructure for MAP-Bayesian dosing of vancomycin or other drugs, a freely available, R-based software package, Advanced Dosing Solutions (AdDS), was created to facilitate clinical implementation of these improved TDM methods. The objective of this study was to utilize AdDS for pre- and post-processing of data in order to streamline the therapeutic management of vancomycin in healthy and obese veterans. Patients from a local Veteran Affairs hospital were utilized to compare the process of full re-estimation versus Bayesian updating of priors on healthy adult and obese patient populations for use with AdDS. Twenty-four healthy veterans were utilized to train (14/24) and test (10/24) the base pharmacokinetic model of vancomycin while comparing the effects of updated and fully re-estimated priors. This process was repeated with a total of 18 obese veterans for both training (11/18) and testing (7/18). Comparison of MAP objective function between the original and re-estimated models for healthy adults indicated that 78.6% of the subjects in the training and 70.0% of the subjects in the testing datasets had similar or improved predictions by the re-estimated model. For obese veterans, 81.8% of subjects in the training dataset and 85.7% of subjects in the testing dataset had similar or improved predictions. Re-estimation of model parameters provided more significant improvements in objective function compared with Bayesian updating, which may be a useful strategy in cases where sufficient samples and subjects are available. The generation of bespoke regimens based on patient-specific clearance and minimal sampling may improve patient care by addressing fundamental pharmacokinetic differences in healthy and obese veteran populations.
Collapse
Affiliation(s)
- Nicholas M Smith
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - Arthur Chan
- Veterans Affair Hospital of Western New York, New York, New York, USA
| | - Laura A Wilkinson
- Veterans Affair Hospital of Western New York, New York, New York, USA
| | - Hubert C Chua
- CHI Baylor St. Luke's Medical Center, Houston, Texas, USA
| | - Thomas D Nguyen
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - Harriet de Souza
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - Anant P Shah
- School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
- New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York, USA
| | - David Z D'Argenio
- Biomedical Simulations Resource, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
16
|
De Winter S, van Hest R, Dreesen E, Annaert P, Wauters J, Meersseman W, Van den Eede N, Desmet S, Verelst S, Vanbrabant P, Peetermans W, Spriet I. Quantification and Explanation of the Variability of First-Dose Amikacin Concentrations in Critically Ill Patients Admitted to the Emergency Department: A Population Pharmacokinetic Analysis. Eur J Drug Metab Pharmacokinet 2021; 46:653-663. [PMID: 34297338 DOI: 10.1007/s13318-021-00698-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND There may be a difference between the determinants of amikacin exposure in emergency department (ED) versus intensive care (ICU) patients, and the peak amikacin concentration varies widely between patients. Moreover, when the first dose of antimicrobials is administered to septic patients admitted to the ED, fluid resuscitation and vasopressors have just been initiated. Nevertheless, population pharmacokinetic modelling data for amikacin in ED patients are unavailable. OBJECTIVE The aim of this study was to quantify the interindividual variability (IIV) in the pharmacokinetics of amikacin in patients admitted to the ED and to identify the patient characteristics that explain this IIV. METHODS Patients presenting at the ED with severe sepsis or septic shock were randomly assigned to receive amikacin 25 mg/kg or 15 mg/kg intravenously. Blood samples were collected at 1, 6 and 24 h after the onset of the first amikacin infusion. Data were analysed using nonlinear mixed-effects modelling. RESULTS A two-compartment population pharmacokinetic model was developed based on 279 amikacin concentrations from 97 patients. The IIV in clearance (CL) and central distribution volume (V1) were 71% and 26%, respectively. Body mass index (BMI), serum total protein level, serum sodium level, and fluid balance 24 h after amikacin administration explained 30% of the IIV in V1, leaving 18% of the IIV unexplained. BMI and creatinine clearance according to the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation 24 h after amikacin administration explained 46% of the IIV in CL, and 39% remained unexplained. CONCLUSION The IIV of amikacin pharmacokinetics in ED patients is large. Higher doses may be considered in patients with low serum sodium levels, low total protein levels, or a high fluid balance. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT02365272.
Collapse
Affiliation(s)
- Sabrina De Winter
- Department of Pharmacy, Univesity Hospitals Leuven, Leuven, Belgium.
| | - Reinier van Hest
- Department of Hospital Pharmacy and Clinical Pharmacology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Erwin Dreesen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium.,BioNotus, Galileilaan 15, 2845, Niel, Belgium
| | - Joost Wauters
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Wouter Meersseman
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Nele Van den Eede
- Laboratory of Clinical Bacteriology and Mycology, University Hospitals Leuven, Leuven, Belgium
| | - Stefanie Desmet
- Laboratory of Clinical Bacteriology and Mycology, University Hospitals Leuven, Leuven, Belgium
| | - Sandra Verelst
- Department of Emergency Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Peter Vanbrabant
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Willy Peetermans
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Isabel Spriet
- Department of Pharmacy, Univesity Hospitals Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Optimizing Antimicrobial Drug Dosing in Critically Ill Patients. Microorganisms 2021; 9:microorganisms9071401. [PMID: 34203510 PMCID: PMC8305961 DOI: 10.3390/microorganisms9071401] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
A fundamental step in the successful management of sepsis and septic shock is early empiric antimicrobial therapy. However, for this to be effective, several decisions must be addressed simultaneously: (1) antimicrobial choices should be adequate, covering the most probable pathogens; (2) they should be administered in the appropriate dose, (3) by the correct route, and (4) using the correct mode of administration to achieve successful concentration at the infection site. In critically ill patients, antimicrobial dosing is a common challenge and a frequent source of errors, since these patients present deranged pharmacokinetics, namely increased volume of distribution and altered drug clearance, which either increased or decreased. Moreover, the clinical condition of these patients changes markedly over time, either improving or deteriorating. The consequent impact on drug pharmacokinetics further complicates the selection of correct drug schedules and dosing during the course of therapy. In recent years, the knowledge of pharmacokinetics and pharmacodynamics, drug dosing, therapeutic drug monitoring, and antimicrobial resistance in the critically ill patients has greatly improved, fostering strategies to optimize therapeutic efficacy and to reduce toxicity and adverse events. Nonetheless, delivering adequate and appropriate antimicrobial therapy is still a challenge, since pathogen resistance continues to rise, and new therapeutic agents remain scarce. We aim to review the available literature to assess the challenges, impact, and tools to optimize individualization of antimicrobial dosing to maximize exposure and effectiveness in critically ill patients.
Collapse
|
18
|
The Synergistic Activity and Optimizing Doses of Tigecycline in Combination with Aminoglycosides against Clinical Carbapenem-Resistant Klebsiella pneumoniae Isolates. Antibiotics (Basel) 2021; 10:antibiotics10060736. [PMID: 34204561 PMCID: PMC8234075 DOI: 10.3390/antibiotics10060736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 01/22/2023] Open
Abstract
Carbapenem-resistant Enterobacteriaceae (CRE), especially carbapenem-resistant Klebsiella pneumoniae (CRKP), are among the largest pathogenic threats to humans. The available antibiotic treatment options for combating CRKP are limited. Colistin-resistant Enterobacteriaceae (CoRE) have also been reported worldwide, including in Thailand. Therefore, this study aimed (1) to determine minimum inhibitory concentrations (MICs) and synergistic activities of antibiotics of CRKP, and (2) to determine the probability target of attainment (PTA) and cumulative fraction of response (CFR) using pharmacokinetic/pharmacodynamic (PK/PD) data. Clinical CRKP isolates were obtained from Phramongkutklao Hospital (June to November 2020). Broth microdilution and checkerboard techniques were used to determine the mono- and synergistic activities of antibiotics. Carbapenemase and mcr-1 genes were also identified by polymerase chain reaction (PCR). The optimal antibiotic regimens were evaluated using Monte Carlo simulations. Forty-nine CRKP isolates were collected, 40 of which were CoRKP strains. The MIC50 and MIC90 of tigecycline, amikacin, and gentamicin were 1 and 2 µg/mL, 4 and 16 µg/mL, and 0.25 and 4 µg/mL, respectively. None of any isolates expressed the mcr-1 gene, whereas blaOXA-48 (53.1%) and blaOXA-48 plus blaNDM (42.9%) were detected. Synergistic activity was observed in 8.2% of isolates for tigecycline combined with amikacin or gentamicin. Additive activity was observed in 75.5% of isolates for tigecycline-amikacin and 69.4% for tigecycline-gentamicin, and no antagonism was observed. High-dose antibiotic regimens achieved the PTA target. The general recommended dose of combination regimens began with 200 mg tigecycline and 25 mg/kg amikacin, or 7 mg/kg gentamicin, followed by 100 mg tigecycline every 12 h and 15 mg/kg amikacin or 5 mg/kg gentamicin every 24 h. In conclusion, tigecycline plus aminoglycosides might be a potential regimen against CRKP and CoRKP. The appropriate combination regimen based on MIC-based dose adjustment can improve optimal antibiotic dosing. Further research via clinical studies will be necessary to confirm these results.
Collapse
|
19
|
Xie F, Wang Y, Peng Y, Cheng Z, Li S. Pharmacokinetic/pharmacodynamic evaluation of tobramycin dosing in critically ill patients: the Hartford nomogram does not fit. J Antimicrob Chemother 2021; 76:2335-2341. [PMID: 34096596 DOI: 10.1093/jac/dkab164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 04/23/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Extended-interval dosing of tobramycin is widely applied in patients with the Hartford nomogram as a representative, while this dosing approach has not been extensively evaluated in critically ill patients. The goal of this study was to characterize the pharmacokinetics of tobramycin and to evaluate the appropriateness of the Hartford nomogram in critically ill patients. METHODS A retrospective analysis was performed based on a medical critical care database. The extracted concentration data of tobramycin were used for the construction of the population pharmacokinetic model using a non-linear mixed-effects modelling approach. Real-world data-based simulations were conducted to evaluate the pharmacodynamic target attainment (Cmax/MIC ≥10) and safety (concentration <0.5 mg/L for at least 4 h) of the Hartford nomogram. RESULTS A population pharmacokinetic model was built based on 307 measurements in 140 unique patients and externally validated by an independent study dataset. A two-compartment model was optimal for the structure model and creatinine clearance remained as the only covariate in the final model correlating to the clearance of tobramycin. Simulations indicated that the Hartford nomogram is effective for infections due to pathogens with an MIC of ≤1 mg/L, but not with an MIC of 2 mg/L. The percentage of patients who reached the non-toxicity target was quite low under the Hartford nomogram and a further extension of the dosing interval was necessary to minimize the toxicity. CONCLUSIONS The Hartford nomogram was not suitable for critically ill patients with pathogen MICs of 2 mg/L and drug monitoring is required to manage efficacy and toxicity.
Collapse
Affiliation(s)
- Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yan Wang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yaru Peng
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Zeneng Cheng
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Sanwang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| |
Collapse
|
20
|
Aminoglycosides in Critically Ill Septic Patients With Acute Kidney Injury Receiving Continuous Renal Replacement Therapy: A Multicenter, Observational Study. Clin Ther 2021; 43:1116-1124. [PMID: 34039478 DOI: 10.1016/j.clinthera.2021.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/19/2021] [Accepted: 04/16/2021] [Indexed: 11/23/2022]
Abstract
PURPOSE Data on aminoglycoside stewardship in critically ill septic patients with acute kidney injury (AKI) needing continuous renal replacement therapy (CRRT) are scarce. The objectives of the study were to determine, during CRRT, the time window with low likelihood for safe reinjection and the proportion of inappropriate reinjection. METHODS A post hoc observational analysis of a multicenter randomized trial comparing the risk of hemodialysis catheter infection with ethanol lock vs placebo in critically ill patients with AKI was conducted. Eligible patients were adults in intensive care units from 6 French hospitals. Any patient with AKI needing CRRT and receiving an antimicrobial therapy for a septic episode occurring before (≤24 hours) or during CRRT was included. The aminoglycoside orders were left to the physicians' discretion, but high dose once daily was the schedule of aminoglycoside administration. FINDINGS A total of 145 septic episodes treated by aminoglycosides were analyzed in patients receiving CRRT. A mean (SD) of 1.6 (0.8) amikacin and 1.8 (1.2) gentamicin administrations per patient were observed. During CRRT, Cmax was 17.3 mg/L (interquartile range, 13.2-22.5 mg/L) for gentamicin and 50 mg/L (interquartile range, 43.7-76.6 mg/L) for amikacin. The plasma drug concentration at 24 hours (CH24) was 2.3 mg/L (interquartile range, 1.6-3.2 mg/L) for gentamicin and 9.3 (interquartile range, 6.6-12.0 mg/L) for amikacin. Sixty-five Cmin dosages remained above the reinjection threshold. Inappropriate reinjection was observed in 11 of 65 episodes (17%). Inappropriate reinjection (defined by, at the reinjection time, Cmin dosages above the threshold; ie, Cmin >2 mg/L for gentamicin and >5 mg/L for amikacin) was observed in 17% of analyzed episodes. Most patients did not need reinjection until approximately ≥30 hours after their initial administration. IMPLICATIONS During CRRT, as indicated by the CH24 value, which was higher than the recommended threshold, the interval to obtain a Cmin low enough to allow for redosing aminoglycosides is significantly longer than 24 hours. This interval is not always respected and leads to an of inappropriate reinjection rate of 17%. ClinicalTrials.gov identifier: ISRCTNCT00875069. (Clin Ther. 2021;XX:XXX-XXX) © 2021 Elsevier HS Journals, Inc.
Collapse
|
21
|
Duong A, Simard C, Wang YL, Williamson D, Marsot A. Aminoglycosides in the Intensive Care Unit: What Is New in Population PK Modeling? Antibiotics (Basel) 2021; 10:antibiotics10050507. [PMID: 33946905 PMCID: PMC8145041 DOI: 10.3390/antibiotics10050507] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Although aminoglycosides are often used as treatment for Gram-negative infections, optimal dosing regimens remain unclear, especially in ICU patients. This is due to a large between- and within-subject variability in the aminoglycoside pharmacokinetics in this population. Objective: This review provides comprehensive data on the pharmacokinetics of aminoglycosides in patients hospitalized in the ICU by summarizing all published PopPK models in ICU patients for amikacin, gentamicin, and tobramycin. The objective was to determine the presence of a consensus on the structural model used, significant covariates included, and therapeutic targets considered during dosing regimen simulations. Method: A literature search was conducted in the Medline/PubMed database, using the terms: ‘amikacin’, ‘gentamicin’, ‘tobramycin’, ‘pharmacokinetic(s)’, ‘nonlinear mixed effect’, ‘population’, ‘intensive care’, and ‘critically ill’. Results: Nineteen articles were retained where amikacin, gentamicin, and tobramycin pharmacokinetics were described in six, 11, and five models, respectively. A two-compartment model was used to describe amikacin and tobramycin pharmacokinetics, whereas a one-compartment model majorly described gentamicin pharmacokinetics. The most recurrent significant covariates were renal clearance and bodyweight. Across all aminoglycosides, mean interindividual variability in clearance and volume of distribution were 41.6% and 22.0%, respectively. A common consensus for an optimal dosing regimen for each aminoglycoside was not reached. Conclusions: This review showed models developed for amikacin, from 2015 until now, and for gentamicin and tobramycin from the past decades. Despite the growing challenges of external evaluation, the latter should be more considered during model development. Further research including new covariates, additional simulated dosing regimens, and external validation should be considered to better understand aminoglycoside pharmacokinetics in ICU patients.
Collapse
Affiliation(s)
- Alexandre Duong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada; (Y.L.W.); (D.W.); (A.M.)
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-514-343-6111
| | - Chantale Simard
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Centre de Recherche, Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, QC G1V 4G5, Canada
| | - Yi Le Wang
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada; (Y.L.W.); (D.W.); (A.M.)
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - David Williamson
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada; (Y.L.W.); (D.W.); (A.M.)
- Hôpital Sacré-Cœur de Montréal, Montréal, QC H4J 1C5, Canada
| | - Amélie Marsot
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada; (Y.L.W.); (D.W.); (A.M.)
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Centre de Recherche, CHU Sainte Justine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
22
|
Roger C, Louart B, Elotmani L, Barton G, Escobar L, Koulenti D, Lipman J, Leone M, Muller L, Boutin C, Amour J, Banakh I, Cousson J, Bourenne J, Constantin JM, Albanese J, Roberts JA, Lefrant JY. An international survey on aminoglycoside practices in critically ill patients: the AMINO III study. Ann Intensive Care 2021; 11:49. [PMID: 33740157 PMCID: PMC7979853 DOI: 10.1186/s13613-021-00834-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/05/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND While aminoglycosides (AG) have been used for decades, debate remains on their optimal dosing strategy. We investigated the international practices of AG usage specifically regarding dosing and therapeutic drug monitoring (TDM) in critically ill patients. We conducted a prospective, multicentre, observational, cohort study in 59 intensive-care units (ICUs) in 5 countries enrolling all ICU patients receiving AG therapy for septic shock. RESULTS We enrolled 931 septic ICU patients [mean ± standard deviation, age 63 ± 15 years, female 364 (39%), median (IQR) SAPS II 51 (38-65)] receiving AG as part of empirical (761, 84%) or directed (147, 16%) therapy. The AG used was amikacin in 614 (66%), gentamicin in 303 (33%), and tobramycin in 14 (1%) patients. The median (IQR) duration of therapy was 2 (1-3) days, the number of doses was 2 (1-2), the median dose was 25 ± 6, 6 ± 2, and 6 ± 2 mg/kg for amikacin, gentamicin, and tobramycin respectively, and the median dosing interval was 26 (23.5-43.5) h. TDM of Cmax and Cmin was performed in 437 (47%) and 501 (57%) patients, respectively, after the first dose with 295 (68%) patients achieving a Cmax/MIC > 8 and 353 (71%) having concentrations above Cmin recommended thresholds. The ICU mortality rate was 27% with multivariable analysis showing no correlation between AG dosing or pharmacokinetic/pharmacodynamic target attainment and clinical outcomes. CONCLUSION Short courses of high AG doses are mainly used in ICU patients with septic shock, although wide variability in AG usage is reported. We could show no correlation between PK/PD target attainment and clinical outcome. Efforts to optimize the first AG dose remain necessary. Trial registration Clinical Trials, NCT02850029, registered on 29th July 2016, retrospectively registered, https://www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Claire Roger
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France. .,Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France.
| | - Benjamin Louart
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France.,Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France
| | - Loubna Elotmani
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France.,Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France
| | - Greg Barton
- St Helens and Knowsley Hospitals NHS Trust, Liverpool, UK
| | - Leslie Escobar
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Despoina Koulenti
- The University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Second Critical Care Department, Attikon University Hospital, Athens, Greece
| | - Jeffrey Lipman
- Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France.,The University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Marc Leone
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Marseille, Marseille, France
| | - Laurent Muller
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France.,Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France
| | - Caroline Boutin
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France
| | - Julien Amour
- Institute of Perfusion, Critical Care Medicine and Anesthesiology in Cardiac Surgery (IPRA), Hôpital Privé Jacques Cartier, Massy, France
| | | | - Joel Cousson
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Reims, Reims, France
| | - Jeremy Bourenne
- Department of Emergency and Intensive Care Medicine, University Hospital of Marseille, Hôpital de La Timone, Marseille, France
| | - Jean-Michel Constantin
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Jacques Albanese
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Marseille, Hôpital de La Conception, Marseille, France
| | - Jason A Roberts
- Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France.,The University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Pharmacy Department, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Jean-Yves Lefrant
- Department of Intensive Care Medicine, Division of Anesthesiology, Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, Place du Professeur Robert Debré, 30 029, Nîmes cedex 9, France.,Equipe D, Caractéristiques Féminines Des Interfaces Vasculaires (IMAGINE), Faculté de Médecine, Univ Montpellier, 2992, Montpellier, France
| | | |
Collapse
|
23
|
[S3 Guideline Sepsis-prevention, diagnosis, therapy, and aftercare : Long version]. Med Klin Intensivmed Notfmed 2021; 115:37-109. [PMID: 32356041 DOI: 10.1007/s00063-020-00685-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Lin Z, Chen DY, Zhu YW, Jiang ZL, Cui K, Zhang S, Chen LH. Population pharmacokinetic modeling and clinical application of vancomycin in Chinese patients hospitalized in intensive care units. Sci Rep 2021; 11:2670. [PMID: 33514803 PMCID: PMC7846798 DOI: 10.1038/s41598-021-82312-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/18/2021] [Indexed: 11/19/2022] Open
Abstract
Management of vancomycin administration for intensive care units (ICU) patients remains a challenge. The aim of this study was to describe a population pharmacokinetic model of vancomycin for optimizing the dose regimen for ICU patients. We prospectively enrolled 466 vancomycin-treated patients hospitalized in the ICU, collected trough or approach peak blood samples of vancomycin and recorded corresponding clinical information from July 2015 to December 2017 at Tai Zhou Hospital of Zhejiang Province. The pharmacokinetics of vancomycin was analyzed by nonlinear mixed effects modeling with Kinetica software. Internal and external validation was evaluated by the maximum likelihood method. Then, the individual dosing regimens of the 92 patients hospitalized in the ICU whose steady state trough concentrations exceeded the target range (10–20 μg/ml) were adjusted by the Bayes feedback method. The final population pharmacokinetic model show that clearance rate (CL) of vancomycin will be raised under the conditions of dopamine combined treatment, severe burn status (Burn-S) and increased total body weight (TBW), but reduced under the conditions of increased serum creatinine (Cr) and continuous renal replacement therapy status; Meanwhile, the apparent distribution volume (V) of vancomycin will be enhanced under the terms of increased TBW, however decreased under the terms of increased age and Cr. The population pharmacokinetic parameters (CL and V) according to the final model were 3.16 (95%CI 2.83, 3.40) L/h and 60.71 (95%CI 53.15, 67.46). The mean absolute prediction error for external validation by the final model was 12.61% (95CI 8.77%, 16.45%). Finally, the prediction accuracy of 90.21% of the patients’ detected trough concentrations that were distributed in the target range of 10–20 μg/ml after dosing adjustment was found to be adequate. There is significant heterogeneity in the CL and V of vancomycin in ICU patients. The constructed model is sufficiently precise for the Bayesian dose prediction of vancomycin concentrations for the population of ICU Chinese patients.
Collapse
Affiliation(s)
- Zhong Lin
- Department of Clinical Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Ximen Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Dan-Yang Chen
- Rehabilitation Department, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Xi Men Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Yan-Wu Zhu
- Department of Clinical Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Ximen Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Zheng-Li Jiang
- Department of Clinical Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Ximen Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Ke Cui
- Intensive Care Unit, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Xi Men Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Sheng Zhang
- Intensive Care Unit, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Xi Men Street No. 150, Linhai, 317000, Zhejiang Province, China
| | - Li-Hua Chen
- Public Scientific Research Platform, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Xi Men Street No. 150, Linhai, 317000, Zhejiang Province, China.
| |
Collapse
|
25
|
Redpath A, Hallowell GD, Bowen IM. Use of aminoglycoside antibiotics in equine clinical practice; a questionnaire-based study of current use. Vet Med Sci 2020; 7:279-288. [PMID: 33099884 PMCID: PMC8025607 DOI: 10.1002/vms3.382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023] Open
Abstract
Veterinary use of the aminoglycoside antibiotics is under increasing scrutiny. This questionnaire-based study aimed to document the use of aminoglycosides with a particular focus on gentamicin. An online questionnaire was delivered to generalist equine veterinary surgeons and specialists in internal medicine to determine the perceived importance, frequency of use and routes of administration of the aminoglycoside antibiotics. A series of hypothetical scenarios were also evaluated regarding gentamicin. Data were compared to evaluate the impact of the level of specialisation on prescribing practices for different antibiotics using Chi-squared and Fischer's exact tests. Data were analysed from 111 responses. Gentamicin was commonly used empirically without culture and susceptibility testing. Generalists were more likely to use gentamicin only after susceptibility testing than specialists in a variety of clinical presentations including respiratory diseases, septic peritonitis, acute febrile diarrhoea, cellulitis and contaminated limb wounds (p < 0.01). Intravenous administration of gentamicin was most common, although inhaled and regional administration of gentamicin and amikacin were also described. Amikacin was most commonly used by intra-articular administration. Gentamicin was more likely to be used in high-risk procedures or contaminated surgeries (86% and 74%, respectively) compared with clean surgery (32%; p < 0.0001). Gentamicin was often used perioperatively in horses undergoing exploratory celiotomy and more commonly used in horses undergoing an enterotomy (90%) than without and enterotomy (79%; p = 0.04). Most respondents (86%) used gentamicin at a dose of 6.6 mg/kg in adults, with few changing their dosing strategies based on the presence of sepsis, although higher doses were more reported in foals (7-15 mg/kg) irrespective of the presence of sepsis. Aminoglycosides are widely used in equine practice and use outside current EU marketing authorisations is common. Stewardship of the aminoglycoside antibiotics could be enhanced in both generalists and specialists through the more frequent use of susceptibility testing, regional administration and dose adjustment, especially in foals.
Collapse
Affiliation(s)
- Adam Redpath
- Oakham Veterinary HospitalSchool of Veterinary Medicine and ScienceUniversity of NottinghamLoughboroughUK
| | - Gayle D. Hallowell
- School of Veterinary Medicine and ScienceUniversity of NottinghamLoughboroughUK
| | - Ian Mark Bowen
- Oakham Veterinary HospitalSchool of Veterinary Medicine and ScienceUniversity of NottinghamLoughboroughUK
| |
Collapse
|
26
|
Chai MG, Cotta MO, Abdul-Aziz MH, Roberts JA. What Are the Current Approaches to Optimising Antimicrobial Dosing in the Intensive Care Unit? Pharmaceutics 2020; 12:pharmaceutics12070638. [PMID: 32645953 PMCID: PMC7407796 DOI: 10.3390/pharmaceutics12070638] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 12/29/2022] Open
Abstract
Antimicrobial dosing in the intensive care unit (ICU) can be problematic due to various challenges including unique physiological changes observed in critically ill patients and the presence of pathogens with reduced susceptibility. These challenges result in reduced likelihood of standard antimicrobial dosing regimens achieving target exposures associated with optimal patient outcomes. Therefore, the aim of this review is to explore the various methods for optimisation of antimicrobial dosing in ICU patients. Dosing nomograms developed from pharmacokinetic/statistical models and therapeutic drug monitoring are commonly used. However, recent advances in mathematical and statistical modelling have resulted in the development of novel dosing software that utilise Bayesian forecasting and/or artificial intelligence. These programs utilise therapeutic drug monitoring results to further personalise antimicrobial therapy based on each patient’s clinical characteristics. Studies quantifying the clinical and cost benefits associated with dosing software are required before widespread use as a point-of-care system can be justified.
Collapse
Affiliation(s)
- Ming G. Chai
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia; (M.G.C.); (M.O.C.); (M.H.A.-A.)
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Woollongabba 4102, Australia
| | - Menino O. Cotta
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia; (M.G.C.); (M.O.C.); (M.H.A.-A.)
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Woollongabba 4102, Australia
| | - Mohd H. Abdul-Aziz
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia; (M.G.C.); (M.O.C.); (M.H.A.-A.)
| | - Jason A. Roberts
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia; (M.G.C.); (M.O.C.); (M.H.A.-A.)
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Woollongabba 4102, Australia
- Departments of Pharmacy and Intensive Care, Royal Brisbane and Women’s Hospital, Brisbane 4006, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nimes University Hospital, University of Montpellier, 30021 Nimes, France
- Correspondence:
| |
Collapse
|
27
|
Cobussen M, Haeseker MB, Stoffers J, Wanrooij VHM, Savelkoul PHM, Stassen PM. Renal safety of a single dose of gentamicin in patients with sepsis in the emergency department. Clin Microbiol Infect 2020; 27:S1198-743X(20)30376-1. [PMID: 32621972 DOI: 10.1016/j.cmi.2020.06.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVES To determine the effect of a single dose of gentamicin on the incidence and persistence of acute kidney injury (AKI) in patients with sepsis in the emergency department (ED). METHODS We retrospectively studied patients with sepsis in the ED in three hospitals. Local antibiotic guidelines recommended a single dose of gentamicin as part of empirical therapy in selected patients in one hospital, whereas the other two hospitals did not. Multivariate analysis was used to evaluate the effect of gentamicin and other potential risk factors on the incidence and persistence of AKI after admission. AKI was defined according to the KDIGO (Kidney Disease Improving Global Outcomes) criteria. RESULTS Of 1573 patients, 571 (32.9%) received a single dose of gentamicin. At admission, 181 (31.7%) of 571 of the gentamicin-treated and 228 (22.8%) of 1002 of the non-gentamicin-treated patients had AKI (p < 0.001). After admission, AKI occurred in 64 (12.0%) of 571 patients who received gentamicin and in 82 (8.9%) of 1002 people in the control group (p 0.06). Multivariate analysis showed that shock (odds ratio (OR), 2.72; 95% CI, 1.31-5.67), diabetes mellitus (OR, 1.49; 95% CI, 1.001-2.23) and higher baseline (i.e. before admission) serum creatinine levels (OR, 1.007; 95% CI, 1.005-1.009) were associated with the development of AKI after admission, but not receipt of gentamicin (OR, 1.29; 95% CI, 0.89-1.86). Persistent AKI was rare in both the group that received gentamicin (16/260, 6.2%) and the group that did not (15/454, 3.3%, p 0.09). CONCLUSIONS With regard to renal function, a single dose of gentamicin in patients with sepsis in the ED is safe. The development of AKI after admission was associated with shock, diabetes mellitus and higher baseline creatinine level.
Collapse
Affiliation(s)
- M Cobussen
- Department of Medical Microbiology, Maastricht University Medical Center, Maastricht, the Netherlands; CAPHRI School for Public Health and Primary Care, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, Rijnstate Hospital, Arnhem, the Netherlands.
| | - M B Haeseker
- Department of Medical Microbiology, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Medical Microbiology, ReinierHaga Medical Diagnostic Center, Delft, the Netherlands
| | - J Stoffers
- Department of Internal Medicine, Division of General Medicine, Section Acute Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Internal Medicine, Zuyderland Medical Center, Heerlen, the Netherlands
| | - V H M Wanrooij
- Department of Internal Medicine, Division of General Medicine, Section Acute Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Internal Medicine, Zuyderland Medical Center, Heerlen, the Netherlands
| | - P H M Savelkoul
- Department of Medical Microbiology, Maastricht University Medical Center, Maastricht, the Netherlands; CAPHRI School for Public Health and Primary Care, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - P M Stassen
- CAPHRI School for Public Health and Primary Care, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, Division of General Medicine, Section Acute Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
28
|
Grucz TM, Kruer RM, Bernice F, Lipsett PA, Dorman T, Sugrue D, Jarrell AS. Aminoglycoside Dosing and Volume of Distribution in Critically Ill Surgery Patients. Surg Infect (Larchmt) 2020; 21:859-864. [PMID: 32302517 DOI: 10.1089/sur.2020.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: At a tertiary referral and Level I trauma center, current institutional guidelines suggest initial aminoglycoside doses of gentamicin or tobramycin 4 mg/kg and amikacin 16 mg/kg for patients admitted to surgical intensive care units (SICUs) with suspected gram-negative infection. The objective of this study was to evaluate initial aminoglycoside dosing and peak serum drug concentrations in critically ill surgery patients to characterize the aminoglycoside volume of distribution (Vd) and determine an optimal standardized dosing strategy. Methods: This retrospective, observational, single-center study included adult SICU patients who received an aminoglycoside for additional gram-negative coverage. Descriptive statistics were used to evaluate the patient population, aminoglycoside dosing, and Vd. Multivariable linear regression was applied to determine variables associated with greater aminoglycoside Vd. The mortality rate was compared in patients who achieved adequate initial peak concentrations versus those who did not. Results: One hundred seventeen patients received an aminoglycoside in the SICUs, of whom 58 had an appropriately timed peak concentration measurement. The mean Acute Physiology, Age, and Chronic Health Evaluation (APACHE) II score was 27.8 ± 8.9. The Vd in patients receiving gentamicin, tobramycin, and amikacin was 0.49 ± 0.10, 0.41 ± 0.09, and 0.53 ± 0.13 L/kg, respectively. Together, the mean aminoglycoside Vd was 0.50 ± 0.12 L/kg. Gentamicin or tobramycin 5 mg/kg achieved goal peak concentrations in 24 patients (63.2%), and amikacin 20 mg/kg achieved the desired concentrations in nine patients (50.0%). Net fluid status, Body Mass Index, and vasopressor use were not predictive of Vd. There was no difference in the in-hospital mortality rate in patients who achieved adequate peak concentrations versus those who did not (26.8% versus 26.7%; p = 0.99). Conclusion: High aminoglycoside doses are needed in critically ill surgery patients to achieve adequate initial peak concentrations because of the high Vd. Goal peak concentrations were optimized at doses of gentamicin or tobramycin 5 mg/kg, and amikacin 20 mg/kg.
Collapse
Affiliation(s)
- Traci M Grucz
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Rachel M Kruer
- Department of Pharmacy, Indiana University Health, Indianapolis, Indiana, USA
| | - Fidelia Bernice
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Pamela A Lipsett
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Todd Dorman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David Sugrue
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Andrew S Jarrell
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Derendorf H, Heinrichs T, Reimers T, Lebert C, Brinkmann A. Calculated initial parenteral treatment of bacterial infections: Pharmacokinetics and pharmacodynamics. GMS INFECTIOUS DISEASES 2020; 8:Doc17. [PMID: 32373442 PMCID: PMC7186811 DOI: 10.3205/id000061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This is the third chapter of the guideline "Calculated initial parenteral treatment of bacterial infections in adults - update 2018" in the 2nd updated version. The German guideline by the Paul-Ehrlich-Gesellschaft für Chemotherapie e.V. (PEG) has been translated to address an international audience. The chapter features the pharmacokinetic and pharmacodynamics properties of the most frequently used antiinfective agents.
Collapse
Affiliation(s)
- Hartmut Derendorf
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, USA
| | | | - Tobias Reimers
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, USA
| | | | - Alexander Brinkmann
- Klinik für Anästhesie, operative Intensivmedizin und spezielle Schmerztherapie, Klinikum Heidenheim, Germany
| |
Collapse
|
30
|
Hartman SJF, Orriëns LB, Zwaag SM, Poel T, de Hoop M, de Wildt SN. External Validation of Model-Based Dosing Guidelines for Vancomycin, Gentamicin, and Tobramycin in Critically Ill Neonates and Children: A Pragmatic Two-Center Study. Paediatr Drugs 2020; 22:433-444. [PMID: 32507958 PMCID: PMC7383037 DOI: 10.1007/s40272-020-00400-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The Dutch Pediatric Formulary (DPF) increasingly bases its guidelines on model-based dosing simulations from pharmacokinetic studies. This resulted in nationwide dose changes for vancomycin, gentamicin, and tobramycin in 2015. OBJECTIVE We aimed to evaluate target attainment of these altered, model-based doses in critically ill neonates and children. METHODS This was a retrospective cohort study in neonatal intensive care unit (NICU) and pediatric ICU (PICU) patients receiving vancomycin, gentamicin, or tobramycin between January 2015 and March 2017 in two university hospitals. The first therapeutic drug monitoring concentration for each patient was collected, as was clinical and dosing information. Vancomycin and tobramycin target trough concentrations were 10-15 and ≤ 1 mg/L, respectively. Target gentamicin trough and peak concentrations were < 1 and 8-12 mg/L, respectively. RESULTS In total, 482 patients were included (vancomycin [PICU] n = 62, [NICU] n = 102; gentamicin [NICU] n = 97; tobramycin [NICU] n = 221). Overall, median trough concentrations were within the target range for all cohorts but showed large interindividual variability, causing nontarget attainment. Trough concentrations were outside the target range in 66.1%, 60.8%, 14.7%, and 23.1% of patients in these four cohorts, respectively. Gentamicin peak concentrations were outside the range in 69% of NICU patients (term neonates 87.1%, preterm infants 57.1%). Higher creatinine concentrations were associated with higher vancomycin and tobramycin trough concentrations. CONCLUSION This study illustrates the need to validate model-based dosing advice in the real-world setting as both sub- and supratherapeutic concentrations of vancomycin, gentamicin, and tobramycin were very prevalent. Our data underline the necessity for further individualization by addressing the high interindividual variability to improve target attainment.
Collapse
Affiliation(s)
- Stan J. F. Hartman
- grid.10417.330000 0004 0444 9382Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud Institute of Health Sciences, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Lynn B. Orriëns
- grid.10417.330000 0004 0444 9382Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud Institute of Health Sciences, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Samanta M. Zwaag
- grid.10417.330000 0004 0444 9382Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud Institute of Health Sciences, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Tim Poel
- grid.10417.330000 0004 0444 9382Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud Institute of Health Sciences, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Marika de Hoop
- grid.489189.50000 0001 0708 7338Royal Dutch Pharmacists Association (KNMP), Den Haag, The Netherlands ,Dutch Knowledge Center Pharmacotherapy for Children, The Hague, The Netherlands
| | - Saskia N. de Wildt
- grid.10417.330000 0004 0444 9382Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud Institute of Health Sciences, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands ,grid.5645.2000000040459992XIntensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands ,Dutch Knowledge Center Pharmacotherapy for Children, The Hague, The Netherlands
| |
Collapse
|
31
|
Sumi CD, Heffernan AJ, Lipman J, Roberts JA, Sime FB. What Antibiotic Exposures Are Required to Suppress the Emergence of Resistance for Gram-Negative Bacteria? A Systematic Review. Clin Pharmacokinet 2019; 58:1407-1443. [PMID: 31325141 DOI: 10.1007/s40262-019-00791-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The rates of antibiotic resistance in Gram-negative bacteria are increasing. One method to minimize resistance emergence may be optimization of antibiotic dosing regimens to achieve drug exposure that suppress the emergence of resistance. OBJECTIVE The aim of this systematic review was to describe the antibiotic exposures associated with suppression of the emergence of resistance for Gram-negative bacteria. METHODS We conducted a search of four electronic databases. Articles were included if the antibiotic exposure required to suppress the emergence of resistance in a Gram-negative bacterial isolate was described. Among studies, 57 preclinical studies (in vitro and in vivo) and 2 clinical studies 59 included investigated the monotherapy of antibiotics against susceptible and/or intermediate Gram-negative bacteria. RESULTS The pharmacokinetic/pharmacodynamic (PK/PD) indices reported to suppress the emergence of antibiotic resistance for various classes were β-lactam antibiotic minimum concentration to minimum inhibitory concentration (Cmin/MIC) ≥ 4; aminoglycoside maximum concentration to MIC (Cmax/MIC) ratio ≥ 20; fluoroquinolones, area under the concentration-time curve from 0 to 24 h to mutant prevention concentration (AUC24/MPC) ≥ 35; tetracyclines, AUC24 to MIC (AUC24/MIC) ratio ≥ 50; polymyxin B, AUC24/MIC ≥ 808; and fosfomycin, AUC24/MIC ≥ 3136. However, the exposures required to suppress the emergence of resistance varied depending on the specific antibiotic tested, the duration of the experiment, the bacterial species and the specific bacterial isolate tested. Importantly, antibiotic exposures required to suppress the emergence of resistance generally exceeded that associated with clinical efficacy. CONCLUSION The benefits of implementing such high PK/PD targets must be balanced with the potential risks of antibiotic-associated toxicity.
Collapse
Affiliation(s)
- Chandra Datta Sumi
- Centre for Translational Anti-Infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Aaron J Heffernan
- Centre for Translational Anti-Infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- School of Medicine, Griffith University, Gold Coast, QLD, Australia
| | - Jeffrey Lipman
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Jason A Roberts
- Centre for Translational Anti-Infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia.
- The University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
- Pharmacy Department, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
| | - Fekade B Sime
- Centre for Translational Anti-Infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
32
|
Abstract
Objective: Sepsis is a deadly infection that causes injury to tissues and organs. Infection and anti-infective treatment are the eternal themes of sepsis. The successful control of infection is a key factor of resuscitation for sepsis and septic shock. This review examines evidence for the treatment of sepsis. This evidence is combined with clinical experiments to reveal the rules and a standard flowchart of anti-infection therapy for sepsis. Data Sources: We retrieved information from the PubMed database up to October 2018 using various search terms and their combinations, including sepsis, septic shock, infection, antibiotics, and anti-infection. Study Selection: We included data from peer-reviewed journals printed in English on the relationships between infections and antibiotics. Results: By combining the literature review and clinical experience, we propose a 6Rs rule for sepsis and septic shock management: right patients, right time, right target, right antibiotics, right dose, and right source control. This rule encompasses rational decisions regarding the timing of treatment, the identification of the correct pathogen, the selection of appropriate antibiotics, the formulation of a scientifically based antibiotic dosage regimen, and the adequate control of infectious foci. Conclusions: This review highlights how to recognize and treat sepsis and septic shock and provides rules and a standard flowchart for anti-infection therapy for sepsis and septic shock for use in the clinical setting.
Collapse
|
33
|
Muller AE, Huttner B, Huttner A. Therapeutic Drug Monitoring of Beta-Lactams and Other Antibiotics in the Intensive Care Unit: Which Agents, Which Patients and Which Infections? Drugs 2019; 78:439-451. [PMID: 29476349 DOI: 10.1007/s40265-018-0880-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibiotics are among the medications most frequently administered to the critically ill, a population with high levels of intra- and inter-individual pharmacokinetic variability. Our knowledge of the relationships among antibiotic dosing, exposure and clinical effect in this population has increased in recent decades. Therapeutic drug monitoring (TDM) of serum antibiotic concentrations is the most practical means of assessing adequate antibiotic exposure, though until recently, it has been underutilised for this end. Now TDM is becoming more widespread, particularly for the beta-lactam antibiotics, a class historically thought to have a wide therapeutic range. We review the basic requirements, indications, and targets for effective TDM of the glycopeptides, aminoglycosides, quinolones and beta-lactam antibiotics in the adult intensive-care setting, with a special focus on TDM of the beta-lactam antibiotics, the most widely used antibiotic class.
Collapse
Affiliation(s)
- Anouk E Muller
- Department of Medical Microbiology, Haaglanden Medisch Centrum, The Hague, The Netherlands.,Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - Benedikt Huttner
- Division of Infectious Diseases, University Hospitals of Geneva, Rue Gabrielle-Gentil-Perret 4, 1205, Geneva, Switzerland
| | - Angela Huttner
- Division of Infectious Diseases, University Hospitals of Geneva, Rue Gabrielle-Gentil-Perret 4, 1205, Geneva, Switzerland.
| |
Collapse
|
34
|
Cobussen M, Stassen PM, Posthouwer D, van Tiel FH, Savelkoul PHM, Havenith T, Haeseker MB. Improving peak concentrations of a single dose regime of gentamicin in patients with sepsis in the emergency department. PLoS One 2019; 14:e0210012. [PMID: 30668571 PMCID: PMC6342306 DOI: 10.1371/journal.pone.0210012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/14/2018] [Indexed: 11/19/2022] Open
Abstract
Objective To achieve an optimal effect in patients with sepsis at the emergency department (ED), the gentamicin peak-concentration should be sufficiently high (i.e. peak-concentration/MIC ≥8–10). ICU patients with sepsis often need higher gentamicin doses to achieve sufficiently high peak-concentrations. The aim of this study is to investigate which dose is needed to reach adequate peak-concentrations in patients presenting with sepsis at the ED. Methods Patients with sepsis at the ED were included from August 2015 until February 2017. Peak-concentrations were measured in blood 30 minutes after the first gentamicin dose. The study consisted of three phases. In the first phase, peak-concentrations were measured after a standard dose of 5mg/kg. In the second phase, a simulation ((peak-concentration/actual dose) × simulated dose) was performed to determine which dose was needed to reach adequate gentamicin peak-concentrations of ≥16mg/L. In the third phase, peak-concentrations were measured for the best simulated dose. Results In phase one, of 86 patients who received a dose of 5mg/kg, 34 (39.5%) patients did not reach the target peak-concentration of ≥16mg/L, and 73 (84.9%) did not reach ≥20mg/L. In phase two, the simulation showed that with a dose of 7mg/kg 83 (96.5%) patients would reach peak-concentrations ≥16mg/L, and 67 (77.9%) of ≥20mg/L. In phase three, 53 patients received a dose of 7mg/kg, of whom 45 (84.9%) reached peak-concentrations of ≥16mg/L, and 31 (58.5%) of ≥20mg/L. Conclusion Patients with sepsis at the ED need higher doses of gentamicin. A dose of 7mg/kg is needed to achieve adequate peak-concentrations in the majority of patients.
Collapse
Affiliation(s)
- Maarten Cobussen
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
- * E-mail:
| | - Patricia M. Stassen
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Dirk Posthouwer
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Frank H. van Tiel
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Thomas Havenith
- Department of Clinical Pharmacy, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Michiel B. Haeseker
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
35
|
Abdul-Aziz MH, Driver E, Lipman J, Roberts JA. 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. [PMID: 29865877 DOI: 10.1080/17425255.2018.1484452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Some special patient populations (e.g. critically ill, burns, hematological malignancy, post-major surgery, post-major trauma) have characteristics that lead to higher rates of failure and mortality associated with infection. Choice of effective antibiotics and optimized doses are challenging in these patients that are commonly infected by multidrug-resistant pathogens. Areas covered: A review of the importance of diagnosis and the place of newer microbiological methods (e.g. whole-genome sequencing) to ensure rapid transition from empiric to directed antibiotic therapy is provided. The effects of pathophysiological changes on antibiotic pharmacokinetics are also provided. Expert opinion: Product information dosing regimens do not address the pharmacokinetic alterations that can occur in special patient populations and increase the likelihood of therapeutic failure and the emergence of bacterial resistance. Altered dosing approaches, supplemented with the use of dosing software and therapeutic drug monitoring, may be needed to ensure optimal antibiotic exposure and better therapeutic outcomes in these patients with severe infection. Dose optimization needs to be coupled with advanced microbiological techniques that enable rapid microbiological identification and characterization of resistance mechanism to ensure that maximally effective directed therapy can be chosen.
Collapse
Affiliation(s)
- Mohd H Abdul-Aziz
- a Faculty of Medicine , University of Queensland Centre for Clinical Research, The University of Queensland , Brisbane , Queensland , Australia
| | - Elicia Driver
- a Faculty of Medicine , University of Queensland Centre for Clinical Research, The University of Queensland , Brisbane , Queensland , Australia
| | - Jeffrey Lipman
- a Faculty of Medicine , University of Queensland Centre for Clinical Research, The University of Queensland , Brisbane , Queensland , Australia.,b Department of Intensive Care Medicine , Royal Brisbane and Women's Hospital , Brisbane , Queensland , Australia
| | - Jason A Roberts
- a Faculty of Medicine , University of Queensland Centre for Clinical Research, The University of Queensland , Brisbane , Queensland , Australia.,b Department of Intensive Care Medicine , Royal Brisbane and Women's Hospital , Brisbane , Queensland , Australia.,c Department of Pharmacy , Royal Brisbane and Women's Hospital , Brisbane , Queensland , Australia.,d School of Pharmacy, Centre for Translational Anti-infective Pharmacodynamics , The University of Queensland , Brisbane , Queensland , Australia
| |
Collapse
|
36
|
Boidin C, Jenck S, Bourguignon L, Torkmani S, Roussey-Jean A, Ledochowski S, Marry L, Ammenouche N, Dupont H, Marçon F, Allaouchiche B, Bohé J, Lepape A, Goutelle S, Friggeri A. Determinants of amikacin first peak concentration in critically ill patients. Fundam Clin Pharmacol 2018; 32:669-677. [PMID: 29660162 DOI: 10.1111/fcp.12374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/26/2018] [Accepted: 04/09/2018] [Indexed: 11/29/2022]
Abstract
Amikacin antimicrobial effect has been correlated with the ratio of the peak concentration (Cmax ) to the minimum inhibitory concentration. A target Cmax ≥ 60-80 mg/L has been suggested. It has been shown that such target is not achieved in a large proportion of critically ill patients in intensive care units. A retrospective analysis was performed to examine the determinants of Cmax ≥ 80 mg/L on the first peak in 339 critically ill patients treated by amikacin. The influence of available variables on Cmax target attainment was analyzed using a classification and regression tree (CART) and logistic regression. Mean Cmax in the 339 patients was 73.0 ± 23.9 mg/L, with a target attainment rate (TAR, Cmax ≥ 80 mg/L) of 37.5%. In CART analysis, the strongest predictor of amikacin target peak attainment was dose per kilogram of lean body weight (dose/LBW). TAR was 60.1% in patients with dose/LBW ≥ 37.8 vs. 19.9% in patients with lower dose/LBW (OR = 6.0 (95% CI: 3.6-10.2)). Renal function was a secondary predictor of Cmax . Logistic regression analysis identified dose per kilogram of ideal body weight (OR = 1.13 (95% CI: 1.09-1.17)) and creatinine clearance (OR = 0.993 (95% CI: 0.988-0.998)) as predictors of target peak achievement. Based on our results, an amikacin dose ≥ 37.8 mg/kg of LBW should be used to optimize the attainment of Cmax ≥ 80 mg/L after the first dose in critically ill patients. An even higher dose may be necessary in patients with normal renal function.
Collapse
Affiliation(s)
- Clément Boidin
- Groupement Hospitalier Nord, Service de Pharmacie, Hôpital Pierre Garraud, Hospices Civils de Lyon, 136 rue du Commandant Charcot, 69322, LyonCedex 5, France.,Laboratoire de Biométrie et Biologie Évolutive, Université Claude Bernard Lyon 1, UMR CNRS 5558, University of Lyon, Bât. Grégor Mendel, 43 bd du 11 novembre 1918, 69622, Villeurbanne Cedex, France
| | - Sophie Jenck
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France
| | - Laurent Bourguignon
- Groupement Hospitalier Nord, Service de Pharmacie, Hôpital Pierre Garraud, Hospices Civils de Lyon, 136 rue du Commandant Charcot, 69322, LyonCedex 5, France.,Laboratoire de Biométrie et Biologie Évolutive, Université Claude Bernard Lyon 1, UMR CNRS 5558, University of Lyon, Bât. Grégor Mendel, 43 bd du 11 novembre 1918, 69622, Villeurbanne Cedex, France.,ISPB - Faculté de Pharmacie de Lyon, Université Claude Bernard Lyon 1, University of Lyon, 8 Avenue Rockefeller, 69003, Lyon, France
| | - Sejad Torkmani
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France
| | - Aurore Roussey-Jean
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France
| | - Stanislas Ledochowski
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France
| | - Lucie Marry
- Service d'Anesthésie et de Réanimation, Centre Hospitalier Universitaire Amiens-Picardie, Site Sud, 80054, Amiens Cedex 1, France
| | - Nacim Ammenouche
- Service d'Anesthésie et de Réanimation, Centre Hospitalier Universitaire Amiens-Picardie, Site Sud, 80054, Amiens Cedex 1, France
| | - Hervé Dupont
- Service d'Anesthésie et de Réanimation, Centre Hospitalier Universitaire Amiens-Picardie, Site Sud, 80054, Amiens Cedex 1, France
| | - Frédéric Marçon
- Service de Pharmacie, Centre Hospitalier Universitaire Amiens-Picardie, Site Sud, 80054, Amiens Cedex 1, France
| | - Bernard Allaouchiche
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France.,Faculté de Médecine Lyon Sud-Charles Mérieux, Université Claude Bernard Lyon 1, University of Lyon, 165 Rue du Petit Revoyet, 69600, Oullins, France
| | - Julien Bohé
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France.,Faculté de Médecine Lyon Sud-Charles Mérieux, Université Claude Bernard Lyon 1, University of Lyon, 165 Rue du Petit Revoyet, 69600, Oullins, France
| | - Alain Lepape
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France.,Laboratoire des Pathogènes Émergents, Université Claude Bernard Lyon 1, UMR CNRS 5308, Inserm U1111, Centre International de Recherche en Infectiologie, University of Lyon, 21 avenue Tony Garnier, 69365, Lyon cedex 07, France
| | - Sylvain Goutelle
- Groupement Hospitalier Nord, Service de Pharmacie, Hôpital Pierre Garraud, Hospices Civils de Lyon, 136 rue du Commandant Charcot, 69322, LyonCedex 5, France.,Laboratoire de Biométrie et Biologie Évolutive, Université Claude Bernard Lyon 1, UMR CNRS 5558, University of Lyon, Bât. Grégor Mendel, 43 bd du 11 novembre 1918, 69622, Villeurbanne Cedex, France.,ISPB - Faculté de Pharmacie de Lyon, Université Claude Bernard Lyon 1, University of Lyon, 8 Avenue Rockefeller, 69003, Lyon, France
| | - Arnaud Friggeri
- Groupement Hospitalier Sud, Service d'Anesthésie et de Réanimation, Hospices Civils de Lyon, 165 Chemin du Grand Revoyet, 69495, Pierre-Bénite, France.,Faculté de Médecine Lyon Sud-Charles Mérieux, Université Claude Bernard Lyon 1, University of Lyon, 165 Rue du Petit Revoyet, 69600, Oullins, France.,Laboratoire des Pathogènes Émergents, Université Claude Bernard Lyon 1, UMR CNRS 5308, Inserm U1111, Centre International de Recherche en Infectiologie, University of Lyon, 21 avenue Tony Garnier, 69365, Lyon cedex 07, France
| |
Collapse
|
37
|
Therapeutic Drug Monitoring of Gentamicin Peak Concentrations in Critically Ill Patients. Ther Drug Monit 2018; 39:522-530. [PMID: 28682925 DOI: 10.1097/ftd.0000000000000432] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Adequate gentamicin peak concentrations (Cmax) are important for optimal clinical efficacy. Within a critically ill patient, substantial variability in Cmax can occur over time, hampering the usefulness of therapeutic drug monitoring (TDM). The aim of this study was to evaluate the effect of gentamicin dosing based on Cmax after the first dose on gentamicin target attainment in critically ill patients. METHODS From gentamicin-treated critically ill patients, dosing information, clinical parameters, and serum concentrations were collected prospectively. A population pharmacokinetic model was developed using nonlinear mixed-effects modeling to estimate Cmax after each dose. To evaluate the usefulness of routine TDM, percentages of Cmax within (%Cther, 15-20 mg/L), above (>20 mg/L), and below (%Csubther, <15 mg/L) the therapeutic range after the first and second doses were compared. In addition, simulations were performed to evaluate the impact of TDM. RESULTS Four hundred sixteen measurements from 59 patients receiving 130 gentamicin doses were included. In the 30 patients who received >1 dose, TDM increased %Cther from 40% after a first median dose of 5.0 mg/kg to 50% after the second dose, and decreased %Csubther from 47% to 30%. Simulations using a 5 mg/kg starting dose revealed %Cther after the second dose of 28.4% without and 36.8% with TDM and %Csubther of 56.9% and 29.3%, respectively. Increasing the simulated starting dose to 6 mg/kg increased %Cther after the first dose from 27.7% to 33.5% and decreased %Csubther from 58.6% to 35.6%. TDM after a first dose of 6 mg/kg had no substantial effect on %Cther or %Csubther after the second dose. CONCLUSIONS Gentamicin dosing based on Cmax after the first dose increased %Cther and decreased %Csubther, but did not result in therapeutic Cmax in half of the patients. When simulating a higher starting dose, %Csubther after the first dose decreased, and TDM showed no additional influence. These data suggest that a starting dose of 6 mg/kg should be considered and that repeated Cmax measurements are not of added value.
Collapse
|
38
|
Bukkems LH, Roger C, Hodiamont CJ, Lefrant JY, Juffermans NP, Roberts JA, van Hest RM. Predictive performance of a gentamicin population pharmacokinetic model in two western populations of critically ill patients. Int J Antimicrob Agents 2018; 52:218-225. [PMID: 29704547 DOI: 10.1016/j.ijantimicag.2018.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/20/2018] [Accepted: 04/19/2018] [Indexed: 12/12/2022]
Abstract
External validation of population pharmacokinetic (PK) models is warranted before they can be clinically applied to aid in antibiotic dose selection. The primary objective of this study was to assess the predictive performance of a gentamicin population PK model in intensive care unit (ICU) patients in two independent western populations of critically ill patients. METHODS Data were collected from the ICU where the model was developed (Academic Medical Centre, Amsterdam [AMC]) and from the Centre Hospitalier Universitaire de Nîmes (CHU Nîmes). Primary endpoints were bias and accuracy. The model was regarded as valid if bias was not significantly different from 0 and accuracy was equal to or less than 2.5 mg/L. Non-linear mixed-effects modelling (NONMEM) was used for data analysis. RESULTS The AMC validation dataset consisted of 192 samples from 66 ICU patients and the CHU Nîmes dataset of 230 gentamicin samples from 50 ICU patients. The structural model predicted the gentamicin plasma concentrations in the AMC population with a non-significant bias (0.35, 95%CI: -0.11-0.81) and a sufficient accuracy of 2.5 mg/L (95%CI: 2.3-2.8). The gentamicin plasma concentrations were overpredicted in the CHU Nîmes population with a significant bias of 4.8 mg/L (95%CI: 4.00-5.62) and an accuracy of 5.5 mg/L (95%CI: 4.7-6.2). CONCLUSION The model is valid for use in the AMC ICU population but not in the CHU Nîmes ICU population. This illustrates that caution is needed when using a population PK model in an external population.
Collapse
Affiliation(s)
- Laura H Bukkems
- Department of Hospital Pharmacy & Clinical Pharmacology, Academic Medical Center, Amsterdam, The Netherlands
| | - Claire Roger
- Service des reanimations, Pôle Anesthésie Réanimation Douleur Urgence, CHU Nîmes, Place du Professeur Robert Debré, 3029 Nîmes Cedex 9, France; EA 2992, Faculté de Médicine Montpellier Nîmes, Avenue JF Kennedy, 30 000 Nîmes, France
| | - Caspar J Hodiamont
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jean-Yves Lefrant
- Service des reanimations, Pôle Anesthésie Réanimation Douleur Urgence, CHU Nîmes, Place du Professeur Robert Debré, 3029 Nîmes Cedex 9, France; EA 2992, Faculté de Médicine Montpellier Nîmes, Avenue JF Kennedy, 30 000 Nîmes, France
| | - Nicole P Juffermans
- Department of Intensive Care, Academic Medical Center, Amsterdam, The Netherlands
| | - Jason A Roberts
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Herston, Australia; Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Herston, Australia; Departments for Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Herston, Australia
| | - Reinier M van Hest
- Department of Hospital Pharmacy & Clinical Pharmacology, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Emergence of polymyxin B resistance in a polymyxin B-susceptible KPC-producing Klebsiella pneumoniae causing bloodstream infection in a neutropenic patient during polymyxin B therapy. Diagn Microbiol Infect Dis 2017; 90:134-138. [PMID: 29150371 DOI: 10.1016/j.diagmicrobio.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/25/2017] [Accepted: 10/13/2017] [Indexed: 01/27/2023]
Abstract
The emergence of resistance to polymyxins in KPC-producing Klebsiella pneumoniae isolates has been a major clinical problem. This study evaluated the molecular mechanisms associated with polymyxin B (PMB) resistance that emerged in a previously PMB-susceptible KPC-2-producing K. pneumoniae during PMB therapy for a bloodstream infection in a neutropenic patient. The first isolate (PMB-susceptible) was obtained while the patient was receiving meropenem and other isolates were recovered from 2 sets of blood cultures in different dates while the patient was receiving PMB therapy (4 of 6 blood cultures bottles yielded isolates with full PMB resistance). The population analysis profile of the first isolate revealed the growth of resistant subpopulations with PFGE profile distinct from the parental isolate but undistinguishable from those obtained in subsequent days under PMB exposure. Resistant subpopulations were obtained from all parental PMB-susceptible and in one PMB-resistant isolate recovered from the patient. The molecular mechanism observed in the hetero-resistant subpopulations (IS1-like in mgrB-promoter region, increased rstB transcription with no mutation and non-identified mechanism) differed from those found in the PMB-resistant isolates, in which no mutation or transcriptional alterations were detected. This study showed that the mechanism of resistance to PMB that emerged during PMB therapy was not related to those observed in subpopulations selected in vitro from PMB-susceptible isolates recovered from the patient. The absence of mutations in the former isolates may be due to adaptive resistance occurred because of sub-optimal PMB levels as well as amikacin and meropenem used in combination.
Collapse
|
40
|
Llanos-Paez CC, Hennig S, Staatz CE. Population pharmacokinetic modelling, Monte Carlo simulation and semi-mechanistic pharmacodynamic modelling as tools to personalize gentamicin therapy. J Antimicrob Chemother 2017; 72:639-667. [PMID: 28062683 DOI: 10.1093/jac/dkw461] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Population pharmacokinetic modelling, Monte Carlo simulation and semi-mechanistic pharmacodynamic modelling are all tools that can be applied to personalize gentamicin therapy. This review summarizes and evaluates literature knowledge on the population pharmacokinetics and pharmacodynamics of gentamicin and identifies areas where further research is required to successfully individualize gentamicin therapy using modelling and simulation techniques. Thirty-five studies have developed a population pharmacokinetic model of gentamicin and 15 studies have made dosing recommendations based on Monte Carlo simulation. Variability in gentamicin clearance was most commonly related to renal function in adults and body weight and age in paediatrics. Nine studies have related aminoglycoside exposure indices to clinical outcomes. Most commonly, efficacy has been linked to a Cmax/MIC ≥7-10 and a AUC24/MIC ≥70-100. No study to date has shown a relationship between predicted achievement of exposure targets and actual clinical success. Five studies have developed a semi-mechanistic pharmacokinetic/pharmacodynamic model to predict bacteria killing and regrowth following gentamicin exposure and one study has developed a deterministic model of aminoglycoside nephrotoxicity. More complex semi-mechanistic models are required that consider the immune response, use of multiple antibiotics, the severity of illness, and both efficacy and toxicity. As our understanding grows, dosing of gentamicin based on sound pharmacokinetic/pharmacodynamic principles should be applied more commonly in clinical practice.
Collapse
|
41
|
Pitiriga V, Dimitroulia E, Saroglou G, Tsakris A. The challenge of curbing aminoglycoside resistance: can antimicrobial stewardship programs play a critical role? Expert Rev Anti Infect Ther 2017; 15:947-954. [DOI: 10.1080/14787210.2017.1382355] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Vassiliki Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Dimitroulia
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George Saroglou
- Department of Internal Medicine, Metropolitan General Hospital, Piraeus, Greece
| | - Athanassios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
42
|
Population Pharmacokinetic Characteristics of Amikacin in Suspected Cases of Neonatal Sepsis in a Low-Resource African Setting: A Prospective Nonrandomized Single-Site Study. Curr Ther Res Clin Exp 2017; 84:e1-e6. [PMID: 28761582 PMCID: PMC5522970 DOI: 10.1016/j.curtheres.2017.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Amikacin exhibits marked pharmacokinetic (PK) variability and is commonly used in combination with other drugs in the treatment of neonatal sepsis. There is a paucity of amikacin PK information in neonates from low-resource settings. OBJECTIVES To determine the PK parameters of amikacin, and explore the influence of selected covariates, including coadministration with aminophylline, on amikacin disposition in neonates of African origin. METHODS Neonates with suspected sepsis admitted to an intensive care unit in Accra, Ghana, and treated with amikacin (15 mg/kg loading followed by 7.5 mg/kg every 12 hours), were recruited. Serum amikacin concentration was measured at specified times after treatment initiation and analyzed using a population PK modeling approach. RESULTS A total of 419 serum concentrations were available for 247 neonates. Mean (SD) trough amikacin concentration (from samples collected 30 minutes before the fourth dose) among term (n = 25), and preterm (<37 weeks' gestation n = 36) neonates were 6.2 (3.4) and 9.2 (5.7) µg/mL, respectively (P = 0.02). A 1-compartment model best fitted amikacin disposition, and birth weight was the most important predictor of amikacin clearance (CL) and distribution (V). The population CL and V of amikacin were related as CL (L/h) = 0.153 (birth weight/2.5)1.31, V (L) = 2.94 (birth weight/2.5)1.18. There was a high between-subject variability (58.9% and 50.7%) in CL and V, respectively. CL and V were 0.058 L/h/kg and 1.15 L/kg, respectively, for a mean birth weight of 2.1 kg, and the mean half-life (based on 1-compartment model), was 13.7 hours. CONCLUSIONS The V and half-life of amikacin in this cohort varied from that reported in non-African populations, and the high trough and low peak amikacin concentrations in both term and preterm neonates suggest strategies to optimize amikacin dosing are required in this population.
Collapse
|
43
|
Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit Care Med 2017; 45:486-552. [PMID: 28098591 DOI: 10.1097/ccm.0000000000002255] [Citation(s) in RCA: 1974] [Impact Index Per Article: 246.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To provide an update to "Surviving Sepsis Campaign Guidelines for Management of Sepsis and Septic Shock: 2012." DESIGN A consensus committee of 55 international experts representing 25 international organizations was convened. Nominal groups were assembled at key international meetings (for those committee members attending the conference). A formal conflict-of-interest (COI) policy was developed at the onset of the process and enforced throughout. A stand-alone meeting was held for all panel members in December 2015. Teleconferences and electronic-based discussion among subgroups and among the entire committee served as an integral part of the development. METHODS The panel consisted of five sections: hemodynamics, infection, adjunctive therapies, metabolic, and ventilation. Population, intervention, comparison, and outcomes (PICO) questions were reviewed and updated as needed, and evidence profiles were generated. Each subgroup generated a list of questions, searched for best available evidence, and then followed the principles of the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system to assess the quality of evidence from high to very low, and to formulate recommendations as strong or weak, or best practice statement when applicable. RESULTS The Surviving Sepsis Guideline panel provided 93 statements on early management and resuscitation of patients with sepsis or septic shock. Overall, 32 were strong recommendations, 39 were weak recommendations, and 18 were best-practice statements. No recommendation was provided for four questions. CONCLUSIONS Substantial agreement exists among a large cohort of international experts regarding many strong recommendations for the best care of patients with sepsis. Although a significant number of aspects of care have relatively weak support, evidence-based recommendations regarding the acute management of sepsis and septic shock are the foundation of improved outcomes for these critically ill patients with high mortality.
Collapse
|
44
|
Zavascki AP, Klee BO, Bulitta JB. Aminoglycosides against carbapenem-resistant Enterobacteriaceae in the critically ill: the pitfalls of aminoglycoside susceptibility. Expert Rev Anti Infect Ther 2017; 15:519-526. [PMID: 28375030 DOI: 10.1080/14787210.2017.1316193] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The emergence of carbapenem-resistant Enterobacteriaceae (CRE) has brought aminoglycosides to the frontline since an aminoglycoside may be the only antimicrobial to which CRE isolates show in vitro susceptibility. The appropriateness of aminoglycoside-based therapies for severe infections by CRE is discussed considering the current breakpoints and recent pharmacokinetic (PK) studies in critically ill patients. Areas covered: Many aminoglycoside-susceptible CRE isolates present minimal inhibitory concentrations (MICs) at or slightly below the breakpoint of amikacin or gentamicin. However, recent PK studies with these aminoglycosides in critically ill have invariably shown that the PK/pharmacodynamic (PD) target is very unlikely attained even when high doses are administered, if the MICs are near the breakpoint. Expert commentary: While new antimicrobials are not widely available, the authors forecast an increasing use of aminoglycosides as backbone antibiotics against CRE isolates. However, the altered PK of aminoglycosides in critically ill patients severely impairs their predicted efficacy in these patients. Aminoglycoside breakpoints may hide 'aminoglycoside-susceptible' CRE isolates for that aminoglycosides will unlikely be effective if used in monotherapy. Therefore, these breakpoints may need to be revised due to the increasing use of aminoglycosides as backbone antibiotics to treat severe infections by CRE isolates in critically ill patients.
Collapse
Affiliation(s)
- Alexandre P Zavascki
- a Infectious Diseases Service , Hospital de Clinicas de Porto Alegre , Porto Alegre , Brazil.,b Department of Internal Medicine, Medical School , Universidade Federal do Rio Grande do Sul , Porto Alegre , Brazil
| | - Brandon O Klee
- c Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics , College of Pharmacy, University of Florida , Orlando , FL , USA
| | - Jürgen B Bulitta
- c Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics , College of Pharmacy, University of Florida , Orlando , FL , USA
| |
Collapse
|
45
|
Cooper TW, Pass SE, Brouse SD, Hall RG. Can Pharmacokinetic and Pharmacodynamic Principles Be Applied to the Treatment of Multidrug-Resistant Acinetobacter? Ann Pharmacother 2017; 45:229-40. [DOI: 10.1345/aph.1p187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE: To discuss treatment options that can be used for treatment of Acinetobacter infections. DATA SOURCES: A MEDLINE search (1966-November 2010) was conducted to identify English-language literature on pharmacotherapy of Acinetobacter and the bibliographies of pertinent articles. Programs and abstracts from infectious diseases meetings were also searched. Search terms included Acinetobacter, multidrug resistance, pharmacokinetics, pharmacodynamics, Monte Carlo simulation, nosocomial pneumonia, carbapenems, polymyxins, sulbactam, aminoglycosides, tetracyclines, tigecycline, rifampin, and fluoroquinolones. DATA SELECTION AND DATA EXTRACTION: All articles were critically evaluated and all pertinent information was included in this review. DATA SYNTHESIS: Multidrug resistant (MDR) Acinetobacter, defined as resistance to 3 or more antimicrobial classes, has increased over the past decade. The incidence of carbapenem-resistant Acinetobacter is also increasing, leading to an increased use of dose optimization techniques and/or alternative antimicrobials, which is driven by local susceptibility patterns. However, Acinetobacter infections that are resistant to all commercially available antibiotics have been reported. General principles are available to guide dose optimization of aminoglycosides, β-lactams, fluoroquinolones, and tigecycline for infections due to gram-negative pathogens. Unfortunately, data specific to patients with Acinetobacter infections are limited. Recent pharmacokinetic-pharmacodynamic information has shed light on colistin dosing. The dilemma with colistin is its concentration-dependent killing, which makes once-daily dosing seem like an attractive option, but its short postantibiotic effect limits a clinician's ability to extend the dosing interval. Localized delivery of antimicrobials is also an attractive option due to the ability to increase drug concentration at the infection site while minimizing systemic adverse events, but more data are needed regarding this approach. CONCLUSIONS: Increased reliance on dosage optimization, combination therapy, and localized delivery of antimicrobials are methods to pursue positive clinical outcomes in MDR Acinetobacter infections since novel antimicrobials will not be available for several years. Well-designed clinical trials with MDR Acinetobacter are needed to define the best treatment options for these patients.
Collapse
|
46
|
Reduced Chance of Hearing Loss Associated with Therapeutic Drug Monitoring of Aminoglycosides in the Treatment of Multidrug-Resistant Tuberculosis. Antimicrob Agents Chemother 2017; 61:AAC.01400-16. [PMID: 28069654 DOI: 10.1128/aac.01400-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/04/2016] [Indexed: 11/20/2022] Open
Abstract
Hearing loss and nephrotoxicity are associated with prolonged treatment duration and higher dosage of amikacin and kanamycin. In our tuberculosis center, we used therapeutic drug monitoring (TDM) targeting preset pharmacokinetic/pharmacodynamic (PK/PD) surrogate endpoints in an attempt to maintain efficacy while preventing (oto)toxicity. To evaluate this strategy, we retrospectively evaluated medical charts of tuberculosis (TB) patients treated with amikacin or kanamycin in the period from 2000 to 2012. Patients with culture-confirmed multiresistant or extensively drug-resistant tuberculosis (MDR/XDR-TB) receiving amikacin or kanamycin as part of their TB treatment for at least 3 days were eligible for inclusion in this retrospective study. Clinical data, including maximum concentration (Cmax), Cmin, and audiometry data, were extracted from the patients' medical charts. A total of 80 patients met the inclusion criteria. The mean weighted Cmax/MIC ratios obtained from 57 patients were 31.2 for amikacin and 12.3 for kanamycin. The extent of hearing loss was limited and correlated with the cumulative drug dose per kg of body weight during daily administration. At follow-up, 35 (67.3%) of all patients had successful outcome; there were no relapses. At a median dose of 6.5 mg/kg, a correlation was found between the dose per kg of body weight during daily dosing and the extent of hearing loss in dB at 8,000 Hz. These findings suggest that the efficacy at this lower dosage is maintained with limited toxicity. A randomized controlled trial should provide final proof of the safety and efficacy of TDM-guided use of aminoglycosides in MDR-TB treatment.
Collapse
|
47
|
Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med 2017; 43:304-377. [PMID: 28101605 DOI: 10.1007/s00134-017-4683-6] [Citation(s) in RCA: 3970] [Impact Index Per Article: 496.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 01/06/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To provide an update to "Surviving Sepsis Campaign Guidelines for Management of Sepsis and Septic Shock: 2012". DESIGN A consensus committee of 55 international experts representing 25 international organizations was convened. Nominal groups were assembled at key international meetings (for those committee members attending the conference). A formal conflict-of-interest (COI) policy was developed at the onset of the process and enforced throughout. A stand-alone meeting was held for all panel members in December 2015. Teleconferences and electronic-based discussion among subgroups and among the entire committee served as an integral part of the development. METHODS The panel consisted of five sections: hemodynamics, infection, adjunctive therapies, metabolic, and ventilation. Population, intervention, comparison, and outcomes (PICO) questions were reviewed and updated as needed, and evidence profiles were generated. Each subgroup generated a list of questions, searched for best available evidence, and then followed the principles of the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system to assess the quality of evidence from high to very low, and to formulate recommendations as strong or weak, or best practice statement when applicable. RESULTS The Surviving Sepsis Guideline panel provided 93 statements on early management and resuscitation of patients with sepsis or septic shock. Overall, 32 were strong recommendations, 39 were weak recommendations, and 18 were best-practice statements. No recommendation was provided for four questions. CONCLUSIONS Substantial agreement exists among a large cohort of international experts regarding many strong recommendations for the best care of patients with sepsis. Although a significant number of aspects of care have relatively weak support, evidence-based recommendations regarding the acute management of sepsis and septic shock are the foundation of improved outcomes for these critically ill patients with high mortality.
Collapse
|
48
|
Determinants of gentamicin concentrations in critically ill patients: a population pharmacokinetic analysis. Int J Antimicrob Agents 2016; 49:204-211. [PMID: 28038961 DOI: 10.1016/j.ijantimicag.2016.10.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/20/2016] [Accepted: 10/30/2016] [Indexed: 11/23/2022]
Abstract
When treating critically ill patients with gentamicin for severe infection, peak concentrations (Cmax) determine clinical efficacy and trough concentrations (Cmin) determine toxicity. Despite administration of body weight-standardised starting doses, a wide range of Cmax is generally observed. Furthermore, in therapeutic drug monitoring, several measures of renal function are used to predict appropriate Cmin and gentamicin dosing intervals, but the most accurate predictor is not known. This study aimed to quantify the impact of several patient parameters on gentamicin Cmax values and to determine which measure of renal function best predicts gentamicin clearance (CL). Clinical data and serum gentamicin levels were retrospectively collected from all critically ill patients treated with gentamicin at our intensive care unit between 1 January and 30 June 2011. Data were analysed using non-linear mixed-effects modelling (NONMEM v.7.1.2). A two-compartmental model was developed based on 303 gentamicin concentration-time data from 44 critically ill patients. Serum albumin levels explained 25% of interindividual variability in the volume of distribution (Vd). Creatinine clearance calculated from the creatinine concentration in a 6-h urine portion (CalcCLCr) resulted in acceptable estimation of gentamicin CL, whilst serum creatinine (SCr) and creatinine clearance estimated by the Cockcroft-Gault formula (CGCLCr) overestimated gentamicin CL and therefore underestimated Cmin. In conclusion, low albumin concentrations resulted in a larger Vd and lower Cmax of gentamicin. These results suggest that use of a higher gentamicin starting dose in critically ill patients with hypoalbuminaemia may prevent underdosing. Urinary CalcCLCr is a better predictor of Cmin than SCr or CGCLCr.
Collapse
|
49
|
Onufrak NJ, Forrest A, Gonzalez D. Pharmacokinetic and Pharmacodynamic Principles of Anti-infective Dosing. Clin Ther 2016; 38:1930-47. [PMID: 27449411 PMCID: PMC5039113 DOI: 10.1016/j.clinthera.2016.06.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/09/2016] [Accepted: 06/23/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE An understanding of the pharmacokinetic (PK) and pharmacodynamic (PD) principles that determine response to antimicrobial therapy can provide the clinician with better-informed dosing regimens. Factors influential on antibiotic disposition and clinical outcome are presented, with a focus on the primary site of infection. Techniques to better understand antibiotic PK and optimize PD are acknowledged. METHODS PubMed (inception-April 2016) was reviewed for relevant publications assessing antimicrobial exposures within different anatomic locations and clinical outcomes for various infection sites. FINDINGS A limited literature base indicates variable penetration of antibiotics to different target sites of infection, with drug solubility and extent of protein binding providing significant PK influences in addition to the major clearing pathway of the agent. PD indices derived from in vitro studies and animal models determine the optimal magnitude and frequency of dosing regimens for patients. PK/PD modeling and simulation has been shown an efficient means of assessing these PD endpoints against a variety of PK determinants, clarifying the unique effects of infection site and patient characteristics to inform the adequacy of a given antibiotic regimen. IMPLICATIONS Appreciation of the PK properties of an antibiotic and its PD measure of efficacy can maximize the utility of these life-saving drugs. Unfortunately, clinical data remain limited for a number of infection site-antibiotic exposure relationships. Modeling and simulation can bridge preclinical and patient data for the prescription of optimal antibiotic dosing regimens, consistent with the tenets of personalized medicine.
Collapse
Affiliation(s)
- Nikolas J Onufrak
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alan Forrest
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
50
|
Allou N, Allyn J, Levy Y, Bouteau A, Caujolle M, Delmas B, Valance D, Brulliard C, Martinet O, Vandroux D, Montravers P, Augustin P. Assessment of the National French recommendations regarding the dosing regimen of 8mg/kg of gentamicin in patients hospitalised in intensive care units. Anaesth Crit Care Pain Med 2016; 35:331-335. [PMID: 27133236 DOI: 10.1016/j.accpm.2015.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/27/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
INTRODUCTION To assess the French National Agency for Medicines and Health Products Safety (ANSM) guidelines concerning the peak plasma concentration (Cmax) of gentamicin when using a loading dose of 8mg/kg administered in patients hospitalised in the intensive care unit (ICU). PATIENTS AND METHODS A prospective observational cohort study conducted in one ICU. RESULTS During the study period, 34 patients with a median simplified acute physiology score 2 of 54 [44-70] received a median dose of 8 [7.9-8.1] mg/kg of gentamicin. The median Cmax was 17.5 [15.4-20.7] mg/L and no patient had a Cmax>30mg/L. Twenty-four of 34 patients (71%) had a Cmax>16mg/L. Following multivariate analysis, the only factor associated with Cmax<16mg/L was a positive fluid balance 24hours before gentamicin administration (per 1000mL increment) (OR: 0.37, 95% CI: 0.18-0.77, P=0.008). CONCLUSIONS These results suggest that a Cmax>30mg/L [which corresponds to approximately 8 times the minimal inhibiting concentrations (MIC) breakpoints for Pseudomonas aeruginosa and Enterobacteriaceae with intermediate sensitivity] of gentamicin as recommended by ANSM guidelines seems impossible to obtain with a loading dose of 8mg/kg in the ICU. A loading dose of 8mg/kg should probably not be used in the empiric antibiotic treatment of infection due to non-fermenting Gram-negative bacilli and Enterobacteriaceae with intermediate sensitivity whose MIC breakpoint is 4mg/L. A Cmax>16mg/L was not reached in almost 30% of patients, particularly in the group with a positive fluid balance who require higher doses than currently recommended.
Collapse
Affiliation(s)
- Nicolas Allou
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Jérôme Allyn
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Yaël Levy
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Astrid Bouteau
- Département d'Anesthésie-Réanimation, AP-HP, CHU Bichat-Claude-Bernard, 46, rue Henri-Huchard, 75018 Paris, France.
| | - Marie Caujolle
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Benjamin Delmas
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Dorothée Valance
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Caroline Brulliard
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Olivier Martinet
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - David Vandroux
- Réanimation polyvalente, CHU Felix-Guyon, allée des Topazes, 97405 Saint-Denis, France.
| | - Philippe Montravers
- Département d'Anesthésie-Réanimation, AP-HP, CHU Bichat-Claude-Bernard, 46, rue Henri-Huchard, 75018 Paris, France.
| | - Pascal Augustin
- Département d'Anesthésie-Réanimation, AP-HP, CHU Bichat-Claude-Bernard, 46, rue Henri-Huchard, 75018 Paris, France.
| |
Collapse
|