1
|
Briand A, Bernier L, Pincivy A, Roumeliotis N, Autmizguine J, Marsot A, Métras MÉ, Thibault C. Prolonged Beta-Lactam Infusions in Children: A Systematic Review and Meta-Analysis. J Pediatr 2024; 275:114220. [PMID: 39097265 DOI: 10.1016/j.jpeds.2024.114220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/02/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE To assess whether beta-lactam extended or continuous beta-lactam infusions (EI/CI) improve clinical outcomes in children with proven or suspected bacterial infections. STUDY DESIGN We included observational and interventional studies that compared beta-lactam EI or CI with standard infusions in children less than 18 years old, and reported on mortality, hospital or intensive care unit length of stay, microbiological cure, and/or clinical cure. Data sources included PubMed, Medline, EBM Reviews, EMBASE, and CINAHL and were searched from January 1, 1980, to November 3, 2023. Thirteen studies (2945 patients) were included: 5 randomized control trials and 8 observational studies. Indications for antimicrobial therapies and clinical severity varied, ranging from cystic fibrosis exacerbation to critically ill children with bacteriemia. RESULTS EI and CI were not associated with a reduction in mortality in randomized control trials (n = 1464; RR 0.93, 95% CI 0.71, 1.21), but were in observational studies (n = 833; RR 0.43, 95% CI 0.19, 0.96). We found no difference in hospital length of stay. Results for clinical and microbiological cures were heterogeneous and reported as narrative review. The included studies were highly heterogeneous, limiting the strength of our findings. The lack of shared definitions for clinical and microbiological cure outcomes precluded analysis. CONCLUSIONS EI and CI were not consistently associated with reduced mortality or length of stay in children. Results were conflicting regarding clinical and microbiological cures. More well-designed studies targeting high-risk populations are necessary to determine the efficacy of these alternative dosing strategies.
Collapse
Affiliation(s)
- Annabelle Briand
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; Department of Pediatrics, CHU Sainte-Justine, Montreal QC, Canada
| | - Laurie Bernier
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Alix Pincivy
- Library Services, CHU Sainte-Justine, Montreal, QC, Canada
| | - Nadia Roumeliotis
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; Division of Critical Care Medicine, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, Canada; CHU Sainte Justine Research Center, Montreal, QC, Canada
| | - Julie Autmizguine
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; CHU Sainte Justine Research Center, Montreal, QC, Canada; Department of Pharmacology and Physiology, Université de Montréal, CHU Sainte-Justine, Montreal, QC, Canada; Division of Infectious Diseases, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, Canada
| | - Amélie Marsot
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada; CHU Sainte Justine Research Center, Montreal, QC, Canada
| | - Marie-Élaine Métras
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada; Division of Critical Care Medicine, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, Canada; CHU Sainte Justine Research Center, Montreal, QC, Canada
| | - Celine Thibault
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; Division of Critical Care Medicine, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, Canada; CHU Sainte Justine Research Center, Montreal, QC, Canada.
| |
Collapse
|
2
|
Wu J, Wang C, Zhang R, Du P, Wang Y, Wu P, Chen X, Huang Y, Jia Y, Shen J. SIL-IS LC-ESI-MS/MS method for simultaneous quick detection of amoxicillin and clavulanic acid in human plasma: Development, validation and its application to a pharmacokinetics study. Biomed Chromatogr 2024; 38:e5964. [PMID: 39252549 DOI: 10.1002/bmc.5964] [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: 04/17/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 09/11/2024]
Abstract
A liquid chromatography electrospray ionization tandem mass spectrometry method with amoxicillin-d4 as the stable isotope-labeled internal standard for simultaneous quick detection of amoxicillin and clavulanic acid in human plasma was developed and validated. Chromatographic separations were performed on a Hedera ODS-2 column (2.1 × 150 mm, 5 μm). The mobile phases for gradient elution were aqueous solution containing 0.2% acetic acid (AA) (mobile phase A) together with organic phase solution (acetonitrile and methanol mixed solution, mobile phase B). Mass spectrometry was performed using negative electrospray ionization in multiple reaction monitoring mode. The target fragment ion pairs of amoxicillin, clavulanic acid and amoxicillin-d4 were m/z 364.1 → 223.1, 198.1 → 135.9 and 368.1 → 227.1, respectively. The linear ranges of this method were 40-5,000 ng/ml for amoxicillin and 30-2,500 ng/ml for clavulanic acid, with coefficient of determination > 0.9900. This method validation included selectivity, standard curve, lower limit of quantitation, accuracy, precision, recovery, matrix effect (hemolytic matrix and hyperlipidemic matrix), carryover, stability, dilution reliability and incurred sample reanalysis study. A successful application of this method was realized in a pharmacokinetic study after administration of amoxicillin-clavulanic acid potassium granules.
Collapse
Affiliation(s)
- Jianbang Wu
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Changmao Wang
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
- The People's Hospital of Lezhi, Ziyang, Sichuan, People's Republic of China
| | - Rong Zhang
- Hainan Simcere Pharmaceutical Co., Ltd., People's Republic of China
| | - Pengfei Du
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Yaqin Wang
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
| | - Ping Wu
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
| | - Xinyan Chen
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Yunzhe Huang
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Yuanwei Jia
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
| | - Jie Shen
- Anhui Provincial Center of Drug Clinical Evaluation, Yijishan Hospital of WannanMedical College, Wuhu, Anhui, People's Republic of China
- School of pharmacy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| |
Collapse
|
3
|
Heffernan AJ, Roberts JA. Is it a case of higher, the worse, or are beta-lactam antibiotics the innocent bystanders? J Crit Care 2024; 85:154934. [PMID: 39490227 DOI: 10.1016/j.jcrc.2024.154934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Aaron J Heffernan
- University of Queensland Centre for Clinical Research, Faculty of Medicine The University of Queensland, Brisbane, Australia
| | - Jason A Roberts
- University of Queensland Centre for Clinical Research, Faculty of Medicine The University of Queensland, Brisbane, Australia; Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, Australia; Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia; UR UM 103, University of Montpellier, Division of Anesthesia Critical Care and Emergency and Pain Medicine, Nimes University Hospital, Nimes, France.
| |
Collapse
|
4
|
Weber L, Moerer O, Wieditz J, Winkler MS, Scheithauer S, Stephani C. A retrospective cohort analysis of factors influencing continuous antibiotic therapy with ampicillin. Life Sci 2024; 358:123168. [PMID: 39454996 DOI: 10.1016/j.lfs.2024.123168] [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: 05/12/2024] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
AIMS There are limited data on ampicillin/sulbactam, both for continuous infusion and for use in critically ill patients. We aimed to identify factors that help predict ampicillin plasma levels during continuous antibiotic therapy in intensive care patients. MAIN METHODS We retrospectively reviewed and retrieved a large dataset of patients who received continuous ampicillin infusion with therapeutic drug monitoring between 2015 and 2022. Patients initially received standard dosing (single shot of 2/1 g followed by continuous infusion of 6/3 g ampicillin/sulbactam per day), which was then adjusted based on the results of regular therapeutic drug monitoring and according to a target range of 30-60 mg/l (equivalent to four to eight times the minimum inhibitory concentration of ampicillin for Enterobacterales). MAIN RESULTS 466 measurements from 225 patients (152 male, mean age 61 years) were analyzed. Initial measurements of ampicillin plasma levels were below the predefined optimal therapeutic range in 50 %, within the range in 30 % and above the range in 20 %. Target attainment increased to 70 % by the 4th measurement. There was a significant negative correlation between ampicillin plasma levels and estimated glomerular filtration rate (eGFR) (r = -0.74; p < 0.001) and, to a lesser extent, with height (r = -0.31; p < 0.001). Based on multiple linear regression, eGFR and body weight or height were the factors accounting for 63 % of the variability in the data. SIGNIFICANCE To optimise target achievement, ampicillin dosing in critically ill patients requires a personalized approach based on renal function. Importantly, patients with normal or augmented eGFR require higher standard doses of ampicillin/sulbactam.
Collapse
Affiliation(s)
- L Weber
- Emergency Department, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - O Moerer
- Clinic for Anesthesiology, University Medical Center Göttingen, Robert Koch-Straße 40, 37075 Göttingen, Germany
| | - J Wieditz
- Clinic for Anesthesiology, University Medical Center Göttingen, Robert Koch-Straße 40, 37075 Göttingen, Germany; Department of Medical Statistics, University Medical Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany
| | - M S Winkler
- Clinic for Anesthesiology, University Medical Center Göttingen, Robert Koch-Straße 40, 37075 Göttingen, Germany
| | - S Scheithauer
- Department of Infection Control and Infectious Diseases, University Medical Center Göttingen, Robert Koch-Straße 40, 37075 Göttingen, Germany
| | - C Stephani
- Clinic for Anesthesiology, University Medical Center Göttingen, Robert Koch-Straße 40, 37075 Göttingen, Germany.
| |
Collapse
|
5
|
O'Jeanson A, Nielsen EI, Friberg LE. Therapeutic drug monitoring (TDM) of β-lactam/β-lactamase inhibitor (BL/BLI) drug combinations: insights from a pharmacometric simulation study. J Antimicrob Chemother 2024:dkae375. [PMID: 39436757 DOI: 10.1093/jac/dkae375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The emergence of β-lactamase-producing bacteria has led to the use of β-lactam (BL) antibiotic and β-lactamase inhibitor (BLI) drug combinations. Despite therapeutic drug monitoring (TDM) being endorsed for BLs, the impact of TDM on BLIs remains unclear. OBJECTIVES Evaluate whether BLIs are available in effective exposures at the site of infection and assess if TDM of BLIs could be of interest. METHODS Population pharmacokinetic models for 9 BL and BLI compounds were used to simulate drug concentrations at infection sites following EMA-approved dose regimens, considering plasma protein binding and tissue penetration. Predicted target site concentrations were used for probability of target attainment (PTA) analysis. RESULTS Using EUCAST targets, satisfactory (≥90%) PTA was observed for BLs in patients with typical renal clearance (CrCL of 80 mL/min) across various sites of infection. However, results varied for BLIs. Avibactam achieved satisfactory PTA only in plasma, with reduced PTAs in abdomen (78%), lung (73%) and prostate (23%). Similarly, tazobactam resulted in unsatisfactory PTAs in intra-abdominal infections (79%), urinary tract infections (64%) and prostatitis (34%). Imipenem-relebactam and meropenem-vaborbactam achieved overall satisfactory PTAs, except in prostatitis and high-MIC infections for the latter combination. CONCLUSIONS This study highlights the risk of solely relying on TDM of BLs, as this can indicate acceptable exposures of the BL while the BLI concentration, and consequently the combination, can result in suboptimal performance in terms of bacterial killing. Thus, dose adjustments also based on plasma concentration measurements of BLIs, in particular for avibactam and tazobactam, can be valuable in clinical practice to obtain effective exposures at the target site.
Collapse
Affiliation(s)
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Laporte-Amargos J, Carmona-Torre F, Huguet M, Puerta-Alcalde P, Rigo-Bonnin R, Ulldemolins M, Arnan M, Del Pozo JL, Torrent A, Garcia-Vidal C, Pallarès N, Tebé C, Muñoz C, Tubau F, Padullés A, Sureda AM, Carratalà J, Gudiol C. Efficacy of extended infusion of β-lactam antibiotics for the treatment of febrile neutropenia in haematologic patients (BEATLE): a randomised, multicentre, open-label, superiority clinical trial. Clin Microbiol Infect 2024:S1198-743X(24)00488-9. [PMID: 39433124 DOI: 10.1016/j.cmi.2024.10.006] [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: 07/25/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVES The efficacy of extended infusions (EI) of β-lactam antibiotics for optimising outcomes in febrile neutropenia is unclear. We assessed whether the administration of β-lactams was more effective in EI than in intermittent infusion (II) for the treatment of febrile neutropenia. METHODS We performed a randomised, open-label, superiority clinical trial of patients with febrile neutropenia at four Spanish university hospitals. Patients undergoing haematopoietic stem cell transplantation or with acute leukaemia receiving chemotherapy who required empirical antibiotic treatment for febrile neutropenia were randomly assigned (1:1) to receive EI of β-lactam or II after a first dose in bolus. The choice of antipseudomonal β-lactam was left to the discretion of the attending physician. The primary endpoint was treatment success at day 5, defined as defervescence without modifying the antibiotic treatment. Secondary endpoints included adverse events, attainment of the pharmacokinetic/pharmacodynamic target of 50, 75 and 100%ƒuT>MIC, and 30-day mortality. RESULTS Between November 19, 2019 and June 22, 2022, 295 patients we screened for eligibility, of whom 150 were randomly assigned to receive EI (n=77) or II (n=73) of the antipseudomonal β-lactam of choice. In the intention-to-treat analysis, treatment success at day 5 was achieved in 39/77 patients (50.6%) receiving EI versus 46/73 patients (63.0%) receiving II (risk difference -12.4%; 95% confidence interval, -29.4 to 4.7, p=0.17). The pharmacokinetic/pharmacodynamic targets of 75 and 100% ƒuT>MIC for empirical treatment were achieved more frequently in the EI group. No statistically-significant differences were found between groups in terms of adverse events or 30-day mortality. CONCLUSIONS Our findings do not support the routine use of empirical EI of β-lactams in febrile neutropenia. Further studies should consider the clinical heterogeneity of febrile neutropenia and focus on patients with sepsis/septic shock and microbiologically documented infections, particularly those with infections caused by microorganisms less susceptible to β-lactams.
Collapse
Affiliation(s)
- Julia Laporte-Amargos
- Department of Infectious Diseases, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Francisco Carmona-Torre
- Department of Infectious Diseases, Clínica Universidad de Navarra, Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maria Huguet
- Department of Clinical Haematology, Institut Català d'Oncologia-Badalona, Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Pedro Puerta-Alcalde
- Infectious Diseases Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raul Rigo-Bonnin
- Department of Clinical Laboratory, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marta Ulldemolins
- Department of Infectious Diseases, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Montserrat Arnan
- Department of Clinical Haematology, Institut Català d'Oncologia-Hospitalet, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Jose Luis Del Pozo
- Department of Infectious Diseases, Clínica Universidad de Navarra, Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Department of Clinical Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Anna Torrent
- Department of Clinical Haematology, Institut Català d'Oncologia-Badalona, Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Carolina Garcia-Vidal
- Infectious Diseases Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Natàlia Pallarès
- University of Barcelona, Barcelona, Spain; Biostatistics Unit, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristian Tebé
- Biostatistics Unit, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carme Muñoz
- Department of Pharmacy, Institut Català d'Oncologia-Hospitalet, IDIBELL, Barcelona, Spain
| | - Fe Tubau
- Department of Microbiology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Ariadna Padullés
- Department of Pharmacy, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana-Maria Sureda
- Department of Clinical Haematology, Institut Català d'Oncologia-Hospitalet, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Institut Català d'Oncologia, Barcelona, Spain
| |
Collapse
|
7
|
Rentsch KM, Khanna N, Halbeisen D, Osthoff M. Enhancing Stability and Investigating Target Attainment of Benzylpenicillin in Outpatient Parenteral Antimicrobial Therapy: Insights from In Vitro and In Vivo Evaluations. Antibiotics (Basel) 2024; 13:970. [PMID: 39452236 PMCID: PMC11504374 DOI: 10.3390/antibiotics13100970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objective: Narrow-spectrum beta-lactam antibiotics such as benzylpenicillin and flucloxacillin are increasingly used in outpatient parenteral antimicrobial therapy (OPAT) programs to mitigate the adverse effects associated with broad-spectrum antibiotics. These beta-lactams require continuous administration via portable infusion devices during OPAT. However, the use of benzylpenicillin in OPAT requires special consideration because of its limited stability at elevated temperatures. Methods: We tested the benzylpenicillin stability, pH, and degradation of products in elastomeric pumps at different concentrations in saline and in buffered solution containing sodium citrate during a prolonged storage and at high temperatures (seven days at 2-8 °C followed by 24 h at 37 °C). Additionally, drug concentrations during intermittent bolus infusion and during OPAT were determined in five patients. The concentrations and degradation products of benzylpenicillin were measured using liquid chromatography mass spectrometry (LC-MS/MS). Results: Unbuffered benzylpenicillin solutions that were already degraded during refrigerator storage and analyte concentration were not measurable after 8 days. The stability of the buffered solutions was acceptable at all three of the tested concentrations (97.6 ± 1.3%, 96.3 ± 0.8%, and 94.9 ± 1.1% for 10 Mio IU, 20 Mio IU, and 40 Mio IU of benzylpenicillin). The stability was influenced by benzylpenicillin concentration, and several breakdown products were identified. Benzylpenicillin concentrations were measured in five patients during OPAT and ranged from 7.2 to 60 mg/L. Conclusions: Benzylpenicillin buffered with sodium citrate is a safe and convenient option for use in continuous infusions during OPAT and should be favored over broad-spectrum antibiotics. Therapeutic drug monitoring data indicate sufficient to high plasma levels when patients received benzylpenicillin as continuous infusions.
Collapse
Affiliation(s)
- Katharina M. Rentsch
- Department of Laboratory Medicine, University Hospital Basel, 4031 Basel, Switzerland;
| | - Nina Khanna
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Delia Halbeisen
- Hospital Pharmacy, University Hospital Basel, 4031 Basel, Switzerland
| | - Michael Osthoff
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland
- Department of Internal Medicine, Cantonal Hospital Winterthur, 8400 Winterthur, Switzerland
| |
Collapse
|
8
|
Dyer CJ, De Waele JJ, Roberts JA. Antibiotic dose optimisation in the critically ill: targets, evidence and future strategies. Curr Opin Crit Care 2024; 30:439-447. [PMID: 39150038 DOI: 10.1097/mcc.0000000000001187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW To highlight the recent evidence for antibiotic pharmacokinetics and pharmacodynamics (PK/PD) in enhancing patient outcomes in sepsis and septic shock. We also summarise the limitations of available data and describe future directions for research to support translation of antibiotic dose optimisation to the clinical setting. RECENT FINDINGS Sepsis and septic shock are associated with poor outcomes and require antibiotic dose optimisation, mostly due to significantly altered pharmacokinetics. Many studies, including some randomised controlled trials have been conducted to measure the clinical outcome effects of antibiotic dose optimisation interventions including use of therapeutic drug monitoring. Current data support antibiotic dose optimisation for the critically ill. Further investigation is required to evolve more timely and robust precision antibiotic dose optimisation approaches, and to clearly quantify whether any clinical and health-economic benefits support expanded use of this treatment intervention. SUMMARY Antibiotic dose optimisation appears to improve outcomes in critically ill patients with sepsis and septic shock, however further research is required to quantify the level of benefit and develop a stronger knowledge of the role of new technologies to facilitate optimised dosing.
Collapse
Affiliation(s)
- Christopher J Dyer
- Herston Institute of Infectious Diseases (HeIDI), Metro North Health
- Pharmacy Department
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital (RBWH), Herston, Australia
| | - Jan J De Waele
- Department of Critical Care Medicine, Ghent University Hospital
- Dept of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jason A Roberts
- Herston Institute of Infectious Diseases (HeIDI), Metro North Health
- Pharmacy Department
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital (RBWH), Herston, Australia
- UQ Centre for Clinical Research (UQCCR), Faculty of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
9
|
Preijers T, Muller AE, Abdulla A, de Winter BCM, Koch BCP, Sassen SDT. Dose Individualisation of Antimicrobials from a Pharmacometric Standpoint: The Current Landscape. Drugs 2024; 84:1167-1178. [PMID: 39240531 PMCID: PMC11512831 DOI: 10.1007/s40265-024-02084-7] [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: 08/11/2024] [Indexed: 09/07/2024]
Abstract
Successful antimicrobial therapy depends on achieving optimal drug concentrations within individual patients. Inter-patient variability in pharmacokinetics (PK) and differences in pathogen susceptibility (reflected in the minimum inhibitory concentration, [MIC]) necessitate personalised approaches. Dose individualisation strategies aim to address this challenge, improving treatment outcomes and minimising the risk of toxicity and antimicrobial resistance. Therapeutic drug monitoring (TDM), with the application of population pharmacokinetic (popPK) models, enables model-informed precision dosing (MIPD). PopPK models mathematically describe drug behaviour across populations and can be combined with patient-specific TDM data to optimise dosing regimens. The integration of machine learning (ML) techniques promises to further enhance dose individualisation by identifying complex patterns within extensive datasets. Implementing these approaches involves challenges, including rigorous model selection and validation to ensure suitability for target populations. Understanding the relationship between drug exposure and clinical outcomes is crucial, as is striking a balance between model complexity and clinical usability. Additionally, regulatory compliance, outcome measurement, and practical considerations for software implementation will be addressed. Emerging technologies, such as real-time biosensors, hold the potential for revolutionising TDM by enabling continuous monitoring, immediate and frequent dose adjustments, and near patient testing. The ongoing integration of TDM, advanced modelling techniques, and ML within the evolving digital health care landscape offers a potential for enhancing antimicrobial therapy. Careful attention to model development, validation, and ethical considerations of the applied techniques is paramount for successfully optimising antimicrobial treatment for the individual patient.
Collapse
Affiliation(s)
- Tim Preijers
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Medical Microbiology, Haaglanden Medisch Centrum, The Hague, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Alan Abdulla
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands.
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands.
| | - Sebastiaan D T Sassen
- Department of Hospital Pharmacy, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
- Centre for Antimicrobial Treatment Optimization Rotterdam (CATOR), Rotterdam, The Netherlands
| |
Collapse
|
10
|
Gandéga H, Poujol H, Mezzarobba M, Muller L, Boyer JC, Lefrant JY, Leguelinel G, Roger C. Determinants of beta-lactam PK/PD target attainment in critically ill patients: A single center retrospective study. J Crit Care 2024; 83:154828. [PMID: 38759580 DOI: 10.1016/j.jcrc.2024.154828] [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: 01/22/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024]
Abstract
PURPOSE We aimed to identify factors associated with achieving target BL plasma concentrations and describe real world data for therapeutic drug monitoring (TDM). METHODS A retrospective single center study was conducted. We collected data from patients admitted to ICU with at least one BL TDM. We assessed the proportion of patients attaining the recommended plasma concentrations (i.e 100%fT > 4 to 8 MIC). Univariate and multivariate analyses was performed to identify the determinants of BL target attainment. RESULTS 156 patients were included. At the first dosing, 34% achieved target BL plasma concentrations, 50% were overdosed, and 16% were underdosed. Median time for 1st TDM were 4 (SD = 2.9) days. Multivariate analysis revealed that CKD-EPI estimated glomerular filtration rate (OR = 1.02; CI [1.01; 1.03]; p < 0.0001) and total body weight (OR = 1.03; CI [1.01; 1.04]; p = 0.0048) were the main determinant of BL target attainment. Conversely, Continuous Renal Replacement Therapy (OR = 0.28; CI [0.09; 0.89]; p = 0.0318) and meropenem use (OR = 0.31; CI [0.14; 0.69]; p = 0.0041) were identified as risk factors for overdosing. No factor was associated with underdosing. CONCLUSION Achieving target BL plasma concentrations remains challenging in ICUs. Identifying predictive factors of BL target attainment would favor implementing rapid dosing optimization strategies in both under and overdosing high risk patients.
Collapse
Affiliation(s)
- H Gandéga
- Department of Pharmacy, Nimes University Hospital, University of Montpellier, Nimes, France
| | - H Poujol
- Department of Pharmacy, Nimes University Hospital, University of Montpellier, Nimes, France; UR-UM103 IMAGINE, Univ Montpellier, Division of Anesthesia and Critical Care, Pain and Emergency Medicine, Nîmes University Hospital, Montpellier, France
| | - M Mezzarobba
- Department of Biostatistics, Clinical Epidemiology, Public Health, and Innovation in Methodology, CHU Nîmes, University of Montpellier, Nîmes, France
| | - L Muller
- UR-UM103 IMAGINE, Univ Montpellier, Division of Anesthesia and Critical Care, Pain and Emergency Medicine, Nîmes University Hospital, Montpellier, France
| | - J C Boyer
- Department of toxicity and biochemistry, Nimes University Hospital, Nimes, France
| | - J Y Lefrant
- UR-UM103 IMAGINE, Univ Montpellier, Division of Anesthesia and Critical Care, Pain and Emergency Medicine, Nîmes University Hospital, Montpellier, France
| | - G Leguelinel
- Department of Pharmacy, Nimes University Hospital, University of Montpellier, Nimes, France; Laboratory of Biostatistics, Epidemiology, Clinical Research and Health Economics, UPRES, EA 2415, University of Montpellier, Montpellier, France
| | - C Roger
- UR-UM103 IMAGINE, Univ Montpellier, Division of Anesthesia and Critical Care, Pain and Emergency Medicine, Nîmes University Hospital, Montpellier, France.
| |
Collapse
|
11
|
Schatz LM, Greppmair S, Kunzelmann AK, Starp J, Brinkmann A, Roehr A, Frey O, Hagel S, Dorn C, Zoller M, Scharf C, Wicha SG, Liebchen U. Predictive performance of multi-model approaches for model-informed precision dosing of piperacillin in critically ill patients. Int J Antimicrob Agents 2024; 64:107305. [PMID: 39146997 DOI: 10.1016/j.ijantimicag.2024.107305] [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: 05/07/2024] [Revised: 07/28/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES Piperacillin (PIP)/tazobactam is a frequently prescribed antibiotic; however, over- or underdosing may contribute to toxicity, therapeutic failure, and development of antimicrobial resistance. An external evaluation of 24 published PIP-models demonstrated that model-informed precision dosing (MIPD) can enhance target attainment. Employing various candidate models, this study aimed to assess the predictive performance of different MIPD-approaches comparing (i) a single-model approach, (ii) a model selection algorithm (MSA) and (iii) a model averaging algorithm (MAA). METHODS Precision, accuracy and expected target attainment, considering either initial (B1) or initial and secondary (B2) therapeutic drug monitoring (TDM)-samples per patient, were assessed in a multicentre dataset (561 patients, 11 German centres, 3654 TDM-samples). RESULTS The results demonstrated a slight superiority in predictive performance using MAA in B1, regardless of the candidate models, compared to MSA and the best single models (MAA, MSA, best single models: inaccuracy ±3%, ±10%, ±8%; imprecision: <25%, <31%, <28%; expected target attainment >77%, >71%, >73%). The inclusion of a second TDM-sample notably improved precision and target attainment for all MIPD-approaches, particularly within the context of MSA and most of the single models. The expected target attainment is maximized (up to >90%) when the TDM-sample is integrated within 24 h. CONCLUSIONS In conclusion, MAA streamlines MIPD by reducing the risk of selecting an inappropriate model for specific patients. Therefore, MIPD of PIP using MAA implicates further optimisation of antibiotic exposure in critically ill patients, by improving predictive performance with only one sample available for Bayesian forecasting, safety, and usability in clinical practice.
Collapse
Affiliation(s)
- Lea Marie Schatz
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Sebastian Greppmair
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Johannes Starp
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexander Brinkmann
- Department of Anesthesiology and Intensive Care Medicine, General Hospital of Heidenheim, Heidenheim, Germany
| | - Anka Roehr
- Department of Pharmacy, General Hospital of Heidenheim, Heidenheim, Germany
| | - Otto Frey
- Department of Pharmacy, General Hospital of Heidenheim, Heidenheim, Germany
| | - Stefan Hagel
- Institute for Infectious Diseases and Infection Control, University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christoph Dorn
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Michael Zoller
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christina Scharf
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Uwe Liebchen
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
12
|
Bahnasawy SM, Parrott NJ, Gijsen M, Spriet I, Friberg LE, Nielsen EI. Physiologically-based pharmacokinetic modelling in sepsis: A tool to elucidate how pathophysiology affects meropenem pharmacokinetics. Int J Antimicrob Agents 2024; 64:107352. [PMID: 39343059 DOI: 10.1016/j.ijantimicag.2024.107352] [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: 03/25/2024] [Revised: 07/26/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES Applying physiologically-based pharmacokinetic (PBPK) modelling in sepsis could help to better understand how PK changes are influenced by drug- and patient-related factors. We aimed to elucidate the influence of sepsis pathophysiology on the PK of meropenem by applying PBPK modelling. METHODS A whole-body meropenem PBPK model was developed and evaluated in healthy individuals, and renally impaired non-septic patients. Sepsis-induced physiological changes in body composition, organ blood flow, kidney function, albumin, and haematocrit were implemented according to a previously proposed PBPK sepsis model. Model performance was evaluated, and a local sensitivity analysis was conducted. RESULTS The model-predicted PK metrics (AUC, Cmax, CL, Vss) were within 1.33-fold-error margin of published data for 87.5% of the simulated profiles in healthy individuals. In sepsis, the model provided good predictions for literature-digitised average plasma and tissue exposure data, where the model-predicted AUC was within 1.33-fold-error margin for 9 out 11 simulated study profiles. Furthermore, the model was applied to individual plasma concentration data from 52 septic patients, where the model-predicted AUC, Cmax, and CL had a fold-error ratio range of 0.98-1.12, with alignment of the predicted and observed variability. For Vss, the fold-error ratio was 0.81, and the model underpredicted the population variability. CL was sensitive to renal plasma clearance, and kidney volume, whereas Vss was sensitive to the unbound fraction, organ volume fraction of the interstitial compartment, and the organ volume. CONCLUSIONS These findings may be extended to more diverse drug types and support a more mechanistic understanding of the effect of sepsis on drug exposure.
Collapse
Affiliation(s)
| | - Neil J Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | - Matthias Gijsen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
| | - Isabel Spriet
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
13
|
Morath B, Schultes L, Frey OR, Röhr AC, Christow H, Hoppe-Tichy T, Brinkmann A, Chiriac U. Development and Validation of a High-Performance Liquid Chromatography-Ultraviolet Spectrometry Method for Ampicillin and its Application in Routine Therapeutic Drug Monitoring of Intensive Care Patients. Ther Drug Monit 2024:00007691-990000000-00261. [PMID: 39289803 DOI: 10.1097/ftd.0000000000001253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Ampicillin/sulbactam, a combination of a β-lactam and β-lactamase inhibitor, is widely used in clinical settings. However, therapeutic drug monitoring (TDM) of ampicillin is not commonly performed, particularly in intensive care units (ICUs). The purpose of this study was to develop and validate a rapid and cost-effective high-performance liquid chromatography (HPLC)-ultraviolet spectrometry method to quantify ampicillin in human serum and evaluate its clinical application in ICU patients. METHODS Sample cleanup included a protein precipitation protocol, followed by chromatographic separation on a C18 reverse-phase HPLC column within 12.5 minutes using gradient elution of the mobile phase. The assay was validated according to the German Society of Toxicology and Forensic Chemistry criteria. Clinical applications involved the retrospective analysis of TDM data from ICU patients receiving continuous infusion of ampicillin/sulbactam, including the attainment of target ranges and individual predicted and observed pharmacokinetics. RESULTS The method was robust, with linear relations between the peak area responses and drug concentrations in the range of 2-128 mg/L. The coefficient of variation for precision and the bias for accuracy (both interday and intraday) were less than 10%. Clinical application revealed variable pharmacokinetics of ampicillin in ICU patients (clearance of 0.5-31.2 L/h). TDM-guided dose adjustments achieved good therapeutic drug exposure, with 92.9% of the samples being within the optimal (16-32 mg/L) or quasioptimal (8-48 mg/L) range. CONCLUSIONS This method provides a practical solution for the routine TDM of ampicillin, facilitating individualized dosing strategies to ensure adequate therapeutic drug exposure. Given its simplicity, cost-effectiveness, and clinical relevance, HPLC-ultraviolet spectrometry holds promise for broad implementation in hospital pharmacies and clinical laboratories.
Collapse
Affiliation(s)
- Benedict Morath
- Hospital Pharmacy, Heidelberg University Hospital, Heidelberg, Germany; and
| | - Linda Schultes
- Hospital Pharmacy, General Hospital Heidenheim, Heidenheim, Germany; and
| | - Otto Roman Frey
- Hospital Pharmacy, General Hospital Heidenheim, Heidenheim, Germany; and
| | - Anka Christa Röhr
- Hospital Pharmacy, General Hospital Heidenheim, Heidenheim, Germany; and
| | - Hannes Christow
- Departments of Internal Medicine and Intensive Care Medicine, and
| | | | - Alexander Brinkmann
- Anaesthesiology and Intensive Care Medicine, General Hospital Heidenheim, Heidenheim, Germany
| | - Ute Chiriac
- Hospital Pharmacy, Heidelberg University Hospital, Heidelberg, Germany; and
| |
Collapse
|
14
|
Dräger S, Ewoldt TMJ, Abdulla A, Rietdijk WJR, Verkaik NJ, van Vliet P, Purmer IM, Osthoff M, Koch BCP, Endeman H. Target attainment of beta-lactam antibiotics and ciprofloxacin in critically ill patients and its association with 28-day mortality. J Crit Care 2024:154904. [PMID: 39277523 DOI: 10.1016/j.jcrc.2024.154904] [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: 06/21/2024] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVES This study aims to assess pharmacodynamic target attainment in critically ill patients and identify factors influencing target attainment and mortality outcomes. METHODS We analysed data from the DOLPHIN trial. Beta-lactam and ciprofloxacin peak and trough concentration were measured within the first 36 h (T1) after initiation of treatment. The study outcome included the rate of pharmacodynamic target attainment of 100 % ƒT>1xEpidemiological cut-off value (ECOFF) for beta-lactams, and of fAUC0-24h/ECOFF>125 for ciprofloxacin at T1. RESULTS The target attainment rates were 78.1 % (n = 228/292) for beta-lactams, and 41.5 % (n = 39/94) for ciprofloxacin, respectively. Lower estimated glomerular filtration rate and higher SOFA score were associated with target attainment. In patients receiving beta-lactams, 28-day mortality was significantly higher in patients who attained 100 % ƒT>1xECOFF (28.9 % vs. 12.5 %; p = 0.01). In the multivariate analysis, attainment of 100 % ƒT>4xECOFF, but not 100 % ƒT>1xECOFF, was associated with a higher 28-day mortality (OR 2.70, 95 % CI 1.36-5.48 vs. OR 1.28, 95 % CI 0.53-3.34). CONCLUSIONS A high rate of target attainment (100 % ƒT>1xECOFF) for beta-lactams and a lower rate for ciprofloxacin was observed. Achieving exposures of 100 % ƒT>4xECOFF was associated with 28-day mortality. The impact of antibiotic target attainment on clinical outcome needs to be a focus of future research.
Collapse
Affiliation(s)
- Sarah Dräger
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands; Department of Internal Medicine, University Hospital Basel, Basel, Switzerland.
| | - Tim M J Ewoldt
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands; Department of Intensive Care Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alan Abdulla
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
| | - Wim J R Rietdijk
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Institutional Affairs, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Nelianne J Verkaik
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Peter van Vliet
- Department of Intensive Care Medicine, Haaglanden Medical Center, The Hague, the Netherlands
| | - Ilse M Purmer
- Department of Intensive Care Medicine, Haga Hospital, The Hague, the Netherlands
| | - Michael Osthoff
- Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands; Rotterdam Clinical Pharmacometrics Group, Rotterdam, the Netherlands
| | - Henrik Endeman
- Department of Intensive Care Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Carrié C, Butruille J, Maingault S, Lannou A, Dubuisson V, Petit L, Biais M, Breilh D. Pharmacokinetics of Piperacillin-Tazobactam in Critically Ill Patients with Open Abdomen and Vacuum-Assisted Wound Closure: Dosing Considerations Using Monte Carlo Simulation. Pharmaceutics 2024; 16:1191. [PMID: 39339227 PMCID: PMC11434833 DOI: 10.3390/pharmaceutics16091191] [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: 07/29/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Open abdomen with vacuum-assisted wound closure therapy (OA/VAC) is frequently used in critically ill patients although the impact of OA/VAC on antibiotics pharmacokinetics (PK) remains unknown. We thus aimed to characterize the PK of piperacillin-tazobactam (PTZ) in critically ill patients with OA/VAC and assess the optimal dosing regimens based on pharmacodynamics (PD) target attainment. METHODS Over a 15-month study period, 45 patients with OA/VAC treated with PTZ administered continuously and adapted to 24 h creatinine clearance (CLCR) underwent measurements of free concentrations in their plasma, urine, VAC exudate, and peritoneal fluid. Population PK modeling was performed considering the effect of covariates, and Monte Carlo simulations were employed to determine the probability of target attainment (PTA) for the PK/PD targets (100% fT > 16 mg/L) in the plasma and at the peritoneal site at steady state. RESULTS Piperacillin concentrations were described using a two-compartment model, with age and total body weight as significant covariates for central volume of distribution (V1) and estimated renal function for clearance (CL). Tazobactam concentrations were described using a two-compartment model with estimated renal function as a significant covariate. The central volume of distributions V1 of piperacillin and tazobactam were 21.2 and 23.2 L, respectively. The VAC-induced peritoneal clearance was negligible compared to renal clearance. Most patients achieved the desirable PK/PD target when using a CLCR-pondered PTZ dosing regimen from 12 g/1.5 g/day to 20 g/2.5 g/day. CONCLUSIONS Despite a wide inter-individual variability, the influence of OA/VAC on piperacillin and tazobactam PK parameters is not straightforward. The use of a CLCR-pondered PTZ dosing regimen from 12 g/1.5 g/day to 20 g/2.5 g/day is needed to reach a PTA > 85%.
Collapse
Affiliation(s)
- Cédric Carrié
- Surgical and Trauma Intensive Care Unit, Anesthesiology and Critical Care Department, Hôpital Pellegrin, CHU Bordeaux, 33076 Bordeaux, France
| | - Jesse Butruille
- Pharmacokinetics and Clinical Pharmacy Laboratory, University of Bordeaux, 33000 Bordeaux, France
- Clinical Pharmacy and City-Hospital Network Department, CHU Bordeaux, 33000 Bordeaux, France
| | - Sophie Maingault
- Surgical and Trauma Intensive Care Unit, Anesthesiology and Critical Care Department, Hôpital Pellegrin, CHU Bordeaux, 33076 Bordeaux, France
| | - Alexandre Lannou
- Surgical and Trauma Intensive Care Unit, Anesthesiology and Critical Care Department, Hôpital Pellegrin, CHU Bordeaux, 33076 Bordeaux, France
| | - Vincent Dubuisson
- Digestive, Emergency and Trauma Surgery, CHU Bordeaux, 33000 Bordeaux, France
| | - Laurent Petit
- Surgical and Trauma Intensive Care Unit, Anesthesiology and Critical Care Department, Hôpital Pellegrin, CHU Bordeaux, 33076 Bordeaux, France
| | - Matthieu Biais
- Surgical and Trauma Intensive Care Unit, Anesthesiology and Critical Care Department, Hôpital Pellegrin, CHU Bordeaux, 33076 Bordeaux, France
| | - Dominique Breilh
- Pharmacokinetics and Clinical Pharmacy Laboratory, University of Bordeaux, 33000 Bordeaux, France
- Clinical Pharmacy and City-Hospital Network Department, CHU Bordeaux, 33000 Bordeaux, France
| |
Collapse
|
16
|
Gonçalves Pereira J, Fernandes J, Mendes T, Gonzalez FA, Fernandes SM. Artificial Intelligence to Close the Gap between Pharmacokinetic/Pharmacodynamic Targets and Clinical Outcomes in Critically Ill Patients: A Narrative Review on Beta Lactams. Antibiotics (Basel) 2024; 13:853. [PMID: 39335027 PMCID: PMC11428226 DOI: 10.3390/antibiotics13090853] [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: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial dosing can be a complex challenge. Although a solid rationale exists for a link between antibiotic exposure and outcome, conflicting data suggest a poor correlation between pharmacokinetic/pharmacodynamic targets and infection control. Different reasons may lead to this discrepancy: poor tissue penetration by β-lactams due to inflammation and inadequate tissue perfusion; different bacterial response to antibiotics and biofilms; heterogeneity of the host's immune response and drug metabolism; bacterial tolerance and acquisition of resistance during therapy. Consequently, either a fixed dose of antibiotics or a fixed target concentration may be doomed to fail. The role of biomarkers in understanding and monitoring host response to infection is also incompletely defined. Nowadays, with the ever-growing stream of data collected in hospitals, utilizing the most efficient analytical tools may lead to better personalization of therapy. The rise of artificial intelligence and machine learning has allowed large amounts of data to be rapidly accessed and analyzed. These unsupervised learning models can apprehend the data structure and identify homogeneous subgroups, facilitating the individualization of medical interventions. This review aims to discuss the challenges of β-lactam dosing, focusing on its pharmacodynamics and the new challenges and opportunities arising from integrating machine learning algorithms to personalize patient treatment.
Collapse
Affiliation(s)
- João Gonçalves Pereira
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Serviço de Medicina Intensiva, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Joana Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Centro Hospitalar de Trás-os-Montes e Alto Douro, 5000-508 Vila Real, Portugal
| | - Tânia Mendes
- Serviço de Medicina Interna, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Filipe André Gonzalez
- Serviço de Medicina Intensiva, Hospital Garcia De Orta, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Susana M Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Hospital Santa Maria, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
17
|
Kim YK, Kang G, Zang DY, Lee DH. Precision Dosing of Meropenem in Adults with Normal Renal Function: Insights from a Population Pharmacokinetic and Monte Carlo Simulation Study. Antibiotics (Basel) 2024; 13:849. [PMID: 39335022 PMCID: PMC11429322 DOI: 10.3390/antibiotics13090849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to develop a population pharmacokinetic (PK) model for meropenem in healthy adults and explore optimal dosing regimens for patients with normal renal function. PK samples were obtained from 12 healthy participants, which were analyzed using noncompartmental analysis and nonlinear mixed-effect modeling. The PK profiles of meropenem were characterized using a two-compartment model, and serum creatinine level was identified as a significant covariate affecting total clearance. Monte Carlo simulations were conducted using this model to inform dosing recommendations. The target index for meropenem efficacy was defined as the cumulative percentage over 24 h during which free (f) drug concentration exceeded the minimum inhibitory concentration (MIC) under steady state conditions (fT>MIC). These simulations indicated that the current dosage regimen of 1 g for 30 min infusions every 8 h achieved a 90% probability of target attainment (PTA) for 40%fT>MIC when the MIC was <2 mg/L. However, to achieve more stringent therapeutic targets, such as a 90%PTA for 100%fT>MIC or a 90%PTA for 100%fT>4MIC, higher doses administered as 3 h extended infusions or as continuous infusions may be necessary. These results highlight the need for model-informed precision dosing to enhance the efficacy of meropenem therapy across various MIC levels and therapeutic targets.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Division of Infectious Diseases, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14066, Republic of Korea
| | - Gaeun Kang
- Division of Clinical Pharmacology, Chonnam National University Hospital, Gfwangju 61469, Republic of Korea
| | - Dae Young Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14066, Republic of Korea
| | - Dong Hwan Lee
- Department of Clinical Pharmacology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14066, Republic of Korea
| |
Collapse
|
18
|
Childs-Kean LM, Rivera CG, Venugopalan V, Johnson MJ, Barreto EF. The OPAT opportunity for beta-lactam individualization. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2024; 4:e115. [PMID: 39257422 PMCID: PMC11384153 DOI: 10.1017/ash.2024.367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 09/12/2024]
Abstract
Beta-lactam therapeutic drug monitoring has been growing in prevalence in the acute care hospital setting. Expansion of its use to outpatient parenteral antimicrobial therapy requires careful consideration of potential logistical and therapeutic barriers.
Collapse
Affiliation(s)
- Lindsey M Childs-Kean
- Department of Pharmacy Education and Practice, University of Florida College of Pharmacy, Gainesville, FL, USA
| | | | - Veena Venugopalan
- Department of Pharmacy Education and Practice, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Madelyn J Johnson
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA
- North Dakota State University College of Pharmacy, Fargo, ND, USA
| | | |
Collapse
|
19
|
Abdul-Aziz MH, Hammond NE, Brett SJ, Cotta MO, De Waele JJ, Devaux A, Di Tanna GL, Dulhunty JM, Elkady H, Eriksson L, Hasan MS, Khan AB, Lipman J, Liu X, Monti G, Myburgh J, Novy E, Omar S, Rajbhandari D, Roger C, Sjövall F, Zaghi I, Zangrillo A, Delaney A, Roberts JA. Prolonged vs Intermittent Infusions of β-Lactam Antibiotics in Adults With Sepsis or Septic Shock: A Systematic Review and Meta-Analysis. JAMA 2024; 332:638-648. [PMID: 38864162 PMCID: PMC11170459 DOI: 10.1001/jama.2024.9803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Importance There is uncertainty about whether prolonged infusions of β-lactam antibiotics improve clinically important outcomes in critically ill adults with sepsis or septic shock. Objective To determine whether prolonged β-lactam antibiotic infusions are associated with a reduced risk of death in critically ill adults with sepsis or septic shock compared with intermittent infusions. Data Sources The primary search was conducted with MEDLINE (via PubMed), CINAHL, Embase, Cochrane Central Register of Controlled Trials (CENTRAL), and ClinicalTrials.gov from inception to May 2, 2024. Study Selection Randomized clinical trials comparing prolonged (continuous or extended) and intermittent infusions of β-lactam antibiotics in critically ill adults with sepsis or septic shock. Data Extraction and Synthesis Data extraction and risk of bias were assessed independently by 2 reviewers. Certainty of evidence was evaluated with the Grading of Recommendations Assessment, Development and Evaluation approach. A bayesian framework was used as the primary analysis approach and a frequentist framework as the secondary approach. Main Outcomes and Measures The primary outcome was all-cause 90-day mortality. Secondary outcomes included intensive care unit (ICU) mortality and clinical cure. Results From 18 eligible randomized clinical trials that included 9108 critically ill adults with sepsis or septic shock (median age, 54 years; IQR, 48-57; 5961 men [65%]), 17 trials (9014 participants) contributed data to the primary outcome. The pooled estimated risk ratio for all-cause 90-day mortality for prolonged infusions of β-lactam antibiotics compared with intermittent infusions was 0.86 (95% credible interval, 0.72-0.98; I2 = 21.5%; high certainty), with a 99.1% posterior probability that prolonged infusions were associated with lower 90-day mortality. Prolonged infusion of β-lactam antibiotics was associated with a reduced risk of intensive care unit mortality (risk ratio, 0.84; 95% credible interval, 0.70-0.97; high certainty) and an increase in clinical cure (risk ratio, 1.16; 95% credible interval, 1.07-1.31; moderate certainty). Conclusions and Relevance Among adults in the intensive care unit who had sepsis or septic shock, the use of prolonged β-lactam antibiotic infusions was associated with a reduced risk of 90-day mortality compared with intermittent infusions. The current evidence presents a high degree of certainty for clinicians to consider prolonged infusions as a standard of care in the management of sepsis and septic shock. Trial Registration PROSPERO Identifier: CRD42023399434.
Collapse
Affiliation(s)
- Mohd H. Abdul-Aziz
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Naomi E. Hammond
- Critical Care Program, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- Malcolm Fisher Department of Intensive Care, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Stephen J. Brett
- Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Menino O. Cotta
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Jan J. De Waele
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent, Belgium
| | - Anthony Devaux
- Statistics Division, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
| | - Gian Luca Di Tanna
- Statistics Division, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- Department of Business Economics, Health and Social Care, University of Applied Sciences and Arts of Southern Switzerland, Manno, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Joel M. Dulhunty
- Critical Care Program, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Redcliffe Hospital, Redcliffe, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Hatem Elkady
- Department of Intensive Care Medicine, Westmead Hospital, Sydney, New South Wales, Australia
| | - Lars Eriksson
- UQ Library, The University of Queensland, Brisbane, Queensland, Australia
| | - M. Shahnaz Hasan
- Department of Anesthesiology, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Ayesha Bibi Khan
- Division of Critical Care, University of Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Jeffrey Lipman
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Jamieson Trauma Institute, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Division of Anesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Xiaoqiu Liu
- Statistics Division, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- School of Population Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Giacomo Monti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - John Myburgh
- Critical Care Program, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- Department of Intensive Care, St George Hospital, Kogarah, New South Wales, Australia
| | - Emmanuel Novy
- Service d’anesthésie-réanimation et médicine péri-opératoire Brabois adulte, CHRU de Nancy, Nancy, France
- Université de Lorraine, SIMPA, Nancy, France
| | - Shahed Omar
- Division of Critical Care, University of Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Dorrilyn Rajbhandari
- Critical Care Program, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
| | - Claire Roger
- Département d’anesthésie et réanimation, douleur et médecine d’urgence, CHU Carémeau, Nîmes, France
- UR UM 103IMAGINE, Faculté de Médecine, Montpellier Université, Nîmes, France
| | - Fredrik Sjövall
- Intensive and Perioperative Care, Skåne University Hospital, Malmö, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Irene Zaghi
- Department of Diagnostic and Experimental Medicine, University of Bologna, Bologna, Italy
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Anthony Delaney
- Critical Care Program, The George Institute for Global Health and University of New South Wales, Sydney, New South Wales, Australia
- Malcolm Fisher Department of Intensive Care, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Jason A. Roberts
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Division of Anesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
- Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, Queensland, Australia
| |
Collapse
|
20
|
Dulhunty JM, Brett SJ, De Waele JJ, Rajbhandari D, Billot L, Cotta MO, Davis JS, Finfer S, Hammond NE, Knowles S, Liu X, McGuinness S, Mysore J, Paterson DL, Peake S, Rhodes A, Roberts JA, Roger C, Shirwadkar C, Starr T, Taylor C, Myburgh JA, Lipman J. Continuous vs Intermittent β-Lactam Antibiotic Infusions in Critically Ill Patients With Sepsis: The BLING III Randomized Clinical Trial. JAMA 2024; 332:629-637. [PMID: 38864155 PMCID: PMC11170452 DOI: 10.1001/jama.2024.9779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Importance Whether β-lactam antibiotics administered by continuous compared with intermittent infusion reduces the risk of death in patients with sepsis is uncertain. Objective To evaluate whether continuous vs intermittent infusion of a β-lactam antibiotic (piperacillin-tazobactam or meropenem) results in decreased all-cause mortality at 90 days in critically ill patients with sepsis. Design, Setting, and Participants An international, open-label, randomized clinical trial conducted in 104 intensive care units (ICUs) in Australia, Belgium, France, Malaysia, New Zealand, Sweden, and the United Kingdom. Recruitment occurred from March 26, 2018, to January 11, 2023, with follow-up completed on April 12, 2023. Participants were critically ill adults (≥18 years) treated with piperacillin-tazobactam or meropenem for sepsis. Intervention Eligible patients were randomized to receive an equivalent 24-hour dose of a β-lactam antibiotic by either continuous (n = 3498) or intermittent (n = 3533) infusion for a clinician-determined duration of treatment or until ICU discharge, whichever occurred first. Main Outcomes and Measures The primary outcome was all-cause mortality within 90 days after randomization. Secondary outcomes were clinical cure up to 14 days after randomization; new acquisition, colonization, or infection with a multiresistant organism or Clostridioides difficile infection up to 14 days after randomization; ICU mortality; and in-hospital mortality. Results Among 7202 randomized participants, 7031 (mean [SD] age, 59 [16] years; 2423 women [35%]) met consent requirements for inclusion in the primary analysis (97.6%). Within 90 days, 864 of 3474 patients (24.9%) assigned to receive continuous infusion had died compared with 939 of 3507 (26.8%) assigned intermittent infusion (absolute difference, -1.9% [95% CI, -4.9% to 1.1%]; odds ratio, 0.91 [95% CI, 0.81 to 1.01]; P = .08). Clinical cure was higher in the continuous vs intermittent infusion group (1930/3467 [55.7%] and 1744/3491 [50.0%], respectively; absolute difference, 5.7% [95% CI, 2.4% to 9.1%]). Other secondary outcomes were not statistically different. Conclusions and Relevance The observed difference in 90-day mortality between continuous vs intermittent infusions of β-lactam antibiotics did not meet statistical significance in the primary analysis. However, the confidence interval around the effect estimate includes the possibility of both no important effect and a clinically important benefit in the use of continuous infusions in this group of patients. Trial Registration ClinicalTrials.gov Identifier: NCT03213990.
Collapse
Affiliation(s)
- Joel M. Dulhunty
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Redcliffe Hospital, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
| | - Stephen J. Brett
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Jan J. De Waele
- Department of Critical Care Medicine, Ghent University Hospital, Ghent, Belgium
| | - Dorrilyn Rajbhandari
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Laurent Billot
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Menino O. Cotta
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
| | - Joshua S. Davis
- Infection Research Program, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
- Department of Infectious Diseases, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Simon Finfer
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
- School of Public Health, Imperial College London, United Kingdom
| | - Naomi E. Hammond
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
- Malcolm Fisher Department of Intensive Care, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Serena Knowles
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Xiaoqiu Liu
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Shay McGuinness
- Auckland City Hospital, Auckland, New Zealand
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Jayanthi Mysore
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - David L. Paterson
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
- Saw Swee Hock School of Public Health and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sandra Peake
- The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew Rhodes
- St George’s University Hospitals NHS Foundation Trust, London, United Kingdom
- St George’s University of London, London, United Kingdom
| | - Jason A. Roberts
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
- Herston Infectious Diseases Institute, Metro North Health, Brisbane, Queensland, Australia
- Department of Anesthesiology, Critical Care, Pain, and Emergency Medicine, University Hospital of Nîmes, Nîmes, France
| | - Claire Roger
- Department of Anesthesiology, Critical Care, Pain, and Emergency Medicine, University Hospital of Nîmes, Nîmes, France
- University of Montpellier, Montpellier, France
| | | | - Therese Starr
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Metro North Health, Brisbane, Queensland, Australia
| | - Colman Taylor
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - John A. Myburgh
- The George Institute for Global Health, The University of New South Wales, Sydney, New South Wales, Australia
- St George Hospital, Sydney, New South Wales, Australia
| | - Jeffrey Lipman
- Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
- Department of Anesthesiology, Critical Care, Pain, and Emergency Medicine, University Hospital of Nîmes, Nîmes, France
| |
Collapse
|
21
|
Gras-Martín L, Plaza-Diaz A, Zarate-Tamames B, Vera-Artazcoz P, Torres OH, Bastida C, Soy D, Ruiz-Ramos J. Risk Factors Associated with Antibiotic Exposure Variability in Critically Ill Patients: A Systematic Review. Antibiotics (Basel) 2024; 13:801. [PMID: 39334976 PMCID: PMC11428266 DOI: 10.3390/antibiotics13090801] [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: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
(1) Background: Knowledge about the behavior of antibiotics in critically ill patients has been increasing in recent years. Some studies have concluded that a high percentage may be outside the therapeutic range. The most likely cause of this is the pharmacokinetic variability of critically ill patients, but it is not clear which factors have the greatest impact. The aim of this systematic review is to identify risk factors among critically ill patients that may exhibit significant pharmacokinetic alterations, compromising treatment efficacy and safety. (2) Methods: The search included the PubMed, Web of Science, and Embase databases. (3) Results: We identified 246 observational studies and ten clinical trials. The most studied risk factors in the literature were renal function, weight, age, sex, and renal replacement therapy. Risk factors with the greatest impact included renal function, weight, renal replacement therapy, age, protein or albumin levels, and APACHE or SAPS scores. (4) Conclusions: The review allows us to identify which critically ill patients are at a higher risk of not reaching therapeutic targets and helps us to recognize the extensive number of risk factors that have been studied, guiding their inclusion in future studies. It is essential to continue researching, especially in real clinical practice and with clinical outcomes.
Collapse
Affiliation(s)
- Laura Gras-Martín
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Adrián Plaza-Diaz
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
| | - Borja Zarate-Tamames
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
| | - Paula Vera-Artazcoz
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Intensive Care Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Olga H Torres
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Geriatric Unit, Internal Medicine Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Carla Bastida
- Pharmacy Department, Division of Medicines, Hospital Clinic of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy, Universitat de Barcelona, Campus Diagonal, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Dolors Soy
- Pharmacy Department, Division of Medicines, Hospital Clinic of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy, Universitat de Barcelona, Campus Diagonal, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Jesús Ruiz-Ramos
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
22
|
Ates HC, Alshanawani A, Hagel S, Cotta MO, Roberts JA, Dincer C, Ates C. Unraveling the impact of therapeutic drug monitoring via machine learning for patients with sepsis. Cell Rep Med 2024; 5:101681. [PMID: 39127039 PMCID: PMC11384951 DOI: 10.1016/j.xcrm.2024.101681] [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: 11/13/2023] [Revised: 05/25/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024]
Abstract
Clinical studies investigating the benefits of beta-lactam therapeutic drug monitoring (TDM) among critically ill patients are hindered by small patient groups, variability between studies, patient heterogeneity, and inadequate use of TDM. Accordingly, definitive conclusions regarding the efficacy of TDM remain elusive. To address these challenges, we propose an innovative approach that leverages data-driven methods to unveil the concealed connections between therapy effectiveness and patient data, collected through a randomized controlled trial (DRKS00011159; 10th October 2016). Our findings reveal that machine learning algorithms can successfully identify informative features that distinguish between healthy and sick states. These hold promise as potential markers for disease classification and severity stratification, as well as offering a continuous and data-driven "multidimensional" Sequential Organ Failure Assessment (SOFA) score. The positive impact of TDM on patient recovery rates is demonstrated by unraveling the intricate connections between therapy effectiveness and clinically relevant data via machine learning.
Collapse
Affiliation(s)
- H Ceren Ates
- University of Freiburg, FIT Freiburg Centre for Interactive Materials and Bioinspired Technology, 79110 Freiburg, Germany; University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg, Germany
| | - Abdallah Alshanawani
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg, Germany
| | - Stefan Hagel
- Institute for Infectious Diseases and Infection Control, Jena University Hospital - Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Menino O Cotta
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Jason A Roberts
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4006, Australia; Departments of Intensive Care Medicine and Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, QLD 4006, Australia; Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, 34295 Nîmes, France
| | - Can Dincer
- University of Freiburg, FIT Freiburg Centre for Interactive Materials and Bioinspired Technology, 79110 Freiburg, Germany; University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg, Germany.
| | - Cihan Ates
- Karlsruhe Institute of Technology (KIT), Machine Intelligence in Energy Systems, 76131 Karlsruhe, Germany; Karlsruhe Institute of Technology (KIT), Center of Health Technologies, 76131 Karlsruhe, Germany.
| |
Collapse
|
23
|
Paice K, Tang Girdwood S, Mizuno T, Pavia K, Punt N, Tang P, Dong M, Curry C, Jones R, Gibson A, Vinks AA, Kaplan J. Pharmacokinetic Factors Associated With Early Meropenem Target Attainment in Pediatric Severe Sepsis. Pediatr Crit Care Med 2024:00130478-990000000-00376. [PMID: 39162600 DOI: 10.1097/pcc.0000000000003599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
OBJECTIVES To determine the frequency of early meropenem concentration target attainment (TA) in critically ill children with severe sepsis; to explore clinical, therapeutic, and pharmacokinetic factors associated with TA; and to assess how fluid resuscitation and volume status relate to early TA. DESIGN Retrospective analysis of prospective observational cohort study. SETTING PICU in a single academic quaternary care children's hospital. PATIENTS Twenty-nine patients starting meropenem for severe sepsis (characterized as need for positive pressure ventilation, vasopressors, or ≥ 40 mL/kg bolused fluid), of which 17 were newly escalated to PICU level care. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Concentration-time profiles were analyzed using modeling software employing opportunistic sampling, Bayesian estimation, and a population pharmacokinetic model. Time above four times minimum inhibitory concentration (T > 4×MIC), using the susceptibility breakpoint of 1 µg/mL, was determined for each patient over the first 24 hours of meropenem therapy, as well as individual clearance and volume of distribution (Vd) estimates. Twenty-one of 29 patients met a target of 40%T > MIC 4 μg/mL. Reaching TA, vs. not, was associated with lower meropenem clearance. We failed to identify a difference in Vd or an association between the TA group and age, weight, creatinine-based estimated glomerular filtration rate (eGFR), or the amount of fluid administered. eGFR was, however, negatively correlated with overall T > MIC. CONCLUSIONS Eight of 29 pediatric patients with early severe sepsis did not meet the selected TA threshold within the first 24 hours of meropenem therapy. Higher clearance was associated with failure to meet targets. Identifying patients likely to have higher meropenem clearance could help with dosing regimens.
Collapse
Affiliation(s)
- Kelli Paice
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sonya Tang Girdwood
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Tomoyuki Mizuno
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kathryn Pavia
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Nieko Punt
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical, Center Groningen, Groningen, The Netherlands
- Medimatics, Maastricht, The Netherlands
| | - Peter Tang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Min Dong
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Calise Curry
- Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Rhonda Jones
- Clinical Quality Improvement Systems, James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Abigayle Gibson
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Alexander A Vinks
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jennifer Kaplan
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
24
|
Carson AA, Bowker KE, Attwood M, Noel AR, MacGowan AP. Pharmacodynamics of piperacillin/tazobactam against Pseudomonas aeruginosa: antibacterial effect and risk of emergence of resistance. JAC Antimicrob Resist 2024; 6:dlae108. [PMID: 39005592 PMCID: PMC11242448 DOI: 10.1093/jacamr/dlae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Affiliation(s)
- Amy A Carson
- Infection Sciences, Pathology Quarter, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
| | - Karen E Bowker
- Bristol Centre for Antimicrobial Research & Evaluation (BCARE), Severn Infection Sciences, Pathology Quarter, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
| | - Marie Attwood
- Bristol Centre for Antimicrobial Research & Evaluation (BCARE), Severn Infection Sciences, Pathology Quarter, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
| | - Alan R Noel
- Bristol Centre for Antimicrobial Research & Evaluation (BCARE), Severn Infection Sciences, Pathology Quarter, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
| | - Alasdair P MacGowan
- Bristol Centre for Antimicrobial Research & Evaluation (BCARE), Severn Infection Sciences, Pathology Quarter, Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, UK
| |
Collapse
|
25
|
Williams P, Cotta MO, Tabah A, Sandaradura I, Kanji S, Scheetz MH, Imani S, Elhadi M, Pardos SL, Schellack N, Sanches C, Timsit JF, Xie J, Farkas A, Wilks K, Roberts JA. Antimicrobial therapeutic drug monitoring in critically ill adult patients - An international perspective on access, utilisation, and barriers. Int J Antimicrob Agents 2024; 64:107192. [PMID: 38734215 DOI: 10.1016/j.ijantimicag.2024.107192] [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: 11/19/2023] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) is an effective method for individualising antimicrobial therapy in critically ill patients. The 2021 ADMIN-intensive care unit survey studied a wide range of intensive care unit clinicians worldwide to gain their perspectives on antimicrobial TDM. This article reports the responses from this survey relating to TDM access, utilisation, and barriers. METHODS An online survey consisted of multiple-choice questions and 5-point Likert scales. The survey examined respondent's access to minimum inhibitory concentration (MIC) results, drug assays, and dosing software, as well as barriers to TDM. RESULTS The survey included 538 clinicians from 409 hospitals in 45 countries, with 71% physicians and 29% pharmacists. Despite most respondents having access to assays, 21% and 26% of respondents lacked access to vancomycin and aminoglycosides, respectively. In lower-income countries, almost 40% reported no access. Delayed drug assay turnaround time was the most significant barrier to TDM, particularly in lower-income countries. Routine access to MIC results was unavailable for 41% of respondents, with 25% of lower-income country respondents having no access to MIC or susceptibility reports. CONCLUSIONS This global survey indicated that consistent TDM usage is hindered by assay access in some sites and the timeliness of assay results in others. Addressing barriers to TDM, particularly in low-income countries, should be a priority to ensure equitable access to affordable TDM.
Collapse
Affiliation(s)
- Paul Williams
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia; Pharmacy Department, Sunshine Coast University Hospital, Birtinya, Queensland, Australia.
| | - Menino Osbert Cotta
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia
| | - Alexis Tabah
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia; Intensive Care Unit, Redcliffe Hospital, Redcliffe, Queensland, Australia; Queensland University of Technology, Brisbane, Queensland, Australia
| | - Indy Sandaradura
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia; Centre for Infectious Diseases and Microbiology, Westmead Hospital, Sydney, New South Wales, Australia; Institute for Clinical Pathology and Medical Research, New South Wales Health Pathology, Sydney, New South Wales, Australia
| | - Salmaan Kanji
- The Ottawa Hospital and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Marc H Scheetz
- Pharmacometric Center of Excellence, Departments of Pharmacy Practice and Pharmacology, College of Pharmacy, Midwestern University, Downers Grove, Illinois, USA
| | - Sahand Imani
- Nepean Blue Mountains Local Health District, Nepean Hospital, Sydney, New South Wales, Australia
| | | | - Sònia Luque Pardos
- Pharmacy Department, Parc de Salut Mar, Barcelona, Spain; Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain; CIBER of Pharmacy, Saint Clare's Infectious Diseases (CIBERINFEC CB21/13/0002) Institute of Health Carlos III, Madrid, Spain
| | - Natalie Schellack
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Cristina Sanches
- Campus Centro Oeste Dona Lindu, Federal University of Sao João del Rei, Divinópolis, Minas Gerais, Brazil
| | - Jean Francois Timsit
- Assistance Publique Hôpitaux de Paris - Bichat Hospital Medical and Infectious Diseases ICU (MI2), Paris France; IAME U 1137 Université Paris-Cité Site Bichat, Paris, France
| | - Jiao Xie
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Andras Farkas
- Optimum Dosing Strategies, Bloomingdale, New Jersey, USA; Department of Pharmacy, Saint Clare's Health, Denville, New Jersey, USA
| | - Kathryn Wilks
- Infectious Diseases Department, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Jason A Roberts
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia; Departments of Intensive Care Medicine and Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, Australia; Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes France
| |
Collapse
|
26
|
M J, Khan MN, R A, Sundararajan S, R NK. Prescribing Trend of Antibiotics Among the Patients Admitted in Intensive Medical Care Unit: A Prospective Observational Study. Cureus 2024; 16:e67101. [PMID: 39290938 PMCID: PMC11406113 DOI: 10.7759/cureus.67101] [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: 07/26/2024] [Accepted: 08/17/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Intensive medical care units (IMCUs) usually admit patients who are in critical medical need and require the utmost attention of healthcare professionals, along with the best treatment options available. These patients are prone to infections and require various antibiotics for the treatment. Varying costs of antibiotics, variable dosage forms, and antibiotic resistance cause an economic burden on patients Methodology: This study was designed and conducted prospectively to evaluate the prescribing pattern of antibiotics at the IMCU in a tertiary care hospital. A total of 102 patients were included in the study based on the exclusion and inclusion criteria, and the collected data was tabulated in an Excel sheet and analyzed using Prism GraphPad software. Data were presented as numbers and percentages. RESULTS Most of the patients were in the age group of 41-50 years. The number of male patients was slightly more than that of female patients. The majority of the patients admitted to the IMCU had acute pulmonary edema and cerebrovascular accidents. Most antibiotics were prescribed empirically and administered parenterally, of which Amoxicillin + Potassium clavulanate was the most commonly used antibiotic. Tigecycline had the highest daily defined dose per 100 bed days value, and injection Sulbactam + Cefoperazone was the costliest of all antibiotic therapy. CONCLUSION Antibiotic therapy used in the IMCU consisting of Sulbactam + Cefoperazone was found to be costlier, and Amoxicillin + Potassium clavulanate were the commonly prescribed antibiotics among the other prescribed antibiotics. The average cost of antibiotics was found to be higher, which increased the economic healthcare burden for patients and their families.
Collapse
Affiliation(s)
- Jegatheeswaran M
- Department of Pharmacy Practice, SRM (Sri Ramaswamy Memorial) College of Pharmacy, Faculty of Medical and Health Sciences, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Mohammad Nezamuddin Khan
- Department of Pharmacy Practice, SRM (Sri Ramaswamy Memorial) College of Pharmacy, Faculty of Medical and Health Sciences, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Ajith R
- Department of Pharmacy Practice, SRM (Sri Ramaswamy Memorial) College of Pharmacy, Faculty of Medical and Health Sciences, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Sarumathy Sundararajan
- Department of Pharmacy Practice, SRM (Sri Ramaswamy Memorial) College of Pharmacy, Faculty of Medical and Health Sciences, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Nanda Kumar R
- Department of General Medicine, SRM (Sri Ramaswamy Memorial) Medical College Hospital and Research Centre, Chengalpattu, IND
| |
Collapse
|
27
|
Shah S, Clarke L, Davis MW, Topal JE, Shields RK. Clinical manifestations and treatment outcomes for patients with Pseudomonas endocarditis. J Antimicrob Chemother 2024; 79:2017-2021. [PMID: 38958234 DOI: 10.1093/jac/dkae205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVES To investigate clinical outcomes of patients with Pseudomonas endocarditis and identify factors associated with treatment failure. METHODS Adult patients meeting definitive Duke's criteria for Pseudomonas endocarditis at 11 hospitals were identified between May 2000 and February 2024. Failure was defined as death or microbiological failure by day 42. First-line therapy consisted of cefepime, piperacillin/tazobactam, ceftazidime or ceftolozane/tazobactam alone or in combination. RESULTS Forty-eight patients met inclusion criteria; 29% were persons who inject drugs and 13% were organ transplant recipients. Pseudomonas aeruginosa was the causative species in 98% of cases. Patients who experienced 42 day cure were more likely to be initially managed with first-line β-lactam agents compared with those who experienced clinical failure (97% versus 62%, P = 0.004). Treatment with first-line β-lactams was associated with shorter time to treatment initiation and a lower likelihood of infection due to MDR Pseudomonas spp. In the univariate model, patients who experienced 90 day mortality were more likely to have prosthetic valve endocarditis (57% versus 24%, P = 0.02), an intracardiac complication (36% versus 9%, P = 0.04) and a higher median (IQR) Pitt bacteraemia score [2.5 (2-3.8) versus 1 (0-2), P = 0.048]. Combination therapy did not improve clinical outcomes but did increase the rate of adverse effects resulting in drug discontinuation compared with monotherapy, (21% versus 0%, P = 0.08). CONCLUSIONS This is the largest study of Pseudomonas endocarditis to date. We identified improved clinical outcomes when cefepime, piperacillin/tazobactam, ceftazidime or ceftolozane/tazobactam were used for initial treatment. We did not identify a clinical benefit for combination treatment.
Collapse
Affiliation(s)
- Sunish Shah
- Antibiotic Management Program, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Pharmacy, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Pharmacy, Yale New Haven Hospital, New Haven, CT, USA
| | - Lloyd Clarke
- Department of Pharmacy, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Matthew W Davis
- Department of Pharmacy, Yale New Haven Hospital, New Haven, CT, USA
| | - Jeffrey E Topal
- Department of Pharmacy, Yale New Haven Hospital, New Haven, CT, USA
| | - Ryan K Shields
- Antibiotic Management Program, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Janssen A, Bennis FC, Cnossen MH, Mathôt RAA. On inductive biases for the robust and interpretable prediction of drug concentrations using deep compartment models. J Pharmacokinet Pharmacodyn 2024; 51:355-366. [PMID: 38532084 PMCID: PMC11255087 DOI: 10.1007/s10928-024-09906-x] [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: 11/17/2023] [Accepted: 02/09/2024] [Indexed: 03/28/2024]
Abstract
Conventional pharmacokinetic (PK) models contain several useful inductive biases guiding model convergence to more realistic predictions of drug concentrations. Implementing similar biases in standard neural networks can be challenging, but might be fundamental for model robustness and predictive performance. In this study, we build on the deep compartment model (DCM) architecture by introducing constraints that guide the model to explore more physiologically realistic solutions. Using a simulation study, we show that constraints improve robustness in sparse data settings. Additionally, predicted concentration-time curves took on more realistic shapes compared to unconstrained models. Next, we propose the use of multi-branch networks, where each covariate can be connected to specific PK parameters, to reduce the propensity of models to learn spurious effects. Another benefit of this architecture is that covariate effects are isolated, enabling model interpretability through the visualization of learned functions. We show that all models were sensitive to learning false effects when trained in the presence of unimportant covariates, indicating the importance of selecting an appropriate set of covariates to link to the PK parameters. Finally, we compared the predictive performance of the constrained models to previous relevant population PK models on a real-world data set of 69 haemophilia A patients. Here, constrained models obtained higher accuracy compared to the standard DCM, with the multi-branch network outperforming previous PK models. We conclude that physiological-based constraints can improve model robustness. We describe an interpretable architecture which aids model trust, which will be key for the adoption of machine learning-based models in clinical practice.
Collapse
Affiliation(s)
- Alexander Janssen
- Department of Clinical Pharmacology, Hospital Pharmacy, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Frank C Bennis
- Follow Me & Emma Neuroscience Group, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Marjon H Cnossen
- Department of Pediatric Hematology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ron A A Mathôt
- Department of Clinical Pharmacology, Hospital Pharmacy, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
van Gelder TG, Schweitzer VA, Uijtendaal EV, Sikma MA. Catching the Culprit: Benzylpenicillin Neurotoxicity Confirmed by Therapeutic Drug Monitoring in a Critically Ill Patient With Continuous Venovenous Hemofiltration. Ther Drug Monit 2024; 46:556-558. [PMID: 38840334 PMCID: PMC11232934 DOI: 10.1097/ftd.0000000000001215] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 06/07/2024]
Abstract
ABSTRACT We present the case of a 65-year-old patient who was treated with high-dose benzylpenicillin for severe invasive pneumococcal pneumonia, complicated by acute renal failure managed with continuous venovenous hemofiltration. After cessation of continuous venovenous hemofiltration, the patient experienced multiple tonic-clonic seizures. Therapeutic drug monitoring revealed high total serum concentrations of benzylpenicillin, identifying it as the likely cause of the neurotoxicity. This case study presents the first documented total serum benzylpenicillin concentration associated with neurotoxicity.
Collapse
Affiliation(s)
- Thomas G. van Gelder
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Valentijn A. Schweitzer
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; and
| | - Esther V. Uijtendaal
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maaike A. Sikma
- Intensive Care and Dutch Poisons Information Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
30
|
Wessel RJ, Rivera CG, Ausman SE, Martin N, Braga SA, Hagy NT, Moreland-Head LN, Abu Saleh OM, Gajic O, Jannetto PJ, Barreto EF. Use of the DMAIC Lean Six Sigma quality improvement framework to improve beta-lactam antibiotic adequacy in the critically ill. Int J Qual Health Care 2024; 36:mzae062. [PMID: 38955670 PMCID: PMC11439991 DOI: 10.1093/intqhc/mzae062] [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: 11/28/2023] [Revised: 04/12/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024] Open
Abstract
Beta-lactam antibiotics are widely used in the intensive care unit due to their favorable effectiveness and safety profiles. Beta-lactams given to patients with sepsis must be delivered as soon as possible after infection recognition (early), treat the suspected organism (appropriate), and be administered at a dose that eradicates the infection (adequate). Early and appropriate antibiotic delivery occurs in >90% of patients, but less than half of patients with sepsis achieve adequate antibiotic exposure. This project aimed to address this quality gap and improve beta-lactam adequacy using the Define, Measure, Analyze, Improve, and Control Lean Six Sigma quality improvement framework. A multidisciplinary steering committee was formed, which completed a stakeholder analysis to define the gap in practice. An Ishikawa cause and effect (Fishbone) diagram was used to identify the root causes and an impact/effort grid facilitated prioritization of interventions. An intervention that included bundled education with the use of therapeutic drug monitoring (TDM; i.e. drug-level testing) was projected to have the highest impact relative to the amount of effort and selected to address beta-lactam inadequacy in the critically ill. The education and TDM intervention were deployed through a Plan, Do, Study, Act cycle. In the 3 months after "go-live," 54 episodes of beta-lactam TDM occurred in 41 unique intensive care unit patients. The primary quality metric of beta-lactam adequacy was achieved in 94% of individuals after the intervention. Ninety-four percent of clinicians gauged the education provided as sufficient. The primary counterbalance of antimicrobial days of therapy, a core antimicrobial stewardship metric, was unchanged over time (favorable result; P = .73). Application of the Define, Measure, Analyze, Improve, and Control Lean Six Sigma quality improvement framework effectively improved beta-lactam adequacy in critically ill patients. The approach taken in this quality improvement project is widely generalizable to other drugs, drug classes, or settings to increase the adequacy of drug exposure.
Collapse
Affiliation(s)
- Rebecca J Wessel
- Strategy Department, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Christina G Rivera
- Department of Pharmacy, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Sara E Ausman
- Department of Pharmacy, Mayo Clinic Health System, 733 W Clairemont Ave, Eau Claire, WI 54701, United States
| | - Nathaniel Martin
- Department of Pharmacy, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Shienna A Braga
- Department of Pharmacy, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Natalie T Hagy
- Department of Pharmacy, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Lindsay N Moreland-Head
- Indiana University School of Medicine, 340 West 10th Street, Fairbanks Hall, Suite 6200, Indianapolis, IN 46202, United States
| | - Omar M Abu Saleh
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Ognjen Gajic
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Paul J Jannetto
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| | - Erin F Barreto
- Department of Pharmacy, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, United States
| |
Collapse
|
31
|
Roger C. Understanding antimicrobial pharmacokinetics in critically ill patients to optimize antimicrobial therapy: A narrative review. JOURNAL OF INTENSIVE MEDICINE 2024; 4:287-298. [PMID: 39035618 PMCID: PMC11258509 DOI: 10.1016/j.jointm.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 07/23/2024]
Abstract
Effective treatment of sepsis not only demands prompt administration of appropriate antimicrobials but also requires precise dosing to enhance the likelihood of patient survival. Adequate dosing refers to the administration of doses that yield therapeutic drug concentrations at the infection site. This ensures a favorable clinical and microbiological response while avoiding antibiotic-related toxicity. Therapeutic drug monitoring (TDM) is the recommended approach for attaining these goals. However, TDM is not universally available in all intensive care units (ICUs) and for all antimicrobial agents. In the absence of TDM, healthcare practitioners need to rely on several factors to make informed dosing decisions. These include the patient's clinical condition, causative pathogen, impact of organ dysfunction (requiring extracorporeal therapies), and physicochemical properties of the antimicrobials. In this context, the pharmacokinetics of antimicrobials vary considerably between different critically ill patients and within the same patient over the course of ICU stay. This variability underscores the need for individualized dosing. This review aimed to describe the main pathophysiological changes observed in critically ill patients and their impact on antimicrobial drug dosing decisions. It also aimed to provide essential practical recommendations that may aid clinicians in optimizing antimicrobial therapy among critically ill patients.
Collapse
Affiliation(s)
- Claire Roger
- Department of Anesthesiology and Intensive Care, Pain and Emergency Medicine, Nîmes-Caremeau University Hospital, Nîmes, France
- UR UM 103 IMAGINE (Initial Management and prévention of orGan failures IN critically ill patiEnts), Faculty of Medicine, Montpellier University, Montpellier, France
| |
Collapse
|
32
|
Pai Mangalore R, Chai MG, Pope J, Lee SJ, Padiglione A, Diehl A, Roberts L, Sim K, Rawson-Harris P, Wicha S, Schneider HG, Peel TN, Jenney A, Ayton D, Peleg AY, Udy AA. Study protocol for ADAPT-TDM: A beta-lactam antibiotic Dose AdaPtation feasibility randomised controlled Trial using Therapeutic Drug Monitoring. BMJ Open 2024; 14:e083635. [PMID: 38951004 PMCID: PMC11328624 DOI: 10.1136/bmjopen-2023-083635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/02/2024] [Indexed: 07/03/2024] Open
Abstract
INTRODUCTION Critically ill patients are at risk of suboptimal beta-lactam antibiotic (beta-lactam) exposure due to the impact of altered physiology on pharmacokinetics. Suboptimal concentrations can lead to treatment failure or toxicity. Therapeutic drug monitoring (TDM) involves adjusting doses based on measured plasma concentrations and individualising dosing to improve the likelihood of improving exposure. Despite its potential benefits, its adoption has been slow, and data on implementation, dose adaptation and safety are sparse. The aim of this trial is to assess the feasibility and fidelity of implementing beta-lactam TDM-guided dosing in the intensive care unit setting. METHODS AND ANALYSIS A beta-lactam antibiotic Dose AdaPtation feasibility randomised controlled Trial using Therapeutic Drug Monitoring (ADAPT-TDM) is a single-centre, unblinded, feasibility randomised controlled trial aiming to enroll up to 60 critically ill adult participants (≥18 years). TDM and dose adjustment will be performed daily in the intervention group; the standard of care group will undergo plasma sampling, but no dose adjustment. The main outcomes include: (1) feasibility of recruitment, defined as the number of participants who are recruited from a pool of eligible participants, and (2) fidelity of TDM, defined as the degree to which TDM as a test is delivered as intended, from accurate sample collection, sample processing to result availability. Secondary outcomes include target attainment, uptake of TDM-guided dosing and incidence of neurotoxicity, hepatotoxicity and nephrotoxicity. ETHICS AND DISSEMINATION This study has been approved by the Alfred Hospital human research ethics committee, Office of Ethics and Research Governance (reference: Project No. 565/22; date of approval: 22/11/2022). Prospective consent will be obtained and the study will be conducted in accordance with the Declaration of Helsinki. The finalised manuscript, including aggregate data, will be submitted for publication in a peer reviewed journal. ADAPT-TDM will determine whether beta-lactam TDM-guided dose adaptation is reproducible and feasible and provide important information required to implement this intervention in a phase III trial. TRIAL REGISTRATION NUMBER Australian New Zealand Clinical Trials Registry, ACTRN12623000032651.
Collapse
Affiliation(s)
- Rekha Pai Mangalore
- Infectious Disease, Alfred Health, Melbourne, Victoria, Australia
- Infectious Diseases, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Ming Gene Chai
- Pharmacy Department, Alfred Health, Melbourne, Victoria, Australia
- Department of Intensive Care and Hyperbaric Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Jeffrey Pope
- Department of Pathology, Alfred Health, Melbourne, Victoria, Australia
| | - Sue J Lee
- Department of Infectious Diseases, Monash University, Clayton, Victoria, Australia
- Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Alexander Padiglione
- Infectious Disease, Alfred Health, Melbourne, Victoria, Australia
- Infectious Diseases, Monash Health, Clayton, Victoria, Australia
| | - Arne Diehl
- Department of Intensive Care and Hyperbaric Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Llyod Roberts
- Department of Intensive Care and Hyperbaric Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Kirsty Sim
- Infectious Disease, Alfred Health, Melbourne, Victoria, Australia
- Infectious Diseases, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Philip Rawson-Harris
- Infectious Disease, Alfred Health, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Clayton, Victoria, Australia
| | - Sebastian Wicha
- Department of Clinical Pharmacology, University of Hamburg, Hamburg, Hamburg, Germany
| | - Hans G Schneider
- Department of Pathology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Trish N Peel
- Infectious Diseases, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Adam Jenney
- Infectious Disease, Alfred Health, Melbourne, Victoria, Australia
- Department of Pathology, Alfred Health, Melbourne, Victoria, Australia
| | - Darshini Ayton
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, Victoria, Australia
- Health and Social Care Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anton Y Peleg
- Infectious Diseases, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Andrew A Udy
- Department of Intensive Care and Hyperbaric Medicine, Alfred Health, Melbourne, Victoria, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
33
|
De Sutter PJ, Hermans E, De Cock P, Van Bocxlaer J, Gasthuys E, Vermeulen A. Penetration of Antibiotics into Subcutaneous and Intramuscular Interstitial Fluid: A Meta-Analysis of Microdialysis Studies in Adults. Clin Pharmacokinet 2024; 63:965-980. [PMID: 38955946 DOI: 10.1007/s40262-024-01394-z] [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: 06/03/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND OBJECTIVE The interstitial fluid of tissues is the effect site for antibiotics targeting extracellular pathogens. Microdialysis studies investigating these concentrations in muscle and subcutaneous tissue have reported notable variability in tissue penetration. This study aimed to comprehensively summarise the existing data on interstitial fluid penetration in these tissues and to identify potential factors influencing antibiotic distribution. METHODS A literature review was conducted, focusing on subcutaneous and intramuscular microdialysis studies of antibiotics in both adult healthy volunteers and patients. Random-effect meta-analyses were used to aggregate effect size estimates of tissue penetration. The primary parameter of interest was the unbound penetration ratio, which represents the ratio of the area under the concentration-time curve in interstitial fluid relative to the area under the concentration-time curve in plasma, using unbound concentrations. RESULTS In total, 52 reports were incorporated into this analysis. The unbound antibiotic exposure in the interstitial fluid of healthy volunteers was, on average, 22% lower than in plasma. The unbound penetration ratio values were higher after multiple dosing but did not significantly differ between muscle and subcutaneous tissue. Unbound penetration ratio values were lower for acids and bases compared with neutral antibiotics. Neither the molecular weight nor the logP of the antibiotics accounted for the variations in the unbound penetration ratio. Obesity was associated with lower interstitial fluid penetration. Conditions such as sepsis, tissue inflammation and tissue ischaemia were not significantly associated with altered interstitial fluid penetration. CONCLUSIONS This study highlights the variability and generally lower exposure of unbound antibiotics in the subcutaneous and intramuscular interstitial fluid compared with exposure in plasma. Future research should focus on understanding the therapeutic relevance of these differences and identify key covariates that may influence them.
Collapse
Affiliation(s)
- Pieter-Jan De Sutter
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| | - Eline Hermans
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Pieter De Cock
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
- Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Jan Van Bocxlaer
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Elke Gasthuys
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| |
Collapse
|
34
|
Tang BH, Yao BF, Zhang W, Zhang XF, Fu SM, Hao GX, Zhou Y, Sun DQ, Liu G, van den Anker J, Wu YE, Zheng Y, Zhao W. Optimal use of β-lactams in neonates: machine learning-based clinical decision support system. EBioMedicine 2024; 105:105221. [PMID: 38917512 PMCID: PMC467072 DOI: 10.1016/j.ebiom.2024.105221] [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: 11/01/2023] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Accurate prediction of the optimal dose for β-lactam antibiotics in neonatal sepsis is challenging. We aimed to evaluate whether a reliable clinical decision support system (CDSS) based on machine learning (ML) can assist clinicians in making optimal dose selections. METHODS Five β-lactam antibiotics (amoxicillin, ceftazidime, cefotaxime, meropenem and latamoxef), commonly used to treat neonatal sepsis, were selected. The CDSS was constructed by incorporating the drug, patient, dosage, pharmacodynamic, and microbiological factors. The CatBoost ML algorithm was used to build the CDSS. Real-world studies were used to evaluate the CDSS performance. Virtual trials were used to compare the CDSS-optimized doses with guideline-recommended doses. FINDINGS For a specific drug, by entering the patient characteristics and pharmacodynamic (PD) target (50%/70%/100% fraction of time that the free drug concentration is above the minimal inhibitory concentration [fT > MIC]), the CDSS can determine whether the planned dosing regimen will achieve the PD target and suggest an optimal dose. The prediction accuracy of all five drugs was >80.0% in the real-world validation. Compared with the PopPK model, the overall accuracy, precision, recall, and F1-Score improved by 10.7%, 22.1%, 64.2%, and 43.1%, respectively. Using the CDSS-optimized doses, the average probability of target concentration attainment increased by 58.2% compared to the guideline-recommended doses. INTERPRETATION An ML-based CDSS was successfully constructed to assist clinicians in selecting optimal β-lactam antibiotic doses. FUNDING This work was supported by the National Natural Science Foundation of China; Distinguished Young and Middle-aged Scholar of Shandong University; National Key Research and Development Program of China.
Collapse
Affiliation(s)
- Bo-Hao Tang
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin-Fang Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shu-Meng Fu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - De-Qing Sun
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gang Liu
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA; Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA; Department of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China.
| |
Collapse
|
35
|
Karaba SM, Cosgrove SE, Lee JH, Fiawoo S, Heil EL, Quartuccio KS, Shihadeh KC, Tamma PD. Extended-Infusion β-Lactam Therapy, Mortality, and Subsequent Antibiotic Resistance Among Hospitalized Adults With Gram-Negative Bloodstream Infections. JAMA Netw Open 2024; 7:e2418234. [PMID: 38954416 PMCID: PMC11220563 DOI: 10.1001/jamanetworkopen.2024.18234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/21/2024] [Indexed: 07/04/2024] Open
Abstract
Importance Current evidence is conflicting for associations of extended-infusion β-lactam (EI-BL) therapy with clinical outcomes. Objective To investigate the association of EI-BL therapy with survival, adverse events, and emergence of antibiotic resistance in adults with gram-negative bloodstream infections (GN-BSI). Design, Setting, and Participants This cohort study of consecutive adults with GN-BSI admitted to 24 United States hospitals between January 1, 2019, and December 31, 2019, receiving EI-BL were compared with adults with GN-BSI receiving the same agents as intermittent infusion β-lactam (II-BL; ≤1-hour infusions). Statistical analysis was performed from January to October 2023. Exposures EI-BL (ie, ≥3-hour infusion). Main Outcomes and Measures EI-BL and II-BL groups underwent 1:3 nearest-neighbor propensity score matching (PSM) without replacement. Multivariable regression was applied to the PSM cohort to investigate outcomes, all censored at day 90. The primary outcome was mortality; secondary outcomes included antibiotic adverse events and emergence of resistance (≥4-fold increase in the minimum inhibitory concentration of the β-lactam used to treat the index GN-BSI). Results Among the 4861 patients included, 2547 (52.4%) were male; and the median (IQR) age was 67 (55-77) years. There were 352 patients in the EI-BL 1:3 PSM group, and 1056 patients in the II-BL 1:3 PSM group. Among 1408 PSM patients, 373 (26.5%) died by day 90. The odds of mortality were lower in the EI-BL group (adjusted odds ratio [aOR], 0.71 [95% CI, 0.52-0.97]). In a stratified analysis, a survival benefit was only identified in patients with severe illness or elevated minimum inhibitory concentrations (ie, in the intermediate range for the antibiotic administered). There were increased odds of catheter complications (aOR, 3.14 [95% CI, 1.66-5.96]) and antibiotic discontinuation because of adverse events (eg, acute kidney injury, cytopenias, seizures) in the EI-BL group (aOR, 3.66 [95% CI, 1.68-7.95]). Emergence of resistance was similar in the EI-BL and II-BL groups at 2.9% vs 7.2%, respectively (P = .35). Conclusions and Relevance In this cohort study of patients with GN-BSI, EI-BL therapy was associated with reduced mortality for patients with severe illness or those infected with nonsusceptible organisms; potential advantages in other groups remain unclear and need to be balanced with potential adverse events. The subsequent emergence of resistance warrants investigation in a larger cohort.
Collapse
Affiliation(s)
- Sara M. Karaba
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sara E. Cosgrove
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jae Hyoung Lee
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Suiyini Fiawoo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emily L. Heil
- Department of Practice, Sciences, and Health-Outcomes Research, University of Maryland School of Pharmacy, Baltimore, Maryland
| | | | | | - Pranita D. Tamma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Alsultan A, Aldawsari MR, Alturaiq NK, Syed SA, Alsubai A, Kurdee Z, Alsubaie S, Alqahtani S, Abouelkheir M. Evaluation of pharmacokinetic pharmacodynamic target attainment of meropenem in pediatric patients. Pediatr Neonatol 2024; 65:386-390. [PMID: 38218717 DOI: 10.1016/j.pedneo.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/27/2023] [Accepted: 09/08/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Meropenem is a widely used carbapenem for treating severe pediatric infections. However, few studies have assessed its pharmacokinetics/pharmacodynamics (PK/PD) in pediatric patients. This study aimed to evaluate the proportion of Saudi pediatric patients achieving the PK/PD target of meropenem. METHODS A prospective observational study was conducted at King Saud University Medical City from July to September 2022. Pediatric patients receiving meropenem for suspected or proven infections were included in the study. The primary outcome was the percentage of patients achieving the recommended PK/PD target for critically ill or non-critically ill pediatric patients. RESULTS The study included 30 patients (nine neonates and 21 older pediatric patients). All neonates were critically ill. Among them, 55 % achieved the PK/PD target of 100 % free time above the MIC. In older ICU pediatric patients, only 11 % attained this target, whereas 58 % of older pediatrics in the general wards achieved the PK/PD target of 50 % free time above the MIC. Augmented renal clearance (ARC) was identified in 57 % of our pediatric patient population, none of whom achieved the recommended PK/PD targets. The median trough concentrations in patients with and without ARC were 0.75 and 1.3 μg/mL, respectively (P < 0.05). CONCLUSIONS The majority of patients in our cohort did not achieve the PK/PD target for meropenem. ARC emerged as a major risk factor for target attainment failure in both critically ill and non-critically ill pediatric patients.
Collapse
Affiliation(s)
- Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia.
| | - Maram R Aldawsari
- Department of Pharmacy, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Nujood Khaled Alturaiq
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Ali Syed
- Department of Pharmaceutical, Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alsubai
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Zeyad Kurdee
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Saudi Arabia
| | - Sarah Alsubaie
- Pediatric Infectious Disease Unit, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Manal Abouelkheir
- Department of Clinical Pharmacy, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| |
Collapse
|
37
|
Wu Y, Lu Z, Liang P, Zhu H, Qi H, Zhang H. Relationship of imipenem therapeutic drug monitoring to clinical outcomes in critically ill patients: a retrospective cohort study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4791-4798. [PMID: 38153513 DOI: 10.1007/s00210-023-02909-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023]
Abstract
The primary objective of this study was to evaluate the predictors associated with target concentration (non-)attainment of imipenem in critically ill patients. The secondary objective was to explore the correlation between achieving imipenem target concentrations and clinical outcomes of therapy. A retrospective cohort study was conducted in critically ill patients treated with imipenem. Clinical data were extracted from the patients' electronic medical records. The pharmacokinetic/pharmacodynamic target was defined as free imipenem concentrations above the minimum inhibitory concentration (MIC) of the pathogen at 100% (100%fT>MIC) of the dosing interval. Factors associated with the non-attainment of target concentrations were evaluated using binomial logistic regression. Kaplan-Meier analysis was used to investigate the correlation between (non-)attainment targets and 30-day mortality. A total of 406 patients were included, and 55.4% achieved the target of 100%fT>MIC. Regression analysis identified an initial daily dose of imipenem ≤ 2 g/day, augmented renal clearance, age ≤ 60 years, recent surgery, and absence of positive microbiology culture as risk factors for target non-attainment. Achieving the 100%fT>MIC target was significantly associated with clinical efficacy but not with 30-day mortality. Selective application of therapeutic drug monitoring in the early stages of imipenem treatment for critically ill patients can improve clinical outcomes. Further research should explore the potential benefits of TDM-guided dosing strategies for imipenem in critical care settings.
Collapse
Affiliation(s)
- Yejing Wu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhangyang Lu
- Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Pei Liang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Huaijun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Hui Qi
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Haixia Zhang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China.
| |
Collapse
|
38
|
Johns K, Eschenauer G, Clark A, Butler S, Dunham S. Antimicrobial Pharmacokinetic Considerations in Extracorporeal Membrane Oxygenation. J Clin Med 2024; 13:3554. [PMID: 38930083 PMCID: PMC11204421 DOI: 10.3390/jcm13123554] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Critical illness creates challenges for healthcare providers in determining the optimal treatment of severe disease, particularly in determining the most appropriate selection and dosing of medications. Critically ill patients experience endogenous physiologic changes that alter the pharmacokinetics (PKs) of medications. These alterations can be further compounded by mechanical support modalities such as extracorporeal membrane oxygenation (ECMO). Specific components of the ECMO circuit have the potential to affect drug PKs through drug sequestration and an increase in the volume of distribution. Factors related to the medications themselves also play a role. These PK alterations create problems when trying to properly utilize antimicrobials in this patient population. The literature seeking to identify appropriate antimicrobial dosing regimens is both limited and difficult to evaluate due to patient variability and an inability to determine the exact role of the ECMO circuit in reduced drug concentrations. Lipophilic and highly protein bound medications are considered more likely to undergo significant drug sequestration in an ECMO circuit, and this general trend represents a logical starting point in antimicrobial selection and dosing in patients on ECMO support. This should not be the only consideration, however, as identifying infection and evaluating the efficacy of treatments in this population is challenging. Due to these challenges, therapeutic drug monitoring should be utilized whenever possible, particularly in cases with severe infection or high concern for drug toxicity.
Collapse
Affiliation(s)
- Kevin Johns
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI 48109, USA; (G.E.); (A.C.); (S.B.)
| | - Gregory Eschenauer
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI 48109, USA; (G.E.); (A.C.); (S.B.)
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Angela Clark
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI 48109, USA; (G.E.); (A.C.); (S.B.)
| | - Simona Butler
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI 48109, USA; (G.E.); (A.C.); (S.B.)
| | - Sabrina Dunham
- Department of Pharmacy Services, University of Michigan Health, Ann Arbor, MI 48109, USA; (G.E.); (A.C.); (S.B.)
| |
Collapse
|
39
|
Dilawri A, Muir J, Brodie D, Abrams D, Agerstrand C, Madahar P, Dzierba AL. Practices surrounding antimicrobial use in patients managed with extracorporeal membrane oxygenation: An international survey. J Crit Care 2024; 81:154534. [PMID: 38367526 DOI: 10.1016/j.jcrc.2024.154534] [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: 11/14/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/19/2024]
Abstract
PURPOSE This study aimed to survey critical care clinicians and characterize their perception of antimicrobial dosing strategies in patients receiving extracorporeal membrane oxygenation (ECMO). METHODS International, cross-sectional survey distributed to members of the Society of Critical Care Medicine in October 2022. RESULTS Respondents were primarily physicians (45%), with 92% practicing in North America. Ninety-seven percent of respondents reported antimicrobial dosing in critically ill patients to be challenging, due to physiological derangements seen in the patient population. Eighty-seven percent reported consideration of physicochemical drug properties when dosing antimicrobials in ECMO-supported patients, with lipophilicity (83%) and degree of protein binding (74%) being the two most common. Respondents' approach to antimicrobial dosing strategies did not significantly differ in critically ill ECMO-supported patients, compared to patients with equal severity of illness not receiving ECMO support. CONCLUSION Approaches to antimicrobial dosing strategies do not significantly differ among respondents between critically ill patients on ECMO support, compared to patients with equal severity of illness not receiving ECMO support. These findings were unexpected considering the added physiologic complexity of the ECMO circuit to critically ill adult patients and the need for well designed and adequately powered studies to inform empiric dosing guidance for ECMO-supported patients.
Collapse
Affiliation(s)
- Atul Dilawri
- Department of Pharmacy, NewYork-Presbyterian Hospital, New York, NY, United States of America
| | - Justin Muir
- Department of Pharmacy, NewYork-Presbyterian Hospital, New York, NY, United States of America
| | - Daniel Brodie
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Darryl Abrams
- Division of Pulmonary and Critical Care Medicine, Columbia College of Physicians and Surgeons/NewYork-Presbyterian Hospital, New York, NY, United States of America
| | - Cara Agerstrand
- Division of Pulmonary and Critical Care Medicine, Columbia College of Physicians and Surgeons/NewYork-Presbyterian Hospital, New York, NY, United States of America
| | - Purnema Madahar
- Division of Pulmonary and Critical Care Medicine, Columbia College of Physicians and Surgeons/NewYork-Presbyterian Hospital, New York, NY, United States of America
| | - Amy L Dzierba
- Department of Pharmacy, NewYork-Presbyterian Hospital, New York, NY, United States of America; Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons and NewYork-Presbyterian Hospital, New York, NY, United States of America.
| |
Collapse
|
40
|
Miano TA, Barreto EF, McNett M, Martin N, Sakhuja A, Andrews A, Basu RK, Ablordeppey EA. Toward Equitable Kidney Function Estimation in Critical Care Practice: Guidance From the Society of Critical Care Medicine's Diversity, Equity, and Inclusion in Renal Clinical Practice Task Force. Crit Care Med 2024; 52:951-962. [PMID: 38407240 PMCID: PMC11098700 DOI: 10.1097/ccm.0000000000006237] [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] [Indexed: 02/27/2024]
Abstract
OBJECTIVES Accurate glomerular filtration rate (GFR) assessment is essential in critically ill patients. GFR is often estimated using creatinine-based equations, which require surrogates for muscle mass such as age and sex. Race has also been included in GFR equations, based on the assumption that Black individuals have genetically determined higher muscle mass. However, race-based GFR estimation has been questioned with the recognition that race is a poor surrogate for genetic ancestry, and racial health disparities are driven largely by socioeconomic factors. The American Society of Nephrology and the National Kidney Foundation (ASN/NKF) recommend widespread adoption of new "race-free" creatinine equations, and increased use of cystatin C as a race-agnostic GFR biomarker. DATA SOURCES Literature review and expert consensus. STUDY SELECTION English language publications evaluating GFR assessment and racial disparities. DATA EXTRACTION We provide an overview of the ASN/NKF recommendations. We then apply an Implementation science methodology to identify facilitators and barriers to implementation of the ASN/NKF recommendations into critical care settings and identify evidence-based implementation strategies. Last, we highlight research priorities for advancing GFR estimation in critically ill patients. DATA SYNTHESIS Implementation of the new creatinine-based GFR equation is facilitated by low cost and relative ease of incorporation into electronic health records. The key barrier to implementation is a lack of direct evidence in critically ill patients. Additional barriers to implementing cystatin C-based GFR estimation include higher cost and lack of test availability in most laboratories. Further, cystatin C concentrations are influenced by inflammation, which complicates interpretation. CONCLUSIONS The lack of direct evidence in critically ill patients is a key barrier to broad implementation of newly developed "race-free" GFR equations. Additional research evaluating GFR equations in critically ill patients and novel approaches to dynamic kidney function estimation is required to advance equitable GFR assessment in this vulnerable population.
Collapse
Affiliation(s)
- Todd A. Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Erin F. Barreto
- Department of Pharmacy, Mayo Clinic, Rochester, MN, United States of America
| | - Molly McNett
- College of Nursing, The Ohio State University, Columbus, Ohio
| | - Niels Martin
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ankit Sakhuja
- Division of Data Driven and Digital Medicine, The Charles Bronfman Institute for Personalized Medicine and Institute for Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adair Andrews
- Society of Critical Care Medicine, Mount Prospect, IL
| | - Rajit K. Basu
- Ann & Robert Lurie Children’s Hospital of Chicago, Northwestern University, Chicago, IL, USA
| | - Enyo Ama Ablordeppey
- Washington University School of Medicine, Department of Anesthesiology and Emergency Medicine, St. Louis, Missouri
| |
Collapse
|
41
|
Damgaard T, Woksepp H, Brudin L, Bonnedahl J, Nielsen EI, Schön T, Hällgren A. Estimated glomerular filtration rate as a tool for early identification of patients with insufficient exposure to beta-lactam antibiotics in intensive care units. Infect Dis (Lond) 2024; 56:451-459. [PMID: 38436273 DOI: 10.1080/23744235.2024.2323002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Only about 50% of intensive care unit (ICU) patients reach a free trough concentration above MIC (100% fT > MIC) of beta-lactam antibiotics. Although dose adjustments based on therapeutic drug monitoring (TDM) could be beneficial, TDM is not widely available. We investigated serum creatinine-based estimated GFR (eGFR) as a rapid screening tool to identify ICU patients at risk of insufficient exposure. METHOD Ninety-three adult patients admitted to four ICUs in southeast Sweden treated with piperacillin/tazobactam, meropenem, or cefotaxime were included. Beta-lactam trough concentrations were measured. The concentration target was set to 100% fT > MICECOFF (2, 4, and 16 mg/L based on calculated free levels for meropenem, cefotaxime, and piperacillin, respectively). eGFR was primarily determined via Chronic Kidney Disease-Epidemiology Collaboration (CKD-EPI) and compared to three other eGFR equations. Data was analysed using logistic regression and receiver operative characteristic (ROC) curves. RESULTS With intermittent standard dosing, insufficient exposure was common in patients with a relative eGFR ≥48mL/min/1.73m2 [85%, (45/53)], particularly when treated with cefotaxime [96%, (24/25)]. This eGFR cut-off had a sensitivity of 92% and specificity of 82% (AUC 0.871, p < 0.001) in identifying insufficient exposure. In contrast, patients with eGFR <48mL/min/1.73m2 had high target attainment [90%, (36/40)] with a wide variability in drug exposure. There was no difference between the four eGFR equations (AUC 0.866-0.872, cut-offs 44-51 ml/min/1.73m2). CONCLUSION Serum creatinine-based eGFR is a simple and widely available surrogate marker with potential for early identification of ICU patients at risk of insufficient exposure to piperacillin, meropenem, and cefotaxime.
Collapse
Affiliation(s)
- Tobias Damgaard
- Pharmaceutical Department in Kalmar, Region Kalmar County, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hanna Woksepp
- Department of Research and Department of Clinical Microbiology in Kalmar, Region Kalmar County, and Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Lars Brudin
- Department of Clinical Physiology in Kalmar, Region Kalmar County, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Jonas Bonnedahl
- Department of Infectious Diseases in Kalmar, Region Kalmar County, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Thomas Schön
- Department of Infectious Diseases in Kalmar, Region Kalmar County, Department of Infectious Diseases in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anita Hällgren
- Department of Infectious Diseases in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
42
|
Butragueño-Laiseca L, Troconiz IF, Grau S, Campillo N, Padilla B, Fernández SN, Slöcker M, Herrera L, Santiago MJ. How to use meropenem in pediatric patients undergoing CKRT? Integrated meropenem pharmacokinetic model for critically ill children. Antimicrob Agents Chemother 2024:e0172923. [PMID: 38656186 DOI: 10.1128/aac.01729-23] [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: 01/11/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Standard dosing could fail to achieve adequate systemic concentrations in ICU children or may lead to toxicity in children with acute kidney injury. The population pharmacokinetic analysis was used to simultaneously analyze all available data (plasma, prefilter, postfilter, effluent, and urine concentrations) and provide the pharmacokinetic characteristics of meropenem. The probability of target fT > MIC attainment, avoiding toxic levels, during the entire dosing interval was estimated by simulation of different intermittent and continuous infusions in the studied population. A total of 16 critically ill children treated with meropenem were included, with 7 of them undergoing continuous kidney replacement therapy (CKRT). Only 33% of children without CKRT achieved 90% of the time when the free drug concentration exceeded the minimum inhibitory concentration (%fT > MIC) for an MIC of 2 mg/L. In dose simulations, only continuous infusions (60-120 mg/kg in a 24-h infusion) reached the objective in patients <30 kg. In patients undergoing CKRT, the currently used schedule (40 mg/kg/12 h from day 2 in a short infusion of 30 min) was clearly insufficient in patients <30 kg. Keeping the dose to 40 mg/kg q8h without applying renal adjustment and extended infusions (40 mg/kg in 3- or 4-h infusion every 12 h) was sufficient to reach 90% fT > MIC (>2 mg/L) in patients >10 kg. In patients <10 kg, only continuous infusions reached the objective. In patients >30 kg, 60 mg/kg in a 24-h infusion is sufficient and avoids toxicity. This population model could help with an individualized dosing approach that needs to be adopted in critically ill pediatric patients. Critically ill patients subjected to or not to CKRT may benefit from the administration of meropenem in an extended or continuous infusion.
Collapse
Affiliation(s)
- Laura Butragueño-Laiseca
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS) RD21/0012/0011, Carlos III Health Institute, Madrid, Spain
| | - Iñaki F Troconiz
- Pharmacometrics and Systems Pharmacology Research Unit, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Santiago Grau
- Pharmacy Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nuria Campillo
- Pharmacy Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Belén Padilla
- Clinical Microbiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Sarah Nicole Fernández
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS) RD21/0012/0011, Carlos III Health Institute, Madrid, Spain
| | - María Slöcker
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS) RD21/0012/0011, Carlos III Health Institute, Madrid, Spain
| | - Laura Herrera
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS) RD21/0012/0011, Carlos III Health Institute, Madrid, Spain
| | - María José Santiago
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS) RD21/0012/0011, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
43
|
Cizmarova I, Mikus P, Svidrnoch M, Piestansky J. Development and Validation of a Capillary Zone Electrophoresis-Tandem Mass Spectrometry Method for Simultaneous Quantification of Eight β-Lactam Antibiotics and Two β-Lactamase Inhibitors in Plasma Samples. Pharmaceuticals (Basel) 2024; 17:526. [PMID: 38675486 PMCID: PMC11054939 DOI: 10.3390/ph17040526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Monitoring plasma concentrations of β-lactam antibiotics is crucial, particularly in critically ill patients, where variations in concentrations can lead to treatment failure or adverse events. Standardized antimicrobial regimens may not be effective for all patients, especially in special groups with altered physiological parameters. Pharmacokinetic/pharmacodynamic (PK/PD) studies highlight the time-dependent antibacterial activity of these antibiotics, emphasizing the need for personalized dosing. Therapeutic drug monitoring (TDM) is essential, requiring rapid and accurate analytical methods for precise determination of drugs in biological material (typically plasma or serum). This study presents a novel capillary zone electrophoresis-tandem mass spectrometry (CZE-MS/MS) method designed for the simultaneous quantification of five penicillin antibiotics, two cephalosporins, one carbapenem, and two β-lactamase inhibitors in a single run. The method involves a simple sample pretreatment-precipitation with organic solvent-and has a run time of 20 min. Optimization of CZE separation conditions revealed that 20 mM ammonium hydrogen carbonate (NH4HCO3) serves as the optimal background electrolyte (BGE). Positive electrospray ionization (ESI) mode, with isopropyl alcohol (IP)/10 mM ammonium formate water solution (50/50, v/v) as the sheath liquid, was identified as the optimal condition for MS detection. Method validation according to the Food and Drug Administration (FDA) guideline for development of bioanalytical methods demonstrated satisfactory selectivity, linearity, recovery, robustness, and stability. The method's practicality was evaluated using the Blue Applicability Grade Index (BAGI), yielding a score of 77.5. Moreover, the greenness of the proposed method was evaluated by two commonly used metric tools-Analytical GREEnness (AGREE) and Green Analytical Procedure Index (GAPI). The developed CZE-MS/MS method offers a practical and reliable approach for quantifying a broad spectrum of β-lactam antibiotics in plasma. Its ability to simultaneously quantify multiple analytes in a single run, coupled with a straightforward sample pretreatment, positions it as a valuable and prospective tool for TDM in critically ill patients.
Collapse
Affiliation(s)
- Ivana Cizmarova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia; (I.C.); (P.M.)
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia; (I.C.); (P.M.)
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
| | - Martin Svidrnoch
- AGEL Lab, Revolucni 2214/35, CZ-741 01 Novy Jicin, Czech Republic;
| | - Juraj Piestansky
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovakia
| |
Collapse
|
44
|
Razazi K, Berti E, Cecchini J, Carteaux G, Habibi A, Bartolucci P, Arrestier R, Gendreau S, de Prost N, Hulin A, Dessap AM. Decreased risk of underdosing with continuous infusion versus intermittent administration of cefotaxime in patients with sickle cell disease and acute chest syndrome. PLoS One 2024; 19:e0302298. [PMID: 38635540 PMCID: PMC11025818 DOI: 10.1371/journal.pone.0302298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
OBJECTIVE Underdosing of antibiotics is common in patients with sickle cell disease (SCD). We hypothesized that in critically-ill patients with SCD receiving cefotaxime during acute chest syndrome, the continuous infusion may outperform the intermittent administration in achieving pharmacokinetic/pharmacodynamic targets. DESIGN Prospective before-after study. SETTINGS Intensive-care unit of a French teaching hospital and sickle cell disease referral center. PATIENTS Sixty consecutive episodes of severe acute chest syndrome in 58 adult patients with sickle cell disease. INTERVENTIONS Patients were treated with intermittent administration during the first period (April 2016 -April 2018) and with continuous infusion during the second period (May 2018 -August 2019). MEASUREMENTS AND MAIN RESULTS We included 60 episodes of acute chest syndrome in 58 patients (29 [25-34] years, 37/58 (64%) males). Daily dose of cefotaxime was similar between groups (59 [48-88] vs. 61 [57-64] mg/kg/day, p = 0.84). Most patients (>75%) presented a glomerular hyperfiltration with no difference between groups (p = 0.25). More patients had a cefotaxime trough level ≥2 mg/L with continuous infusion than intermittent administration: 28 (93%) vs. 5 (16%), p<0.001. The median residual concentration was higher in the continuous infusion than intermittent administration group: 10.5 [7.4-13.3] vs. 0 [0-0] mg/L, p<0.001. No infection relapse was observed in the entire cohort. Hospital length of stay was similar between groups. CONCLUSION As compared to intermittent administration, continuous infusion of cefotaxime maximizes the pharmacokinetic/pharmacodynamic parameters in patients with SCD. The clinical outcome did not differ between the two administration methods; however, the study was underpowered to detect such a difference.
Collapse
Affiliation(s)
- Keyvan Razazi
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Enora Berti
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Jerome Cecchini
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
- Hôpital Intercommunal de Créteil, Service de Réanimation et Surveillance Continue Adulte, 94000, Créteil, France
| | - Guillaume Carteaux
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Anoosha Habibi
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Centre de Référence de la Drépanocytose, Créteil, France
| | - Pablo Bartolucci
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Centre de Référence de la Drépanocytose, Créteil, France
| | - Romain Arrestier
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Ségolène Gendreau
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Nicolas de Prost
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| | - Anne Hulin
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Biochimie, Créteil, 94010 France
| | - Armand Mekontso Dessap
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, F-94010, France
- Université Paris Est Créteil, CARMAS, Créteil, F-94010, France
| |
Collapse
|
45
|
Areskog Lejbman I, Torisson G, Resman F, Sjövall F. Beta-lactam antibiotic concentrations in critically ill patients with standard and adjusted dosages: A prospective observational study. Acta Anaesthesiol Scand 2024; 68:530-537. [PMID: 38407447 DOI: 10.1111/aas.14382] [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: 10/13/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/27/2024]
Abstract
INTRODUCTION Antibiotic concentration target attainment is known to be poor in critically ill patients. Dose adjustment is recommended in patients with altered clearance, obesity and those with bacterial species with intermediate susceptibility. The aim of this study was to investigate the variation of antibiotic concentration in critically ill patients with standard or adjusted dosing regimens. METHODS The concentration of three beta-lactam antibiotics used in the intensive care unit (ICU) setting, cefotaxime, piperacillin/tazobactam, and meropenem, was measured in patients with confirmed or suspected infection. Mid-dose and trough values were collected during a single dosing interval. The pharmacokinetic endpoints were a free antibiotic concentration that, during the whole dosing interval, was above MIC (100% ƒT > MIC, primary endpoint) or above four times MIC (100% ƒT > 4MIC, secondary endpoint). Non-species related MIC breakpoints were used (1 mg/L for cefotaxime, 8 mg/L for piperacillin/tazobactam, and 2 mg/L for meropenem). RESULTS We included 102 patients (38 cefotaxime, 30 piperacillin/tazobactam, and 34 meropenem) at a single ICU, with a median age of 66 years. In total, 73% were males, 40% were obese (BMI ≥30) and the median SAPS 3 score was 63 points. Of all patients, 78 patients (76%) reached the primary endpoint (100%ƒT > MIC), with 74% for cefotaxime, 67% for piperacillin/tazobactam and 88% for meropenem. Target attainment for 100% ƒT > 4MIC was achieved in 40 (39%) patients, overall, with 34% for cefotaxime, 30% for piperacillin/tazobactam and 53% for meropenem. In patients with standard dose 71% attained 100%ƒT > MIC and 37% for 100%ƒT > 4MIC. All patients with reduced dose attained 100%ƒT > MIC and 27% attained 100% ƒT > 4MIC. In patients with increased dose 79% attained 100%ƒT > MIC and 48% 100%ƒT > 4MIC respectively. CONCLUSIONS Beta-lactam antibiotics concentration vary widely in critically ill patients. The current standard dosing regimens employed during the study were not sufficient to reach 100% ƒT > MIC in approximately a quarter of the patients. In patients where dose adjustment was performed, the group with increased dose also had low target attainment, as opposed to patients with dose reduction, who all reached target. This suggests the need for further individualization of dosing where therapeutic drug monitoring can be an alternative to further increase target attainment.
Collapse
Affiliation(s)
- Ilja Areskog Lejbman
- Department of Intensive and Perioperative Care, Skåne University Hospital, Malmö, Sweden
| | - Gustav Torisson
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | - Fredrik Resman
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | - Fredrik Sjövall
- Department of Intensive and Perioperative Care, Skåne University Hospital, Malmö, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
46
|
El-Haffaf I, Marsot A, Hachemi D, Pesout T, Williams V, Smith MA, Albert M, Williamson D. Exposure levels and target attainment of piperacillin/tazobactam in adult patients admitted to the intensive care unit: a prospective observational study. Can J Anaesth 2024; 71:511-522. [PMID: 38243099 DOI: 10.1007/s12630-023-02689-8] [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: 03/13/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 01/21/2024] Open
Abstract
PURPOSE The objective of this study was to evaluate the exposure and the pharmacodynamic target attainment of piperacillin/tazobactam (PTZ) in adult critically ill patients. METHODS We conducted a prospective observational study in the intensive care unit (ICU) of the Hôpital du Sacré-Cœur de Montréal (a Level I trauma centre in Montreal, QC, Canada) between January 2021 and June 2022. We included patients aged 18 yr or older admitted to the ICU who received PTZ by intravenous administration. Demographic and clinical characteristics were collected, and clinical scores were calculated. On study day 1 of antimicrobial therapy, three blood samples were collected at the following timepoints: one hour after PTZ dose administration and at the middle and at the end of the dosing interval. The sampling schedule was repeated on days 4 and 7 of therapy if possible. Samples were analyzed by ultra-high performance liquid chromatography with diode array detector to determine the total piperacillin concentration. Middle- and end-of-interval concentrations were used for target attainment analyses, and were defined as a concentration above the minimal inhibitory concentration of 16 mg·L-1, corresponding to the breakpoint of Enterobacteriaceae and Pseudomonas aeruginosa. RESULTS Forty-three patients were recruited and 202 blood samples were analyzed. The most prevalent dose was 3/0.375 g every six hours (n = 50/73 doses administered, 68%) with a 30-min infusion. We observed marked variability over the three sampling timepoints, and the median [interquartile range] piperacillin concentrations at peak, middle of interval, and end of interval were 109.4 [74.0-152.3], 59.3 [21.1-74.4], and 25.3 [6.8-44.6] mg·L-1, respectively. When assessing target attainment, 37% of patients did not reach the efficacy target of a trough concentration of 16 mg·L-1. The majority of patients who were underexposed were patients with normal to augmented renal clearance. CONCLUSION In this prospective observational study of adult ICU patients receiving intravenous PTZ, a large proportion had subtherapeutic concentrations of piperacillin. This was most notable in patients with normal to augmented renal clearance. More aggressive dosage regimens may be required for this subpopulation to ensure attainment of efficacy targets.
Collapse
Affiliation(s)
- Ibrahim El-Haffaf
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada.
- Faculty of Pharmacy, Université de Montréal, 2940 chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada.
| | - Amélie Marsot
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Centre de recherche, CHU Sainte-Justine, Montréal, QC, Canada
| | - Djamila Hachemi
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
| | - Thomas Pesout
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
| | - Virginie Williams
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
| | - Marc-André Smith
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
| | - Martin Albert
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - David Williamson
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- CIUSSS-NIM-Hôpital du Sacré-Cœur de Montréal and CIUSSS-NIM Research Center, Montreal, QC, Canada
| |
Collapse
|
47
|
Wenker SAM, Alabdulkarim N, Readman JB, Slob EMA, Satta G, Ali S, Gadher N, Shulman R, Standing JF. Defining the pharmacokinetic/pharmacodynamic index of piperacillin/tazobactam within a hollow-fibre infection model to determine target attainment in intensive care patients. JAC Antimicrob Resist 2024; 6:dlae036. [PMID: 38476774 PMCID: PMC10928666 DOI: 10.1093/jacamr/dlae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Background It is important to optimize dosing schemes of antibiotics to maximize the probability of therapeutic success. The recommended pharmacokinetic/pharmacodynamic (PK/PD) index for piperacillin/tazobactam therapy in clinical studies ranges widely (50%-100% fT>1-4×MIC). Dosing schemes failing to achieve PK/PD targets may lead to negative treatment outcomes. Objectives The first aim of this study was to define the optimal PK/PD index of piperacillin/tazobactam with a hollow-fibre infection model (HFIM). The second aim was to predict whether these PK/PD targets are currently achieved in critically ill patients through PK/PD model simulation. Patients and methods A dose-fractionation study comprising 21 HFIM experiments was performed against a range of Gram-negative bacterial pathogens, doses and infusion times. Clinical data and dose histories from a case series of nine patients with a known bacterial infection treated with piperacillin/tazobactam in the ICU were collected. The PK/PD index and predicted plasma concentrations and therefore target attainment of the patients were simulated using R version 4.2.1. Results fT >MIC was found to be the best-fitting PK/PD index for piperacillin/tazobactam. Bactericidal activity with 2 log10 cfu reduction was associated with 77% fT>MIC. Piperacillin/tazobactam therapy was defined as clinically 'ineffective' in ∼78% (7/9) patients. Around seventy-one percent (5/7) of these patients had a probability of >10% that 2 log10 cfu reduction was not attained. Conclusions Our dose-fractionation study indicates an optimal PK/PD target in piperacillin/tazobactam therapies should be 77% fT>MIC for 2 log10 kill. Doses to achieve this target should be considered when treating patients in ICU.
Collapse
Affiliation(s)
- Suzanne A M Wenker
- Infection, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Najla Alabdulkarim
- Infection, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Clinical Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - John B Readman
- Infection, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Elise M A Slob
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, Haaglanden Medical Center, The Hague, The Netherlands
| | - Giovanni Satta
- Department of Infection, University College London Hospitals NHS Foundation Trust, London, UK
| | - Shanom Ali
- Environmental Research Laboratory, University College London Hospitals NHS Foundation Trust, London, UK
| | - Nishma Gadher
- Pharmacy Department, CMORE, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rob Shulman
- Pharmacy Department, CMORE, University College London Hospitals NHS Foundation Trust, London, UK
| | - Joseph F Standing
- Infection, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Pharmacy, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
48
|
Chai MG, Tu Q, Cotta MO, Bauer MJ, Balch R, Okafor C, Comans T, Kruger P, Meyer J, Shekar K, Brady K, Fourie C, Sharp N, Vlad L, Whiley D, Ungerer JPJ, Mcwhinney BC, Farkas A, Paterson DL, Clark JE, Hajkowicz K, Raman S, Bialasiewicz S, Lipman J, Forde BM, Harris PNA, Schlapbach LJ, Coin L, Roberts JA, Irwin AD. Achievement of therapeutic antibiotic exposures using Bayesian dosing software in critically unwell children and adults with sepsis. Intensive Care Med 2024; 50:539-547. [PMID: 38478027 PMCID: PMC11018654 DOI: 10.1007/s00134-024-07353-3] [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: 10/29/2023] [Accepted: 02/11/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE Early recognition and effective treatment of sepsis improves outcomes in critically ill patients. However, antibiotic exposures are frequently suboptimal in the intensive care unit (ICU) setting. We describe the feasibility of the Bayesian dosing software Individually Designed Optimum Dosing Strategies (ID-ODS™), to reduce time to effective antibiotic exposure in children and adults with sepsis in ICU. METHODS A multi-centre prospective, non-randomised interventional trial in three adult ICUs and one paediatric ICU. In a pre-intervention Phase 1, we measured the time to target antibiotic exposure in participants. In Phase 2, antibiotic dosing recommendations were made using ID-ODS™, and time to target antibiotic concentrations were compared to patients in Phase 1 (a pre-post-design). RESULTS 175 antibiotic courses (Phase 1 = 123, Phase 2 = 52) were analysed from 156 participants. Across all patients, there was no difference in the time to achieve target exposures (8.7 h vs 14.3 h in Phase 1 and Phase 2, respectively, p = 0.45). Sixty-one courses in 54 participants failed to achieve target exposures within 24 h of antibiotic commencement (n = 36 in Phase 1, n = 18 in Phase 2). In these participants, ID-ODS™ was associated with a reduction in time to target antibiotic exposure (96 vs 36.4 h in Phase 1 and Phase 2, respectively, p < 0.01). These patients were less likely to exhibit subtherapeutic antibiotic exposures at 96 h (hazard ratio (HR) 0.02, 95% confidence interval (CI) 0.01-0.05, p < 0.01). There was no difference observed in in-hospital mortality. CONCLUSIONS Dosing software may reduce the time to achieve target antibiotic exposures. It should be evaluated further in trials to establish its impact on clinical outcomes.
Collapse
Affiliation(s)
- Ming G Chai
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Quyen Tu
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, South Brisbane, QLD, Australia
| | - Menino O Cotta
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Herston Infectious Disease Institute, Metro North, QLD Health, Herston, QLD, Australia
| | - Michelle J Bauer
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Ross Balch
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Charles Okafor
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
| | - Tracy Comans
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
| | - Peter Kruger
- Intensive Care Unit, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Jason Meyer
- Intensive Care Unit, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Kiran Shekar
- Adult Intensive Care Services and Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Kara Brady
- Adult Intensive Care Services and Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Cheryl Fourie
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Natalie Sharp
- Paediatric Intensive Care Unit, Queensland Children's Hospital, South Brisbane, QLD, Australia
| | - Luminita Vlad
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - David Whiley
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Jacobus P J Ungerer
- Department of Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Biomedical Science, University of Queensland, Brisbane, QLD, Australia
| | - Brett C Mcwhinney
- Department of Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
| | - Andras Farkas
- Optimum Dosing Strategies, Bloomingdale, NJ, 07403, USA
- Department of Pharmacy, Saint Clare's Health, Denville, NJ, 07834, USA
| | - David L Paterson
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- ADVANCE-ID, Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Julia E Clark
- Infection Management and Prevention Service, Queensland Children's Hospital, Brisbane, Australia
| | - Krispin Hajkowicz
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sainath Raman
- Paediatric Intensive Care Unit, Queensland Children's Hospital, South Brisbane, QLD, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Seweryn Bialasiewicz
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jeffrey Lipman
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- ICU and Jameson Trauma Institute, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Nimes University Hospital, University of Montpellier, Nimes, France
| | - Brian M Forde
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Patrick N A Harris
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Herston Infectious Disease Institute, Metro North, QLD Health, Herston, QLD, Australia
- Central Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Luregn J Schlapbach
- Paediatric Intensive Care Unit, Queensland Children's Hospital, South Brisbane, QLD, Australia
- Department of Pediatric and Neonatal Intensive Care, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lachlan Coin
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jason A Roberts
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Herston Infectious Disease Institute, Metro North, QLD Health, Herston, QLD, Australia
| | - Adam D Irwin
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia.
- Infection Management and Prevention Service, Queensland Children's Hospital, Brisbane, Australia.
| |
Collapse
|
49
|
Zhang X, Wang Y, Li S, Xie F, Yi H. Simulated drug disposition in critically ill patients to evaluate effective PK/PD targets for combating Pseudomonas aeruginosa resistance to meropenem. Antimicrob Agents Chemother 2024; 68:e0154123. [PMID: 38319075 PMCID: PMC10916391 DOI: 10.1128/aac.01541-23] [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: 11/22/2023] [Accepted: 01/13/2024] [Indexed: 02/07/2024] Open
Abstract
Bacterial infections, including those caused by Pseudomonas aeruginosa, often lead to sepsis, necessitating effective antibiotic treatment like carbapenems. The key pharmacokinetic/pharmacodynamic (PK/PD) index correlated to carbapenem efficacy is the fraction time of unbound plasma concentration above the minimum inhibitory concentration (MIC) of the pathogen (%fT > MIC). While multiple targets exist, determining the most effective one for critically ill patients remains a matter of debate. This study evaluated meropenem's bactericidal potency and its ability to combat drug resistance in Pseudomonas aeruginosa under three representative PK/PD targets: 40% fT > MIC, 100% fT > MIC, and 100% fT > 4× MIC. The hollow fiber infection model (HFIM) was constructed, validated, and subsequently inoculated with a substantial Pseudomonas aeruginosa load (1 × 108 CFU/mL). Different meropenem regimens were administered to achieve the specified PK/PD targets. At specified intervals, samples were collected from the HFIM system and subjected to centrifugation. The resulting supernatant was utilized to determine drug concentrations, while the precipitates were used to track changes in both total and drug-resistant bacterial populations over time by the spread plate method. The HFIM accurately reproduced meropenem's pharmacokinetics in critically ill patients. All three PK/PD target groups exhibited a rapid bactericidal response within 6 h of the initial treatment. However, the 40% fT > MIC and 100% fT > MIC groups subsequently showed bacterial resurgence and resistance, whereas the 100% fT > 4× MIC group displayed sustained bactericidal activity with no evidence of drug resistance. The HFIM system revealed that maintaining 100% fT > 4× MIC offers a desirable microbiological response for critically ill patients, demonstrating strong bactericidal capacity and effective prevention of drug resistance.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yan Wang
- Yueyang Inspection and Testing Center, Yueyang, China
| | - Sanwang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Hanxi Yi
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
50
|
Chai MG, Roberts NA, Dobbins C, Roberts JA, Cotta MO. Factors Influencing Integration and Usability of Model-Informed Precision Dosing Software in the Intensive Care Unit. Appl Clin Inform 2024; 15:388-396. [PMID: 38754464 PMCID: PMC11098592 DOI: 10.1055/s-0044-1786978] [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: 10/28/2023] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Antimicrobial dosing in critically ill patients is challenging and model-informed precision dosing (MIPD) software may be used to optimize dosing in these patients. However, few intensive care units (ICU) currently adopt MIPD software use. OBJECTIVES To determine the usability of MIPD software perceived by ICU clinicians and identify implementation barriers and enablers of software in the ICU. METHODS Clinicians (pharmacists and medical staff) who participated in a wider multicenter study using MIPD software were invited to participate in this mixed-method study. Participants scored the industry validated Post-study System Usability Questionnaire (PSSUQ, assessing software usability) and Technology Acceptance Model 2 (TAM2, assessing factors impacting software acceptance) survey. Semistructured interviews were used to explore survey responses. The framework approach was used to identify factors influencing software usability and integration into the ICU from the survey and interview data. RESULTS Seven of the eight eligible clinicians agreed to participate in the study. The PSSUQ usability scores ranked poorer than the reference norms (2.95 vs. 2.62). The TAM2 survey favorably ranked acceptance in all domains, except image. Qualitatively, key enablers to workflow integration included clear and accessible data entry, visual representation of recommendations, involvement of specialist clinicians, and local governance of software use. Barriers included rigid data entry systems and nonconformity of recommendations to local practices. CONCLUSION Participants scored the MIPD software below the threshold that implies good usability. Factors such as availability of software support by specialist clinicians was important to participants while rigid data entry was found to be a deterrent.
Collapse
Affiliation(s)
- Ming G. Chai
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Pharmacy Department, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Natasha A. Roberts
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
| | - Chelsea Dobbins
- School of Electrical Engineering and Computer Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Jason A. Roberts
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Pharmacy Department, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nimes University Hospital, University of Montpellier, Nimes, France
- Herston Infectious Diseases Institute, Metro North Health, Brisbane, Australia
| | - Menino O. Cotta
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|