1
|
Wolthuis DFGJ, Freriksen JJM, Ter Avest M, Kartha RV, de Wildt SN, Wijnsma K, van de Kar NCAJ, Ter Heine R. Model-informed repurposing of eliglustat for treatment and prophylaxis of Shiga toxin-producing Escherichia coli hemolytic-uremic syndrome (STEC-HUS) in children. Pediatr Nephrol 2025; 40:2009-2019. [PMID: 39900743 PMCID: PMC12031897 DOI: 10.1007/s00467-025-06688-3] [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: 12/03/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Shiga toxin-producing Escherichia coli hemolytic-uremic syndrome (STEC-HUS) is a severe illness predominantly affecting young children, with limited treatment options beyond supportive care. Eliglustat, approved for Gaucher disease, shows potential in reducing Shiga toxin binding to target glomerular endothelial cells in vitro, prompting interest as a treatment for STEC-HUS. However, it remains unknown what dose is likely to be effective and safe for treatment of STEC-HUS in the pediatric population. We hypothesize that effective and safe levels of eliglustat can be reached in children. METHODS We identified pharmacokinetic targets of efficacy for treatment and prophylaxis of STEC-HUS based on a preclinical model and human cardiac safety data. Then, we developed oral and intravenous dosing regimens using population pharmacokinetic (popPK) simulations based on an existing model enriched to allow extrapolation to a simulated virtual pediatric population. These dosing regimens were then confirmed using a verified physiologically based pharmacokinetic (PBPK) model. RESULTS We simulated, using popPK data, oral and intravenous dosing regimens resulting in adequate target exposure in > 90% of all patients, with minimal expected risk for cardiotoxicity. Confirmation of these dosing regimens with PBPK modeling resulted in very similar exposure, with lower interindividual variability and minimal toxicity potential. CONCLUSIONS Based on pharmacokinetic modeling, we developed oral and intravenous eliglustat dosing regimens that are likely safe and effective for treatment of STEC-HUS and prophylaxis in case of outbreaks of STEC infections. Clinical evaluation of these dosing regimens in children suspected of or diagnosed with STEC-HUS is required and should include assessment of pharmacokinetics, efficacy, and safety (e.g., ECG monitoring).
Collapse
Affiliation(s)
- David F G J Wolthuis
- Department of Internal Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands.
| | | | - Mendy Ter Avest
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, USA
| | - Saskia N de Wildt
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
- Department of Intensive Care, Radboudumc, Nijmegen, The Netherlands
- Department of Neonatal and Pediatric Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Kioa Wijnsma
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Nicole C A J van de Kar
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Gao Y, Wu T, Pu L, Wang M, Wang C, Guo Y, Qiu W. Risk Factors for Nephrotoxicity Among Neonates Receiving Vancomycin: A Systematic Review and Meta-analysis. Paediatr Drugs 2025:10.1007/s40272-025-00690-w. [PMID: 40208565 DOI: 10.1007/s40272-025-00690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Nephrotoxicity may increase the risk of neonatal mortality. Early identification of risk factors for nephrotoxicity is essential to improve clinical outcomes. This study aimed to determine the risk factors for nephrotoxicity in neonates receiving vancomycin to promote the safe use of vancomycin. METHODS We searched international and Chinese databases from 1990 to 24 March 2024 for studies involving neonates receiving vancomycin and reporting their nephrotoxic outcomes. Effects were estimated with 95% confidence intervals (CIs), odds ratios (ORs), and standardized mean differences. We evaluated the certainty of the evidence using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) tool. RESULTS In total, the study included 12 retrospective cohort studies involving 2079 neonates. In neonates receiving vancomycin, the incidence of nephrotoxicity varied from 2.7 to 20.0%. Moderate-quality evidence indicated that furosemide (OR 5.80; 95% CI 2.98-11.28), amphotericin B (OR 2.75; 95% CI 1.23-6.12), patent ductus arteriosus (OR 5.93; 95% CI 2.80-12.55), and necrotizing enterocolitis (OR 4.49; 95% CI 2.12-9.49) were risk factors for nephrotoxicity in neonates receiving vancomycin. Low-quality evidence suggested that vasoactive agents (OR 9.23; 95% CI 1.06-80.62) were risk factors. Subgroup analysis with moderate-quality evidence identified a steady-state vancomycin trough concentration > 20 mg·L-1 (OR 6.87; 95% CI 3.81-12.39) as a risk factor. Very low-quality evidence indicated that aminoglycosides (OR 0.29; 95% CI 0.13-0.62) were not risk factors. CONCLUSIONS This study reveals the nephrotoxic risk factors for neonates receiving vancomycin, which could help in the implementation of measures to prevent further renal impairment. PROSPERO REGISTRATION NUMBER CRD42024564584.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Tong Wu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Libin Pu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Mengjie Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Chang Wang
- Pharmacy department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yinyin Guo
- Pharmacy department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Wen Qiu
- Pharmacy department, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- National Drug Clinical Trial Institution, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
3
|
Adamiszak A, Pietrzkiewicz K, Bartkowska-Śniatkowska A, Smuszkiewicz P, Kusza K, Grześkowiak E, Bienert A. Do Critically Ill Patients Undergoing Continuous Renal Replacement Therapy Require Ceftaroline Dosage Adjustments? Ceftaroline PopPK Model and Dosage Simulations with the Probability of Target Attainment Analysis Based on Retrospective Data. Antibiotics (Basel) 2025; 14:347. [PMID: 40298514 PMCID: PMC12024021 DOI: 10.3390/antibiotics14040347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives: We aimed to develop a population pharmacokinetic (PopPK) model and evaluate dosing regimens for different renal clearances and continuous renal replacement therapy (CRRT) settings. Methods: Data were collected from four studies in intensive care unit (ICU) adult patients receiving 400-600 mg of ceftaroline every 8-12 h in a one-hour infusion. The PopPK model was developed according to non-linear mixed effects modeling implemented in Monolix 2024R1. To investigate dosing recommendations, Monte Carlo simulations and probability of target attainment (PTA) analysis were performed in Simulx 2024R1. Results: We collected 296 plasma concentrations from 29 non-CRRT patients and 24 pre-filter (systemic), 23 post-filter, and 23 effluent concentrations from four CRRT patients using WebPlotDigitizer (Version 4.7). A five-compartment model, with the first-order elimination from the central compartment and additional elimination with the effluent during CRRT, best described the ceftaroline concentrations. Creatinine clearance (ClCr) was identified as a covariate on the clearance of elimination (Cl) and CRRT modality on the central and peripheral compartments' volumes and intercompartmental clearance. The results of dosage simulations for different CRRT modalities and ClCr, S. pneumoniae (MIC = 0.25 mg/L) and methicillin-resistant S. aureus (MRSA) (MIC = 1 mg/L) infections, and assumed 100%ƒT>MIC target, revealed that registered ceftaroline dosages are sufficient to achieve assumed PTA, except MRSA infection in patients with augmented renal clearance (ARC). Conclusions: Our successfully developed model allows flexible PK simulations of ceftaroline, including real-time changes in settings and even temporary or permanent cessation of CRRT. However, the results of our study warrant clinical validation and should be used with caution primarily due to the limited CRRT patient number included in the analysis.
Collapse
Affiliation(s)
- Arkadiusz Adamiszak
- Department of Pharmacology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Krzysztof Pietrzkiewicz
- Department of Paediatric Anaesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (K.P.); (A.B.-Ś.)
| | - Alicja Bartkowska-Śniatkowska
- Department of Paediatric Anaesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (K.P.); (A.B.-Ś.)
| | - Piotr Smuszkiewicz
- Department of Anesthesiology, Intensive Therapy and Pain Management, Poznan University of Medical Sciences, 60-352 Poznan, Poland; (P.S.); (K.K.)
| | - Krzysztof Kusza
- Department of Anesthesiology, Intensive Therapy and Pain Management, Poznan University of Medical Sciences, 60-352 Poznan, Poland; (P.S.); (K.K.)
| | - Edmund Grześkowiak
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Agnieszka Bienert
- Department of Pharmacology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| |
Collapse
|
4
|
Downes KJ, Sharova A, Amajor V, Gianchetti L, Himebauch AS, Fitzgerald JC, Zuppa AF. Urinary Biomarkers and Attainment of Cefepime Therapeutic Targets in Critically Ill Children. Pediatr Infect Dis J 2025:00006454-990000000-01249. [PMID: 40073372 DOI: 10.1097/inf.0000000000004784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
BACKGROUND Critically ill children are at risk for subtherapeutic antibiotic concentrations. The frequency of target attainment and risk factors for subtherapeutic concentrations of cefepime in children have not been extensively studied. METHODS We performed an observational study in critically ill children receiving a new prescription of standard dosing of cefepime for suspected sepsis (≥2 systemic inflammatory response syndrome criteria within 48 hours of cefepime start). Three plasma cefepime concentrations were measured at steady state and, a urine sample was collected prior to pharmacokinetics (PK) sampling for measurement of urinary biomarkers. Bayesian analysis determined cefepime PK for each individual, and simulations were used to estimate time above minimum inhibitory concentration (fT > MIC) for 8 µg/mL (breakpoint for Pseudomonas). Clinical factors and urinary biomarkers were compared between patients who did and did not achieve 100% fT > MIC. Correlations between covariates and cefepime PK parameters, as well as optimal cut points to identify <100% fT > MIC, were evaluated. RESULTS Twenty-one subjects were enrolled and PK sampling occurred after a median of 5 doses (range, 3-9); 43% of children achieved 100% fT > MIC for an MIC of 8 µg/mL. Younger age and lower urinary biomarkers (neutrophil gelatinase-associated lipocalin and kidney injury molecule-1) were significantly associated with failure to attain 100% fT > 8 µg/mL. Urinary neutrophil gelatinase-associated lipocalin (<122.1-ng/mg creatinine) best identified individuals who failed to attain this putative target (positive predictive value, 91.7%). CONCLUSIONS A large proportion of critically ill children failed to attain target concentrations for empiric treatment of Pseudomonas aeruginosa with cefepime. Urinary biomarkers may be a noninvasive means to identify those at higher risk for increased cefepime clearance and subtherapeutic concentrations.
Collapse
Affiliation(s)
- Kevin J Downes
- From the Department of Pediatrics
- Division of Infectious Diseases
- Center for Clinical Pharmacology
- Clinical Futures
| | - Anna Sharova
- Center for Clinical Pharmacology
- Clinical Futures
| | | | | | - Adam S Himebauch
- Department of Anesthesiology and Critical Care, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA
- Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA
- Clinical Futures
- Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Athena F Zuppa
- From the Department of Pediatrics
- Center for Clinical Pharmacology
| |
Collapse
|
5
|
Butragueño-Laiseca L, García-Orueta G, Riva N, Trocóniz IF, Fernández SN, Camacho Vicente V, Padilla B, Slöcker M, Santiago MJ. Population pharmacokinetic analysis of teicoplanin in paediatric patients, including those receiving continuous kidney replacement therapy: a prospective cohort study. J Antimicrob Chemother 2025; 80:868-875. [PMID: 39846780 PMCID: PMC11879232 DOI: 10.1093/jac/dkaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVES Teicoplanin is a commonly used antibiotic in critically ill children. However, teicoplanin dosing is often inaccurate, especially in children undergoing continuous kidney replacement therapy (CKRT). This study aims to develop a population pharmacokinetic (PK) model to optimize teicoplanin dosing in critically ill children, including those on CKRT. METHODS Data from 26 critically ill children (12 with CKRT) receiving the standard dosing regimen were analysed. In total, 172 teicoplanin concentration measurements from plasma, pre- and post-filter ports were modelled simultaneously using NONMEM 7.4. Simulations were conducted to assess the target attainment (Cmin = 10 mg/L and AUC24/MIC > 800 h) of the current standard dosing regimen and of different alternative dosing regimens. RESULTS A two-compartment model was selected. Weight significantly affected renal clearance and volume of distribution of the central compartment, while filter surface area affected haemofilter clearance. Only 16 patients (59%) achieved a Cmin of >10 mg/L with the standard dosing regimen, and only 1 achieved the target AUC/MIC. Based on simulation results, 3 × 15 mg/kg q12h + 10 mg/kg q24h (CKRT) and 3 × 15 mg/kg q12h + 15 mg/kg q24h (no CKRT) could be better alternative regimens. CONCLUSIONS This population model is a good proof of concept to develop modelling approaches that could help in an individualized dosing approach that needs to be adopted in critically ill paediatric patients. The standard paediatric dosage for teicoplanin could be insufficient for optimal exposure, and higher doses may benefit both CKRT and non-CKRT patients.
Collapse
Affiliation(s)
- Laura Butragueño-Laiseca
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Departamento de Salud Departamento de Salud Pública y Materno Infantil, Universidad Complutense de Madrid, Madrid, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS RD21/0012/0011), Institute of Health Carlos III, Madrid, Spain
| | - Gastón García-Orueta
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Natalia Riva
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Iñaki F Trocóniz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Institute of Data Science and Artificial Intelligence, DATAI, University of Navarra, Pamplona, Spain
| | - Sarah N Fernández
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Departamento de Salud Departamento de Salud Pública y Materno Infantil, Universidad Complutense de Madrid, Madrid, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS RD21/0012/0011), Institute of Health Carlos III, Madrid, Spain
| | - Verónica Camacho Vicente
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Departamento de Salud Departamento de Salud Pública y Materno Infantil, Universidad Complutense de Madrid, Madrid, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS RD21/0012/0011), Institute of Health Carlos III, Madrid, Spain
| | - Belén Padilla
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Clinical Microbiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Slöcker
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Departamento de Salud Departamento de Salud Pública y Materno Infantil, Universidad Complutense de Madrid, Madrid, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS RD21/0012/0011), Institute of Health Carlos III, Madrid, Spain
| | - María José Santiago
- Pediatric Intensive Care Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Departamento de Salud Departamento de Salud Pública y Materno Infantil, Universidad Complutense de Madrid, Madrid, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS RD21/0012/0011), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Pavia K, Girdwood ST, Paice K, Dong M, Mizuno T, Tang P, Mangeot C, Vinks AA, Kaplan J. Acute kidney injury is associated with abnormal cefepime exposure among critically ill children and young adults. Pediatr Nephrol 2025; 40:513-521. [PMID: 39150525 PMCID: PMC11666613 DOI: 10.1007/s00467-024-06477-4] [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: 03/22/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Elevated cefepime blood concentrations can cause neurotoxicity in adults. The consequences of elevated cefepime concentrations among pediatric patients are unknown. Future exploration of such effects requires first identifying patients at risk for elevated cefepime exposure. We investigated the role of acute kidney injury as a risk factor for increased cefepime concentrations in critically ill children. METHODS This was a retrospective analysis at a single pediatric intensive care unit. Analyzed patients received at least 24 h of cefepime and had at least two opportunistic samples collected for total cefepime concentration measurement. Individual pharmacokinetic (PK) profiles during treatment courses were reconstructed using Bayesian estimation with an established population PK model. Elevated trough concentration (Cmin) was defined as ≥ 30 mg/L based on adult toxicity studies. The effect of kidney dysfunction on cefepime PK profiles was interrogated using a mixed-effect model. RESULTS Eighty-seven patients were included, of which 13 (14.9%) had at least one estimated Cmin ≥ 30 mg/L. Patients with elevated Cmin were more likely to have acute kidney injury (AKI) during their critical illness (92% vs. 57%, p = 0.015 for any AKI; 62% vs. 26%, p = 0.019 for severe AKI). Patients who had AKI during critical illness had significantly higher cefepime exposure, as quantified by the area under the concentration-time curve over 24 h (AUC24h) and Cmin. CONCLUSIONS Among critically ill children, AKI is associated with elevated cefepime concentrations. Identifying these high-risk patients is the first step toward evaluating the clinical consequences of such exposures.
Collapse
Affiliation(s)
- Kathryn Pavia
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| | - Sonya Tang Girdwood
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kelli Paice
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Min Dong
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peter Tang
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Colleen Mangeot
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jennifer Kaplan
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
7
|
Paice KM, Girdwood ST, 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; 25:1103-1116. [PMID: 39162600 PMCID: PMC11617271 DOI: 10.1097/pcc.0000000000003599] [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] [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 M. Paice
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- 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, USA
- 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, USA
| | - 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, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
8
|
Li M, Gao L, Wang Z, Zeng L, Chen C, Wang J, Li S, Liu M, Wang Y. Population pharmacokinetics and dose optimization of ceftazidime in critically ill children. Front Pharmacol 2024; 15:1470350. [PMID: 39664522 PMCID: PMC11631598 DOI: 10.3389/fphar.2024.1470350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Objective The aim of this study was to develop a population pharmacokinetic model for ceftazidime in critically ill children in the pediatric intensive care unit (PICU) and optimize an appropriate dosing regimen for this population. Methods We performed a prospective pharmacokinetic study on critically ill children aged 0.03-15 years. A population pharmacokinetic model was developed using the NLME program. Statistical and graphical methods were used to assess the stability and predictive performance of the model. Monte Carlo simulations were conducted to determine the optimal ceftazidime dosing regimen to achieve 70% fT > minimum inhibitory concentration (MIC). Results This study included 88 critically ill children and 100 ceftazidime serum concentrations. The pharmacokinetic characteristics of ceftazidime were best described by a one-compartment linear elimination model. The weight and estimated glomerular filtration rate (eGFR) were determinant covariates for the clearance (CL) of ceftazidime. The recommended ceftazidime dosage regimens achieved a probability of target attainment (PTA) >90% for critically ill children at MIC values of 2, 4, and 8 mg/L. For bacterial infection at an MIC of 16 mg/L, it is difficult to achieve effective pharmacodynamic (PD) targets in vivo with the commonly used dose of ceftazidime. Conclusion The population pharmacokinetic model of ceftazidime was established in critically ill children. Based on this model, we recommend evidence-based, individualized dosing regimens for subgroups with different weights and renal functions. The current daily dosage for children adequately meets the treatment requirements for MICs of 2, 4, and 8 mg/L, while for bacterial infection at an MIC of 16 mg/L, an elevated dosage regimen may be required. Clinical Trial Registration https://www.medicalresearch.org.cn/login, Identifier MR-42-22-000220.
Collapse
Affiliation(s)
- Mengting Li
- Department of Clinical Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuliu Gao
- Department of Clinical Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuo Wang
- Department of Neonatology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingkong Zeng
- Department of Neonatology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Wang
- Department of Clinical Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sichan Li
- Department of Clinical Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maochang Liu
- Department of Clinical Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Wang
- Drug Clinical Trial Institution, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Boast A, Zhang W, Soeorg H, Gonis G, Di Carlo A, Daley A, Curtis N, McWhinney B, Ungerer JPJ, Lei A, Standing JF, Gwee A. Population pharmacokinetic modeling of ceftriaxone in cerebrospinal fluid in children: should we be using once- or twice-daily dosing for meningitis? Antimicrob Agents Chemother 2024; 68:e0074724. [PMID: 39377581 PMCID: PMC11539224 DOI: 10.1128/aac.00747-24] [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: 05/25/2024] [Accepted: 08/02/2024] [Indexed: 10/09/2024] Open
Abstract
Guidelines for bacterial meningitis in children recommend intravenous ceftriaxone 50 mg/kg (max 2 g) twice daily (BD) or 100 mg/kg (max 4 g) once daily (OD), leaving the decision regarding the dose frequency to the prescriber. We investigated the cerebrospinal fluid (CSF) penetration of ceftriaxone to evaluate whether one dosing regimen is superior. Unbound ceftriaxone concentrations were measured in serum and CSF samples from children aged 0-18 years treated with ceftriaxone if there was a sample remaining after clinical tests were performed. A serum-CSF population pharmacokinetic model was developed using non-linear mixed-effects modeling. The once- and twice-daily dosing regimens were simulated, and the probability of target attainment (PTA) was determined for maintaining a CSF concentration above a minimum inhibitory concentration (MIC) of 1 mg/L for common meningitis pathogens and 4 mg/L for Staphylococcus aureus for 100% of the dosing interval. Sixteen serum and 87 CSF samples were collected from 98 children (age range 0.1-18.5 years). The final two-compartment serum-CSF model included a renal maturation function with weight scaling on clearance and volume of distribution. The estimated serum:CSF uptake was 20.1%. For MIC 1 mg/L, the 24 h PTA was higher for OD (88%) compared with BD (53%) dosing, although both achieved a 100% PTA at steady state. For S. aureus (MIC 4 mg/L), neither dosing regimen was sufficient. Our findings support the use of a 100 mg/kg once daily regimen for empirical treatment of bacterial meningitis due to earlier achievement of the pharmacodynamic target. Neither dosing regimen was adequate for S. aureus meningitis.
Collapse
Affiliation(s)
- A. Boast
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
- Antimicrobials and Infectious Diseases Group, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - W. Zhang
- Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - H. Soeorg
- Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - G. Gonis
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
| | - A. Di Carlo
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
| | - A. Daley
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - N. Curtis
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
- Antimicrobials and Infectious Diseases Group, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - B. McWhinney
- Department of Chemical Pathology, Pathology Queensland, Brisbane, Australia
| | - J. P. J. Ungerer
- Department of Chemical Pathology, Pathology Queensland, Brisbane, Australia
- Faculty of Health and Behavioural Sciences, University of Queensland, Brisbane, Australia
| | - A. Lei
- Antimicrobials and Infectious Diseases Group, Murdoch Children’s Research Institute, Melbourne, Australia
| | - J. F. Standing
- Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Pharmacy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - A. Gwee
- Department of General Medicine and Laboratory Services, The Royal Children’s Hospital Melbourne, Parkville, Australia
- Antimicrobials and Infectious Diseases Group, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Al-Sulaiti FK, Alkhiyami D, Elmekaty EZI, Awaisu A, Kheir N, El-Zubair A, Al-Sulaiti HK. Appropriateness of gentamicin therapeutic drug monitoring at a Middle Eastern tertiary hospital setting: a retrospective evaluation and quality audit. J Pharm Policy Pract 2024; 17:2375753. [PMID: 39011355 PMCID: PMC11249166 DOI: 10.1080/20523211.2024.2375753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction The use of gentamicin in the treatment of infectious diseases requires frequent monitoring to attain the best treatment outcomes. Objective This study aimed to evaluate the appropriateness of gentamicin therapeutic drug monitoring (TDM) at a tertiary care hospital in Qatar. Methods A one-year quantitative retrospective chart review of all gentamicin TDM records was conducted. Evidence-based criteria were applied to evaluate the appropriateness of gentamicin TDM in terms of indication, sampling times, and post-analytical actions. Results Out of 59 captured gentamicin TDM records, 58 gentamicin samples were eligible for evaluation. Overall, gentamicin TDM appropriateness was achieved in 50% (n = 29) of the evaluated records. However, 12% (n = 7) of gentamicin drug concentrations were below the assay quantification limits or were not sampled appropriately. Inappropriate post-analytical actions (22.4%, n = 13) and inappropriate sampling times (44.8%, n = 26) were recorded. Most of the gentamicin blood samples (n = 43; 74.2%) were taken appropriately at steady-state. Inappropriate sampling time relative to the last dose was captured in 31% (n = 18) of the cases. Although 27.6% (n = 16) of gentamicin concentrations were non-therapeutic, continuing gentamicin dosing without adjustment was the most frequent post-analytical action (69.8%, n = 37). Gentamicin dose regimen continuations, dose regimen decreases and dose regimen discontinuations were inappropriately applied in 27% (n = 10), 25% (n = 2) and 14% (n = 1) of the times, respectively. Conclusion Suboptimal gentamicin TDM practices exist in relation to sampling time and post-analytical actions. Studies exploring setting-specific reasons behind inappropriate TDM practices and methods of its optimisation are needed.
Collapse
Affiliation(s)
- Fatima Khalifa Al-Sulaiti
- Clinical Pharmacy and Practice Department, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
- Qatar National Research Fund, Qatar Foundation, Doha, Qatar
| | - Dania Alkhiyami
- Clinical Pharmacy Department, Al-Wakrah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Eman Zeyad I Elmekaty
- Clinical Pharmacy Department, Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Awaisu
- Clinical Pharmacy and Practice Department, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Nadir Kheir
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
- School of Pharmacy, The University of Auckland, Auckland, New Zealand
| | - Ahmed El-Zubair
- Clinical Pharmacy Department, Al-Khor Hospital, Hamad Medical Corporation, Doha, Qatar
| | | |
Collapse
|
11
|
Wang H, Sheng Y, Ou Y, Xu M, Tao M, Lin S, Deng Z, Bai L, Ding W, Kang Q. Streptomyces-based whole-cell biosensors for detecting diverse cell envelope-targeting antibiotics. Biosens Bioelectron 2024; 249:116004. [PMID: 38199083 DOI: 10.1016/j.bios.2024.116004] [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: 09/22/2023] [Revised: 12/25/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
Cell envelope-targeting antibiotics are potent therapeutic agents against various bacterial infections. The emergence of multiple antibiotic-resistant strains underscores the significance of identifying potent antimicrobials specifically targeting the cell envelope. However, current drug screening approaches are tedious and lack sufficient specificity and sensitivity, warranting the development of more efficient methods. Genetic circuit-based whole-cell biosensors hold great promise for targeted drug discovery from natural products. Here, we performed comparative transcriptomic analysis of Streptomyces coelicolor M1146 exposed to diverse cell envelope-targeting antibiotics, aiming to identify regulatory elements involved in perceiving and responding to these compounds. Differential gene expression analysis revealed significant activation of VanS/R two-component system in response to the glycopeptide class of cell envelope-acting antibiotics. Therefore, we engineered a pair of VanS/R-based biosensors that exhibit functional complementarity and possess exceptional sensitivity and specificity for glycopeptides detection. Additionally, through promoter screening and characterization, we expanded the biosensor's detection range to include various cell envelope-acting antibiotics beyond glycopeptides. Our genetically engineered biosensor exhibits superior performance, including a dynamic range of up to 887-fold for detecting subtle antibiotic concentration changes in a rapid 2-h response time, enabling high-throughput screening of natural product libraries for antimicrobial agents targeting the bacterial cell envelope.
Collapse
Affiliation(s)
- Hengyu Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Sheng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yixin Ou
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Min Xu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, West 7th Avenue No. 32, 300308, Tianjin, China
| | - Meifeng Tao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Shuangjun Lin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Linquan Bai
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Ding
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Qianjin Kang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China.
| |
Collapse
|
12
|
Papachatzi E, Gkentzi D, Tzifas S, Dassios T, Dimitriou G. Daptomycin Use for Persistent Coagulase-Negative Staphylococcal Bacteremia in a Neonatal Intensive Care Unit. Antibiotics (Basel) 2024; 13:254. [PMID: 38534689 DOI: 10.3390/antibiotics13030254] [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: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
During the last two decades, the incidence of late-onset sepsis (LOS) has increased due to improved survival of premature neonates. Persistent bacteremia (PB) in LOS is defined as more than two positive blood cultures obtained on different calendar days during the same infectious episode. Although rare, PB should be treated aggressively to prevent adverse outcomes. Daptomycin, a lipopeptide antibiotic, has been used in neonates with persistent coagulase-negative staphylococci (CoNS) bacteremia with promising results, but studies reporting on the efficacy and safety of the agent are scarce. The purpose of this study was to evaluate the efficacy and safety of daptomycin use for persistent CoNS bacteremia in a neonatal cohort. This is a retrospective, observational, single-center study of neonates treated with daptomycin during 2011-2022 in the Tertiary Neonatal Intensive Care Unit (NICU) of the University General Hospital of Patras, Greece. For the years 2011-2022, there were 3.413 admissions to the NICU. During the last 3 years (2020-2022)-the active epidemiological surveillance period-123 infants (out of 851 admissions, 14.4%) developed CoNS bacteremia (LOS). During the study period, twelve infants with PB were treated with daptomycin. They had a median gestational age of 32 weeks (IQR 31-34) and mean (SD) birth weight of 1.840 (867) grams. CoNS bacteremia isolates were s. epidermidis (50%), s. haemolyticus (20%), s. hominis (20%) and s. warneri (10%). The decision to start daptomycin (6 mg/kg/dose twice daily) was taken on median day 10 (ΙQR 7-15) of infection. None of the infants had focal complications or meningitis. Daptomycin therapy caused no renal, hepatic, muscular or gastrointestinal adverse events. One neonate developed seizures, and one death occurred due to multiple complications of prematurity. Most infants (11/12) were successfully treated and eventually had negative blood culture. Daptomycin monotherapy showed an adequate cure rate in premature neonates with persistent CoNS bacteremia in a tertiary NICU. In our study, daptomycin was effective and well tolerated; the safety profile, however, needs to be confirmed in larger studies and randomized controlled trials.
Collapse
Affiliation(s)
- Eleni Papachatzi
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Despoina Gkentzi
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| | - Sotiris Tzifas
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Theodore Dassios
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
| | - Gabriel Dimitriou
- Neonatal Intensive Care Unit, University General Hospital of Patras, 26504 Patras, Greece
- Department of Pediatrics, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
13
|
Wong Vega M, Starr MC, Brophy PD, Devarajan P, Soranno DE, Akcan-Arikan A, Basu R, Goldstein SL, Charlton JR, Barreto E. Advances in pediatric acute kidney injury pharmacology and nutrition: a report from the 26th Acute Disease Quality Initiative (ADQI) consensus conference. Pediatr Nephrol 2024; 39:981-992. [PMID: 37878137 PMCID: PMC10817838 DOI: 10.1007/s00467-023-06178-4] [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: 06/28/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND In the past decade, there have been substantial advances in our understanding of pediatric AKI. Despite this progress, large gaps remain in our understanding of pharmacology and nutritional therapy in pediatric AKI. METHODS During the 26th Acute Disease Quality Initiative (ADQI) Consensus Conference, a multidisciplinary group of experts reviewed the evidence and used a modified Delphi process to achieve consensus on recommendations for gaps and advances in care for pharmacologic and nutritional management of pediatric AKI. The current evidence as well as gaps and opportunities were discussed, and recommendations were summarized. RESULTS Two consensus statements were developed. (1) High-value, kidney-eliminated medications should be selected for a detailed characterization of their pharmacokinetics, pharmacodynamics, and pharmaco-"omics" in sick children across the developmental continuum. This will allow for the optimization of real-time modeling with the goal of improving patient care. Nephrotoxin stewardship will be identified as an organizational priority and supported with necessary resources and infrastructure. (2) Patient-centered outcomes (functional status, quality of life, and optimal growth and development) must drive targeted nutritional interventions to optimize short- and long-term nutrition. Measures of acute and chronic changes of anthropometrics, body composition, physical function, and metabolic control should be incorporated into nutritional assessments. CONCLUSIONS Neonates and children have unique metabolic and growth parameters compared to adult patients. Strategic investments in multidisciplinary translational research efforts are required to fill the knowledge gaps in nutritional requirements and pharmacological best practices for children with or at risk for AKI.
Collapse
Affiliation(s)
- Molly Wong Vega
- Renal and Apheresis Services, Texas Children's Hospital, Houston, TX, USA
| | - Michelle C Starr
- Department of Pediatrics, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Pediatric and Adolescent Comparative Effectiveness Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Patrick D Brophy
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester, Rochester, NY, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Danielle E Soranno
- Department of Pediatrics, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Bioengineering, Purdue University, West Lafayette, IN, USA
| | - Ayse Akcan-Arikan
- Divisions of Critical Care and Nephrology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Rajit Basu
- Division of Critical Care, Department of Pediatrics, Northwestern University, Chicago, IL, USA
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Jennifer R Charlton
- Division of Nephrology, Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA, 22901, USA.
| | - Erin Barreto
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Budai KA, Tímár ÁE, Obeidat M, Máté V, Nagy R, Harnos A, Kiss-Dala S, Hegyi P, Garami M, Hankó B, Lódi C. Extended infusion of β-lactams significantly reduces mortality and enhances microbiological eradication in paediatric patients: a systematic review and meta-analysis. EClinicalMedicine 2023; 65:102293. [PMID: 38021371 PMCID: PMC10651452 DOI: 10.1016/j.eclinm.2023.102293] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Background Paediatric patients are often exposed to subtherapeutic levels or treatment failure of β-lactams, and prolonged infusion may be beneficial. We aimed to investigate the efficacy and safety of extended infusion (EI; defined as ≥3 h) or continuous infusion vs. short, intermittent infusion (SI; defined as ≤60 min) of β-lactams in patients <21 years of age. Methods A systematic review and meta-analysis was conducted to compare EI and continuous infusion with SI of β-lactams in children. A systematic search was performed in MEDLINE (via PubMed), Embase, CENTRAL, and Scopus databases for randomised controlled trials (RCTs) and observational studies published from database inception up to August 22, 2023. Any comparative study concerned with mortality, clinical efficacy, adverse events, or plasma concentrations of β-lactams for any infection was eligible. Case reports, case series, and patients aged >21 years were excluded. Odds ratios (OR) and median differences with 95% confidence intervals (CI) were calculated using a random-effects model. Risk of bias (ROB) was assessed using ROB2 and ROBINS-I tools. The protocol was registered with PROSPERO, CRD42022375397. Findings In total, 19,980 articles were screened, out of which 19 studies (4195 patients) were included in the meta-analysis. EI administration was associated with a significantly lower all-cause mortality in both RCTs and non-RCTs [OR 0.74; CI 0.55-0.99; I2 = 0%; CI 0-58%]. Early microbiological eradication was higher with EI [OR 3.18; CI 2.24-4.51; I2 = 0%; CI 0-90%], but the clinical cure did not differ significantly between the two groups [OR 1.20; CI 0.17-8.71; I2 = 79%; CI 32-93%]. Achieving the optimal plasma level (50-100% fT > MIC) appeared favourable in the EI group compared to the SI. No significant differences were observed in the adverse events. The overall ROB was high because of the small sample sizes and clinically heterogeneous populations. Interpretation Our findings suggest that extended infusion of β-lactams was associated with lower mortality and increased microbiological eradication and was considered safe compared to short-term infusion. Funding None.
Collapse
Affiliation(s)
- Kinga Anna Budai
- University Pharmacy, Department of Pharmacy Administration, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ágnes Eszter Tímár
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Mahmoud Obeidat
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Vanda Máté
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Rita Nagy
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Andrea Harnos
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Biostatistics at the University of Veterinary Medicine, Budapest, Hungary
| | - Szilvia Kiss-Dala
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Miklós Garami
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Balázs Hankó
- University Pharmacy, Department of Pharmacy Administration, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Csaba Lódi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Gatti M, Campoli C, Latrofa ME, Ramirez S, Sasso T, Mancini R, Caramelli F, Viale P, Pea F. Relationship Between Real-time TDM-guided Pharmacodynamic Target Attainment of Continuous Infusion Beta-lactam Monotherapy and Microbiologic Outcome in the Treatment of Critically Ill Children With Severe Documented Gram-negative Infections. Pediatr Infect Dis J 2023; 42:975-982. [PMID: 37523585 PMCID: PMC10569676 DOI: 10.1097/inf.0000000000004054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVES To explore the relationship between real-time therapeutic drug monitoring (TDM)-guided pharmacodynamic target attainment of continuous infusion (CI) beta-lactam monotherapy and microbiological outcome in the treatment of critically ill children with severe documented Gram-negative infections. METHODS Observational, monocentric, retrospective study of critically ill patients receiving CI piperacillin-tazobactam, ceftazidime, or meropenem in monotherapy for documented Gram-negative infections optimized by means of a real-time TDM-guided strategy. Average steady-state beta-lactam concentrations (C ss ) were calculated for each patient, and the beta-lactam C ss /minimum inhibitory concentration (MIC) ratio was selected as a pharmacodynamic parameter of efficacy. The C ss /MIC ratio was defined as optimal if ≥4, quasi-optimal if between 1 and 4, and suboptimal if <1. The relationship between C ss /MIC and microbiological outcome was assessed. RESULTS Forty-six TDM assessments were carried out in 21 patients [median age 2 (interquartile range: 1-8) years]. C ss /MIC ratios were optimal in 76.2% of cases. Patients with optimal C ss /MIC ratios had both a significantly higher microbiological eradication rate (75.0% vs. 0.0%; P = 0.006) and lower resistance development rate (25.0% vs. 80.0%; P = 0.047) than those with quasi-optimal or suboptimal C ss /MIC ratios. Quasi-optimal/suboptimal C ss /MIC ratio occurred more frequently when patients had infections caused by pathogens with MIC values above the European Committee on Antimicrobial Susceptibility Testing clinical breakpoint (100.0% vs. 6.3%; P < 0.001). CONCLUSIONS Real-time TDM-guided pharmacodynamic target attainment of CI beta-lactam monotherapy allowed to maximize treatment efficacy in most critically ill children with severe Gram-negative infections. Attaining early optimal C ss /MIC ratios of CI beta-lactams could be a key determinant associated with microbiologic eradication during the treatment of Gram-negative infections. Larger prospective studies are warranted for confirming our findings.
Collapse
Affiliation(s)
- Milo Gatti
- From the Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Caterina Campoli
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria Elena Latrofa
- Pediatric Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Tommaso Sasso
- Pediatric Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Rita Mancini
- LUM Metropolitan Laboratory, AUSL Bologna, Bologna, Italy
| | - Fabio Caramelli
- Pediatric Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Viale
- From the Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Federico Pea
- From the Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
16
|
Pavia K, Hambrick HR, Paice K, Tang P, Punt N, Kaplan J, Goldstein SL, Vinks AA, Mizuno T, Tang Girdwood S. Cefepime pharmacokinetics in critically ill children and young adults undergoing continuous kidney replacement therapy. J Antimicrob Chemother 2023; 78:2140-2147. [PMID: 37466170 PMCID: PMC10477133 DOI: 10.1093/jac/dkad192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/05/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES Cefepime is an antibiotic commonly used to treat sepsis and is cleared by renal excretion. Cefepime dosing requires adjustment in patients with decreased kidney function and in those receiving continuous kidney replacement therapy (CKRT). We aimed to characterize cefepime PK in a diverse cohort of critically ill paediatric patients on CKRT. METHODS Patients were identified from an ongoing pharmacokinetic/pharmacodynamic (PK/PD) study of beta-lactam antibiotics, and were included if they had received at least two cefepime doses in the ICU and were on CKRT for at least 24 h. PK parameters were estimated using MwPharm++ with Bayesian estimation and a paediatric population PK model. Target attainment was assessed as time of free cefepime concentrations above minimum inhibitory concentration (fT > 1× or 4 × MIC). RESULTS Seven patients were included in the study (ages 2 to 20 years). CKRT indications included liver failure (n = 1), renal failure (n = 4) and fluid overload (n = 2). Total effluent flow rates ranged from 1833 to 3115 (mean 2603) mL/1.73 m2/h, while clearance was 2.11-3.70 (mean 3.0) L/h/70 kg. Effluent flows were lower, but clearance and fT > MIC were similar to paediatric data published previously. Using Pseudomonas aeruginosa MIC breakpoints, all patients had 100% of dosing interval above MIC, but only one had 100% of dosing interval above 4× MIC. CONCLUSIONS Since most patients failed to attain stringent targets of 100% fT > 4× MIC, model-informed precision dosing may benefit such patients.
Collapse
Affiliation(s)
- Kathryn Pavia
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - H Rhodes Hambrick
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kelli Paice
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Peter Tang
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nieko Punt
- University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
- Medimatics, Maastricht, The Netherlands
| | - Jennifer Kaplan
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sonya Tang Girdwood
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Hospital Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
17
|
Zheng P, Mo L, Zhao B, Li L, Cen B, Xu Z, Li Y. Pharmaceutical care model in precision medicine in China. FARMACIA HOSPITALARIA 2023; 47:T218-T223. [PMID: 37598018 DOI: 10.1016/j.farma.2023.07.004] [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: 11/27/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 08/21/2023] Open
Abstract
Pharmacy service is to provide individualized pharmaceutical care for patients, which should follow the current evidence-based pharmacy, and constantly verify the evidence and then produce new evidence. In pharmaceutical care, differences are often found in the efficacy and adverse reactions of drugs among individuals, even within individuals, which are closely related to patients' genetics, liver and kidney functions, disease states, and drug interactions. Back in the 1980s, therapeutic drug monitoring (TDM) has been applied to routinely monitor the blood drug concentration of patients taking antiepileptic drugs or immunosuppressants after transplantation to provide individualized dosage recommendations and accumulate a large amount of pharmacokinetic (PK)/pharmacodynamic (PD) data. As individualized pharmaceutical care proceeds, the concept of precision medicine was introduced into pharmacy services in combination with evidence-based pharmacy, PK/PD theories, and big data to further promote the TDM technology and drugs, and carry out pharmacogenomics analysis. The TDM and pharmacogenomics have been applied gradually to the fields of antimicrobial, antitumor, and antipsychotic drugs and immunosuppressants. Based on the concept of precision pharmacy, we adopted approaches including PK/PD, quantitative pharmacology, population pharmacokinetics, and big data machine learning to provide more personalized pharmacy services, which is mainly for special patients, such as critical patients, patients with interaction risk of multiple drugs, patients with liver and renal insufficiency, pregnant women, children, and elderly patients. As the service pattern of precision pharmacy has been constructed and constantly improved, better evidence in clinical practice will be produced to provide patients with better precision pharmacy service.
Collapse
Affiliation(s)
- Ping Zheng
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| | - Liqian Mo
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| | - Boxin Zhao
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| | - Liren Li
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| | - Baihong Cen
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| | - Zhongyuan Xu
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China.
| | - Yilei Li
- Unidad de Farmacia Clínica, Hospital Nanfang, Universidad Médica del Sur, Guangzhou, China
| |
Collapse
|
18
|
Zheng P, Mo L, Zhao B, Li L, Cen B, Xu Z, Li Y. Pharmaceutical care model in precision medicine in China. FARMACIA HOSPITALARIA 2023; 47:218-223. [PMID: 37248115 DOI: 10.1016/j.farma.2023.04.005] [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: 11/27/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Pharmacy service is to provide individualized pharmaceutical care for patients, which should follow the current evidence-based pharmacy, and constantly verify the evidence and then produce new evidence. In pharmaceutical care, differences are often found in the efficacy and adverse reactions of drugs among individuals, even within individuals, which are closely related to patient's genetics, liver and kidney functions, disease states, and drug interactions. Back in the 1980s, therapeutic drug monitoring (TDM) has been applied to routinely monitor the blood drug concentration of patients taking antiepileptic drugs or immunosuppressants after transplantation to provide individualized dosage recommendations and accumulate a large amount of pharmacokinetic (PK)/pharmacodynamic (PD) data. As individualized pharmaceutical care proceeds, the concept of precision medicine was introduced into pharmacy services in combination with evidence-based pharmacy, PK/PD theories and big data to further promote the TDM technology and drugs, and carry out pharmacogenomics analysis. The TDM and pharmacogenomics have been applied gradually to the fields of antimicrobial, antitumor and antipsychotic drugs and immunosuppressants. Based on the concept of precision pharmacy, we adpoted approaches including PK/PD, quantitative pharmacology, population pharmacokinetics, and big data machine learning to provide more personalized pharmacy services, which is mainly for special patients, such as critical patients, patients with interaction risk of multiple drugs, patients with liver and renal insufficiency, pregnant women, children and elderly patients. As the service pattern of precision pharmacy has been constructed and constantly improved, better evidence in clinical practice will be produced to provide patients with better precision pharmacy service.
Collapse
Affiliation(s)
- Ping Zheng
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China
| | - Liqian Mo
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China
| | - Boxin Zhao
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China
| | - Liren Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China
| | - Baihong Cen
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China.
| | - Yilei Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
19
|
Venuti F, Romani L, De Luca M, Tripiciano C, Palma P, Chiriaco M, Finocchi A, Lancella L. Novel Beta Lactam Antibiotics for the Treatment of Multidrug-Resistant Gram-Negative Infections in Children: A Narrative Review. Microorganisms 2023; 11:1798. [PMID: 37512970 PMCID: PMC10385558 DOI: 10.3390/microorganisms11071798] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Infections due to carbapenem-resistant Enterobacterales (CRE) are increasingly prevalent in children and are associated with poor clinical outcomes, especially in critically ill patients. Novel beta lactam antibiotics, including ceftolozane-tazobactam, ceftazidime-avibactam, meropenem-vaborbactam, imipenem-cilastatin-relebactam, and cefiderocol, have been released in recent years to face the emerging challenge of multidrug-resistant (MDR) Gram-negative bacteria. Nonetheless, several novel agents lack pediatric indications approved by the Food and Drug Administration (FDA) and the European Medicine Agency (EMA), leading to uncertain pediatric-specific treatment strategies and uncertain dosing regimens in the pediatric population. In this narrative review we have summarized the available clinical and pharmacological data, current limitations and future prospects of novel beta lactam antibiotics in the pediatric population.
Collapse
Affiliation(s)
- Francesco Venuti
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, 10149 Torino, Italy
| | - Lorenza Romani
- Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Maia De Luca
- Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Costanza Tripiciano
- Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Paolo Palma
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Chiriaco
- Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Andrea Finocchi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Laura Lancella
- Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
20
|
Gatti M, Pea F. The expert clinical pharmacological advice program for tailoring on real-time antimicrobial therapies with emerging TDM candidates in special populations: how the ugly duckling turned into a swan. Expert Rev Clin Pharmacol 2023; 16:1035-1051. [PMID: 37874608 DOI: 10.1080/17512433.2023.2274984] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION The growing spread of infections caused by multidrug-resistant pathogens makes the need of tailoring antimicrobial therapies by means of a 'patient-centered' approach fundamental. In this scenario, therapeutic drug monitoring (TDM) of emerging antimicrobial candidates may be a valuable approach, but expert interpretation of TDM results should be granted for making them more clinically useful. The MD Clinical Pharmacologist may take over this task since this specialist may couple PK/PD expertise on drugs with a medical background and may provide expert interpretation of TDM results of antimicrobials for tailoring therapy on real-time in each single patient based on specific both drug/pathogen issues and patient issues. AREAS COVERED This article aims to highlight the main key-points and organizational aspects for implementing a successful TDM-based expert clinical pharmacological advice (ECPA) program for tailoring antimicrobial therapies on real-time in different hospitalized patient special populations. EXPERT OPINION TDM-based ECPA programs lead by the MD Clinical Pharmacologist may represent a way forward for maximizing clinical efficacy and for minimizing the risk of resistance developments and/or toxicity of antimicrobials. Stakeholders should be aware of the fact that this innovative approach may be cost-effective.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
21
|
Črček M, Grabnar I, Zdovc JA, Grosek Š, Kos MK. External validation of population pharmacokinetic models of gentamicin in paediatric population from preterm newborns to adolescents. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:175-194. [PMID: 37307377 DOI: 10.2478/acph-2023-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 06/14/2023]
Abstract
The aim of this study was to externally validate the predictive performance of published population pharmacokinetic models of gentamicin in all paediatric age groups, from preterm newborns to adolescents. We first selected published population pharmacokinetic models of gentamicin developed in the paediatric population with a wide age range. The parameters of the literature models were then re-estimated using the PRIOR subroutine in NONMEM®. The predictive ability of the literature and the tweaked models was evaluated. Retrospectively collected data from a routine clinical practice (512 concentrations from 308 patients) were used for validation. The models with covariates characterising developmental changes in clearance and volume of distribution had better predictive performance, which improved further after re-estimation. The tweaked model by Wang 2019 performed best, with suitable accuracy and precision across the complete paediatric population. For patients treated in the intensive care unit, a lower proportion of patients would be expected to reach the target trough concentration at standard dosing. The selected model could be used for model-informed precision dosing in clinical settings where the entire paediatric population is treated. However, for use in clinical practice, the next step should include additional analysis of the impact of intensive care treatment on gentamicin pharmacokinetics, followed by prospective validation.
Collapse
Affiliation(s)
- Mateja Črček
- 1University of Ljubljana, Faculty of Pharmacy, Department of Biopharmacy and Pharmacokinetics, 1000 Ljubljana Slovenia
| | - Iztok Grabnar
- 1University of Ljubljana, Faculty of Pharmacy, Department of Biopharmacy and Pharmacokinetics, 1000 Ljubljana Slovenia
| | - Jurij Aguiar Zdovc
- 1University of Ljubljana, Faculty of Pharmacy, Department of Biopharmacy and Pharmacokinetics, 1000 Ljubljana Slovenia
| | - Štefan Grosek
- 2University of Ljubljana, Faculty of Medicine, Department of Pediatrics 1000 Ljubljana, Slovenia
- 3University Medical Centre Ljubljana Division of Obstetrics and Gynecology, Department of Perinatology Neonatology Section, 1000 Ljubljana Slovenia
- 4University Medical Centre Ljubljana Division of Paediatrics, Department of Paediatric Intensive Therapy, 1000 Ljubljana, Slovenia
| | - Mojca Kerec Kos
- 1University of Ljubljana, Faculty of Pharmacy, Department of Biopharmacy and Pharmacokinetics, 1000 Ljubljana Slovenia
| |
Collapse
|
22
|
van der Veer MAA, de Haan TR, Franken LGW, Hodiamont CJ, Groenendaal F, Dijk PH, de Boode WP, Simons S, Dijkman KP, van Straaten HLM, Rijken M, Cools F, Nuytemans DHGM, van Kaam AH, Bijleveld YA, Mathôt RAA. Population Pharmacokinetics and Dosing Optimization of Ceftazidime in Term Asphyxiated Neonates during Controlled Therapeutic Hypothermia. Antimicrob Agents Chemother 2023; 67:e0170722. [PMID: 37010414 PMCID: PMC10190683 DOI: 10.1128/aac.01707-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/23/2023] [Indexed: 04/04/2023] Open
Abstract
Ceftazidime is an antibiotic commonly used to treat bacterial infections in term neonates undergoing controlled therapeutic hypothermia (TH) for hypoxic-ischemic encephalopathy after perinatal asphyxia. We aimed to describe the population pharmacokinetics (PK) of ceftazidime in asphyxiated neonates during hypothermia, rewarming, and normothermia and propose a population-based rational dosing regimen with optimal PK/pharmacodynamic (PD) target attainment. Data were collected in the PharmaCool prospective observational multicenter study. A population PK model was constructed, and the probability of target attainment (PTA) was assessed during all phases of controlled TH using targets of 100% of the time that the concentration in the blood exceeds the MIC (T>MIC) (for efficacy purposes and 100% T>4×MIC and 100% T>5×MIC to prevent resistance). A total of 35 patients with 338 ceftazidime concentrations were included. An allometrically scaled one-compartment model with postnatal age and body temperature as covariates on clearance was constructed. For a typical patient receiving the current dose of 100 mg/kg of body weight/day in 2 doses and assuming a worst-case MIC of 8 mg/L for Pseudomonas aeruginosa, the PTA was 99.7% for 100% T>MIC during hypothermia (33.7°C; postnatal age [PNA] of 2 days). The PTA decreased to 87.7% for 100% T>MIC during normothermia (36.7°C; PNA of 5 days). Therefore, a dosing regimen of 100 mg/kg/day in 2 doses during hypothermia and rewarming and 150 mg/kg/day in 3 doses during the following normothermic phase is advised. Higher-dosing regimens (150 mg/kg/day in 3 doses during hypothermia and 200 mg/kg/day in 4 doses during normothermia) could be considered when achievements of 100% T>4×MIC and 100% T>5×MIC are desired.
Collapse
Affiliation(s)
- Marlotte A. A. van der Veer
- Department of Hospital Pharmacology and Clinical Pharmacology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Timo R. de Haan
- Department of Neonatology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Linda G. W. Franken
- Department of Hospital Pharmacology and Clinical Pharmacology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Caspar J. Hodiamont
- Medical Microbiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children’s Hospital, Utrecht, The Netherlands
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter H. Dijk
- University Medical Center Groningen, Beatrix Children’s Hospital, Department of Pediatrics, Division of Neonatology, University of Groningen, Groningen, The Netherlands
| | - Willem P. de Boode
- Department of Neonatology, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Nijmegen, The Netherlands
| | - Sinno Simons
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Koen P. Dijkman
- Department of Neonatology, Máxima Medical Center Veldhoven, Veldhoven, The Netherlands
| | | | - Monique Rijken
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Filip Cools
- Department of Neonatology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Debbie H. G. M. Nuytemans
- Department of Neonatology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Anton H. van Kaam
- Department of Neonatology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Yuma A. Bijleveld
- Department of Hospital Pharmacology and Clinical Pharmacology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A. A. Mathôt
- Department of Hospital Pharmacology and Clinical Pharmacology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Leegwater E, Wewerinke L, de Grauw AM, van Veen M, Storm BN, Kruizinga MD. Optimization of β-Lactam Dosing Regimens in Neonatal Infections: Continuous and Extended Administration versus Intermittent Administration. Clin Pharmacokinet 2023; 62:715-724. [PMID: 36972008 DOI: 10.1007/s40262-023-01230-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND AND OBJECTIVE In neonates, β-Lactam antibiotics are almost exclusively administered by intermittent infusion. However, continuous or prolonged infusion may be more beneficial because of the time-dependent antibacterial activity. In this pharmacokinetic/pharmacodynamic simulation study, we aimed to compare treatment with continuous, extended and intermittent infusion of β-lactam antibiotics for neonates with infectious diseases. METHODS We selected population pharmacokinetic models of penicillin G, amoxicillin, flucloxacillin, cefotaxime, ceftazidime and meropenem, and performed a Monte Carlo simulation with 30,000 neonates. Four different dosing regimens were simulated: intermittent infusion in 30 min, prolonged infusion in 4 h, continuous infusion, and continuous infusion with a loading dose. The primary endpoint was 90% probability of target attainment (PTA) for 100% ƒT>MIC during the first 48 h of treatment. RESULTS For all antibiotics except cefotaxime, continuous infusion with a loading dose resulted in a higher PTA compared with other dosing regimens. Sufficient exposure (PTA >90%) using continuous infusion with a loading dose was reached for amoxicillin (90.3%), penicillin G (PTA 98.4%), flucloxacillin (PTA 94.3%), cefotaxime (PTA 100%), and ceftazidime (PTA 100%). Independent of dosing regimen, higher meropenem (PTA for continuous infusion with a loading dose of 85.5%) doses might be needed to treat severe infections in neonates. Ceftazidime and cefotaxime dose might be unnecessarily high, as even with dose reductions, a PTA > 90% was retained. CONCLUSIONS Continuous infusion after a loading dose leads to a higher PTA compared with continuous, intermittent or prolonged infusion, and therefore has the potential to improve treatment with β-lactam antibiotics in neonates.
Collapse
Affiliation(s)
- Emiel Leegwater
- The Hague Hospital Pharmacy, The Hague, The Netherlands.
- Department of Hospital Pharmacy, Haga Teaching Hospital, The Hague, The Netherlands.
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden University, Leiden, The Netherlands.
| | - Leo Wewerinke
- Juliana Children's Hospital, Haga Teaching Hospital, The Hague, The Netherlands
| | - Anne M de Grauw
- Juliana Children's Hospital, Haga Teaching Hospital, The Hague, The Netherlands
| | - Mirjam van Veen
- Juliana Children's Hospital, Haga Teaching Hospital, The Hague, The Netherlands
| | - Bert N Storm
- Department of Hospital Pharmacy, Haga Teaching Hospital, The Hague, The Netherlands
| | - Matthijs D Kruizinga
- Juliana Children's Hospital, Haga Teaching Hospital, The Hague, The Netherlands
- Department of Paediatrics, Willem Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Hollander EM, van Tuinen EL, Schölvinck EH, Bergman KA, Bourgonje AR, Gracchi V, Kneyber MCJ, Touw DJ, Mian P. Evaluation of Dosing Guidelines for Gentamicin in Neonates and Children. Antibiotics (Basel) 2023; 12:antibiotics12050810. [PMID: 37237713 DOI: 10.3390/antibiotics12050810] [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: 03/24/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Although aminoglycosides are frequently prescribed to neonates and children, the ability to reach effective and safe target concentrations with the currently used dosing regimens remains unclear. This study aims to evaluate the target attainment of the currently used dosing regimens for gentamicin in neonates and children. We conducted a retrospective single-center cohort study in neonates and children receiving gentamicin between January 2019 and July 2022, in the Beatrix Children's Hospital. The first gentamicin concentration used for therapeutic drug monitoring was collected for each patient, in conjunction with information on dosing and clinical status. Target trough concentrations were ≤1 mg/L for neonates and ≤0.5 mg/L for children. Target peak concentrations were 8-12 mg/L for neonates and 15-20 mg/L for children. In total, 658 patients were included (335 neonates and 323 children). Trough concentrations were outside the target range in 46.2% and 9.9% of neonates and children, respectively. Peak concentrations were outside the target range in 46.0% and 68.7% of neonates and children, respectively. In children, higher creatinine concentrations were associated with higher gentamicin trough concentrations. This study corroborates earlier observational studies showing that, with a standard dose, drug concentration targets were met in only approximately 50% of the cases. Our findings show that additional parameters are needed to improve target attainment.
Collapse
Affiliation(s)
- Esther M Hollander
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Eline L van Tuinen
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Elisabeth H Schölvinck
- Department of Pediatric Infectious Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Klasien A Bergman
- Division of Neonatology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Valentina Gracchi
- Division of Pediatric Nephrology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Martin C J Kneyber
- Division of Peadiatric Critical Care Medicine, Department of Paediatrics, Beatrix Children's Hospital Groningen, University Medical Center Groningen, University of Groningen Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute for Pharmacy, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
25
|
Aquino M, Tinoco M, Bicker J, Falcão A, Rocha M, Fortuna A. Therapeutic Drug Monitoring of Amikacin in Neutropenic Oncology Patients. Antibiotics (Basel) 2023; 12:antibiotics12020373. [PMID: 36830283 PMCID: PMC9952017 DOI: 10.3390/antibiotics12020373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Amikacin is the antibiotic of choice for the treatment of Gram-negative infections, namely, those in neutropenic oncology patients. No populational pharmacokinetic studies are currently available reporting amikacin pharmacokinetics in neutropenic oncology patients despite their specific pathophysiological features and treatments. A large-scale retrospective study was herein conducted to specifically investigate the effects that tumor diseases have on the pharmacokinetic parameters of amikacin and identify whether chemotherapy, the lag time between administration of chemotherapy and amikacin, age and renal function contribute to amikacin pharmacokinetics in neutropenic cancer patients. A total of 1180 pharmacokinetic analysis from 629 neutropenic patients were enrolled. The daily dose administered to oncology patients was higher than that administered to non-oncology patients (p < 0.0001). No statistical differences were found in amikacin concentrations, probably because drug clearance was increased in cancer patients (p < 0.0001). Chemotherapy influenced amikacin pharmacokinetics and drug clearance decreased as the lag time enhanced. The elderly group revealed no statistical differences between the doses administered to both the oncology groups, suggesting that the impact of ageing is stronger than chemotherapy. Our research suggests that cancer patients require higher initial doses of amikacin, as well as when chemotherapy is received less than 30 days before amikacin treatment has started.
Collapse
Affiliation(s)
- Maria Aquino
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Tinoco
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Marília Rocha
- Centro Hospitalar e Universitário de Coimbra (CHUC, EPE), 3000-548 Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
26
|
Singu BS, Ndeunyema MN, Ette EI, Pieper CH, Verbeeck RK. Plasma concentration and eGFR in preterm and term neonates receiving gentamicin or successive amikacin therapy. BMC Pediatr 2023; 23:24. [PMID: 36647065 PMCID: PMC9841723 DOI: 10.1186/s12887-023-03834-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Gentamicin and amikacin are aminoglycoside antibiotics which are renally excreted and known to be nephrotoxic. Estimate of glomerular filtration rate (eGFR) per body surface area is lower in neonates than in adults and exposure to these drugs could lead to more suppression in kidney function. The aim of this study was to determine maximum and minimum plasma concentrations (Cmax and Cmin), time to reach Cmin levels of gentamicin and amikacin, and to assess eGFR in preterm and term neonates. METHODS Two groups of patients were recruited, 44 neonates receiving gentamicin (5 mg/kg/24 h) and 35 neonates receiving amikacin (15 mg/kg/24 h) by slow intravenous injection. Patients on amikacin had been on gentamicin before being switched to amikacin. Two blood samples were drawn for the determination of the maximum and minimum plasma concentration. Primary outcomes were determination of Cmax, Cmin, and the time it took to clear the aminoglycoside to a plasma concentration below the toxicity threshold (gentamicin: < 1 mcg/mL; amikacin: < 5 mcg/mL. RESULTS Therapeutic range for Cmax of gentamicin (15-25 mcg/mL) or amikacin (30-40 mcg/mL) was achieved in only 27.3 and 2.9% of neonates, respectively. Percentage of neonates reaching plasma concentrations below the toxicity threshold within the 24-hour dosing interval was 72.7% for gentamicin and 97.1% for amikacin. Positive correlation between gentamicin clearance and postnatal age borderline statistical significance (p = 0.007), while the correlation between amikacin clearance and postnatal age was poor and not statistically significant (r2 = - 0.30, p = 0.971). CONCLUSION Although eGFR decreased significantly as a function of postnatal age in neonates receiving amikacin, the majority (91.4%) of these neonates were able to clear the drug to < 5 mcg/mL within a 24-hour dosing interval.
Collapse
Affiliation(s)
- Bonifasius Siyuka Singu
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Private Bag, Windhoek, 13301 Namibia
| | - Milka Ndapandula Ndeunyema
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Private Bag, Windhoek, 13301 Namibia
| | - Ene I. Ette
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Private Bag, Windhoek, 13301 Namibia
| | | | - Roger Karel Verbeeck
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Private Bag, Windhoek, 13301 Namibia
| |
Collapse
|
27
|
Franzese R, Riccobene T, Carrothers T, Vourvahis M, Winter E, Lovern M, McFadyen L. Population Pharmacokinetic Modeling for Ceftazidime-Avibactam Renal Dose Adjustments in Pediatric Patients 3 months and Older. Clin Pharmacol Ther 2023; 113:182-195. [PMID: 36239478 DOI: 10.1002/cpt.2764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/02/2022] [Indexed: 12/24/2022]
Abstract
Ceftazidime-avibactam is a novel β-lactam/β-lactamase inhibitor combination developed to treat serious Gram-negative bacterial infections; approved indications include complicated urinary tract infection, complicated intra-abdominal infection, and hospital-acquired pneumonia including ventilator-associated pneumonia in patients ≥ 3 months old. Because of the predominantly renal clearance of ceftazidime and avibactam, dose adjustments (reductions) are required for patients with estimated creatinine clearance (CrCL) ≤ 50 mL/min. We describe the application of combined adult and pediatric population pharmacokinetic models in developing ceftazidime-avibactam dose recommendations for pediatric patients ≥ 2 to < 18 years old with body surface area-normalized CrCL ≤ 50 mL/min/1.73 m2 , including moderate, severe, or very severe renal impairment, or end-stage renal disease requiring hemodialysis, and for patients ≥ 3 months to < 2 years old with mild, moderate, or severe renal impairment. Models included allometric scaling for all subjects and simulations (1,000 subjects per age group, renal function group, and indication) were performed nonparametrically using post hoc random effects. Doses were selected based on simulated pediatric patients achieving steady-state exposures similar to adults and high probability of target attainment (using a simultaneous joint target for both ceftazidime and avibactam). Because there were few children with renal impairment in the ceftazidime-avibactam clinical trials, selected pediatric doses were guided by extrapolation and matching of adult exposures associated with efficacy and within established safety margins. The recommended doses for pediatric patients with estimated CrCL ≤ 50 mL/min/1.73 m2 use equivalent adjustments in dose quantity and/or administration interval (vs. the corresponding age group with normal renal function) as those for adults.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark Lovern
- Certara Strategic Consulting, Raleigh, North Carolina, USA
| | | |
Collapse
|
28
|
Kato H, Hagihara M, Matsuda H, Iwamoto T. Gentamicin Pharmacokinetics and Optimal Dosage in Infant Patients: A Case Report and Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15360. [PMID: 36430078 PMCID: PMC9691146 DOI: 10.3390/ijerph192215360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 06/16/2023]
Abstract
Gentamicin is an aminoglycoside antibiotic that is mostly used for the pediatric population. While the pediatric population is classified into neonates, infants, children, and adolescents based on developmental or maturational changes, infants are often overlooked in research. Three infant cases receiving gentamicin are presented to illustrate the pharmacokinetics and optimum dosage of gentamicin. Three infant patients received gentamicin (5.6-7.5 mg/kg/day) for urinary tract infections (UTIs) or bacteremia caused by Enterobacter aerogenes. The trough (Cmin) and peak (Cpeak) concentrations of gentamicin were 0.2-1.8 and 8.9 mg/L, respectively. The Cmin of a patient receiving gentamicin at 9.0 mg/kg/day was 3.3 mg/L, and the patient showed a decrease in urinary volume. The other two patients fully recovered from the infection and did not experience any adverse events. Additionally, we reviewed three studies regarding infant patients receiving gentamicin. The studies used gentamicin therapy for Gram-negative pathogen infections and UTIs caused by Escherichia coli and Enterococcus faecalis. The Cmin and Cpeak of patients receiving gentamicin at 2.2-7.5 mg/kg/day were 0.58-2.15 mg/kg and 4.67-8.88 mg/L, respectively. All patients were cured without any adverse events. Gentamicin dosages below 7.5 mg/kg/day may be effective and safe for use in infant patients. However, the optimal dosing regimen of gentamicin in infant patients is controversial, and limited data are available.
Collapse
Affiliation(s)
- Hideo Kato
- Department of Pharmacy, Mie University Hospital, Tsu 514-8507, Japan
- Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Mao Hagihara
- Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University Hospital, Nagakute 480-1195, Japan
| | - Hiroko Matsuda
- Department of Pharmacy, Mie University Hospital, Tsu 514-8507, Japan
| | - Takuya Iwamoto
- Department of Pharmacy, Mie University Hospital, Tsu 514-8507, Japan
- Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| |
Collapse
|
29
|
Zhou P, Cheng Y, Cao G, Xing Y, Zhai S, Tong X, Yang K. The OBTAINS study: A nationwide cross-sectional survey on the implementation of extended or continuous infusion of β-lactams and vancomycin among neonatal sepsis patients in China. Front Pharmacol 2022; 13:1001924. [PMID: 36299905 PMCID: PMC9589050 DOI: 10.3389/fphar.2022.1001924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/23/2022] [Indexed: 01/03/2023] Open
Abstract
Background: Dosing strategies of β-lactams and vancomycin should be optimized according to pharmacokinetic/pharmacodynamic principles. However, there is no available data indicating the implementation of extended infusion (EI) or continuous infusion (CI) administration in the management of neonatal sepsis. Methods: A nationwide cross-sectional survey was conducted and the pediatricians from 31 provinces in China were enrolled. A multidisciplinary team created the questionnaire, which had three sections and a total of 21 questions with open- and closed-ended responses. The survey was then conducted using an internet platform in an anonymous way. The data was eventually gathered, compiled, and examined. To identify the risk factors associated with the implementation of EI/CI, logistic regression was carried out. Results: A total of 1501 respondents answered the questionnaires. The implementation of EI/CI of β-lactams and vancomycin were only available to one-third of the respondents, and the prolonged strategy was primarily supported by guidelines (71.25%) and advice from medical specialists (55.18%). A significant fraction (72.94%-94.71%) lacked a strong understanding of the infusions' stability. Additionally, it was discovered that more frequent MDT discussions about antibiotic use and the appropriate time pediatricians worked in the neonatal ward were associated with an increase in the use of the EI/CI strategy. Conclusion: The EI/CI strategy in neonatal sepsis was not well recognized in China, and it is necessary to establish a solid MDT team with regularly collaborates. In the near future, guidelines regarding prolonged infusion management in neonatal sepsis should be developed.
Collapse
Affiliation(s)
- Pengxiang Zhou
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China,Department of Pharmacy, Peking University Third Hospital, Beijing, China,Institute for Drug Evaluation, Peking University Health Science Center, Beijing, China
| | - Yinchu Cheng
- Department of Pharmacy, Peking University Third Hospital, Beijing, China,Institute for Drug Evaluation, Peking University Health Science Center, Beijing, China
| | - Guangna Cao
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Suodi Zhai
- Department of Pharmacy, Peking University Third Hospital, Beijing, China,Institute for Drug Evaluation, Peking University Health Science Center, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China,*Correspondence: Kehu Yang, ; Xiaomei Tong,
| | - Kehu Yang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China,*Correspondence: Kehu Yang, ; Xiaomei Tong,
| |
Collapse
|
30
|
Luxton TN, King N, Wälti C, Jeuken LJC, Sandoe JAT. A Systematic Review of the Effect of Therapeutic Drug Monitoring on Patient Health Outcomes during Treatment with Carbapenems. Antibiotics (Basel) 2022; 11:antibiotics11101311. [PMID: 36289971 PMCID: PMC9598625 DOI: 10.3390/antibiotics11101311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Adjusting dosing regimens based on measurements of carbapenem levels may improve carbapenem exposure in patients. This systematic review aims to describe the effect carbapenem therapeutic drug monitoring (TDM) has on health outcomes, including the emergence of antimicrobial resistance (AMR). Four databases were searched for studies that reported health outcomes following adjustment to dosing regimens, according to measurements of carbapenem concentration. Bias in the studies was assessed with risk of bias analysis tools. Study characteristics and outcomes were tabulated and a narrative synthesis was performed. In total, 2 randomised controlled trials (RCTs), 17 non-randomised studies, and 19 clinical case studies were included. Significant variation in TDM practice was seen; consequently, a meta-analysis was unsuitable. Few studies assessed impacts on AMR. No significant improvement on health outcomes and no detrimental effects of carbapenem TDM were observed. Five cohort studies showed significant associations between achieving target concentrations and clinical success, including suppression of resistance. Studies in this review showed no obvious improvement in clinical outcomes when TDM is implemented. Optimisation and standardisation of carbapenem TDM practice are needed to improve intervention success and enable study synthesis. Further suitably powered studies of standardised TDM are required to assess the impact of TMD on clinical outcomes and AMR.
Collapse
Affiliation(s)
- Timothy N. Luxton
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
- Correspondence:
| | - Natalie King
- Leeds Institute of Health Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Lars J. C. Jeuken
- Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | |
Collapse
|
31
|
Haseeb A, Faidah HS, Alghamdi S, Alotaibi AF, Elrggal ME, Mahrous AJ, Abuhussain SSA, Obaid NA, Algethamy M, AlQarni A, Khogeer AA, Saleem Z, Iqbal MS, Ashgar SS, Radwan RM, Mutlaq A, Fatani N, Sheikh A. Dose optimization of β-lactams antibiotics in pediatrics and adults: A systematic review. Front Pharmacol 2022; 13:964005. [PMID: 36210807 PMCID: PMC9532942 DOI: 10.3389/fphar.2022.964005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background: β-lactams remain the cornerstone of the empirical therapy to treat various bacterial infections. This systematic review aimed to analyze the data describing the dosing regimen of β-lactams. Methods: Systematic scientific and grey literature was performed in accordance with Preferred Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. The studies were retrieved and screened on the basis of pre-defined exclusion and inclusion criteria. The cohort studies, randomized controlled trials (RCT) and case reports that reported the dosing schedule of β-lactams are included in this study. Results: A total of 52 studies met the inclusion criteria, of which 40 were cohort studies, 2 were case reports and 10 were RCTs. The majority of the studies (34/52) studied the pharmacokinetic (PK) parameters of a drug. A total of 20 studies proposed dosing schedule in pediatrics while 32 studies proposed dosing regimen among adults. Piperacillin (12/52) and Meropenem (11/52) were the most commonly used β-lactams used in hospitalized patients. As per available evidence, continuous infusion is considered as the most appropriate mode of administration to optimize the safety and efficacy of the treatment and improve the clinical outcomes. Conclusion: Appropriate antibiotic therapy is challenging due to pathophysiological changes among different age groups. The optimization of pharmacokinetic/pharmacodynamic parameters is useful to support alternative dosing regimens such as an increase in dosing interval, continuous infusion, and increased bolus doses.
Collapse
Affiliation(s)
- Abdul Haseeb
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hani Saleh Faidah
- Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saleh Alghamdi
- Department of Clinical Pharmacy, Faculty of Clinical Pharmacy, Al Baha University, Al Baha, Saudi Arabia
| | - Amal F. Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mahmoud Essam Elrggal
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmad J. Mahrous
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Najla A. Obaid
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Manal Algethamy
- Department of Infection Prevention and Control Program, Alnoor Specialist Hospital, Makkah, Saudi Arabia
| | - Abdullmoin AlQarni
- Infectious Diseases Department, Alnoor Specialist Hospital, Makkah, Saudi Arabia
| | - Asim A. Khogeer
- Plan and Research Department, General Directorate of Health Affairs of Makkah Region, Ministry of Health, Makkah, Saudi Arabia
- Medical Genetics Unit, Maternity and Children Hospital, Makkah Healthcare Cluster, Ministry of Health, Makkah, Saudi Arabia
| | - Zikria Saleem
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya Univrsity, Multan, Pakistan
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sami S. Ashgar
- Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Rozan Mohammad Radwan
- Pharmaceutical Care Department, Alnoor Specialist Hospital, Ministry of Health, Makkah, Saudi Arabia
| | - Alaa Mutlaq
- General Department of Pharmaceutical Care, Ministry of Health, Riyadh, Saudi Arabia
| | | | - Aziz Sheikh
- Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
32
|
Abstract
OBJECTIVES In critically ill children, severely altered pharmacokinetics may result in subtherapeutic β-lactam antibiotic concentrations when standard pediatric dosing regimens are applied. However, it remains unclear how to recognize patients most at risk for suboptimal exposure and their outcome. This study aimed to: 1) describe target attainment for β-lactam antibiotics in critically ill children, 2) identify risk factors for suboptimal exposure, and 3) study the association between target nonattainment and clinical outcome. DESIGN Post hoc analysis of the "Antibiotic Dosing in Pediatric Intensive Care" study (NCT02456974, 2012-2019). Steady-state trough plasma concentrations were classified as therapeutic if greater than or equal to the minimum inhibitory concentration of the (suspected) pathogen. Factors associated with subtherapeutic concentrations and clinical outcome were identified by logistic regression analysis. SETTING The pediatric and cardiac surgery ICU of a Belgian tertiary-care hospital. PATIENTS One hundred fifty-seven patients (aged 1 mo to 15 yr) treated intravenously with amoxicillin-clavulanic acid, piperacillin-tazobactam, or meropenem. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Three hundred eighty-two trough concentrations were obtained from 157 patients (median age, 1.25 yr; interquartile range, 0.4-4.2 yr). Subtherapeutic concentrations were measured in 39 of 60 (65%), 43 of 48 (90%), and 35 of 49 (71%) of patients treated with amoxicillin-clavulanic acid, piperacillin-tazobactam, and meropenem, respectively. Estimates of glomerular filtration rate (eGFR; 54% increase in odds for each sd increase in value, 95% CI, 0.287-0.736; p = 0.001) and the absence of vasopressor treatment (2.8-fold greater odds, 95% CI, 1.079-7.253; p = 0.034) were independently associated with target nonattainment. We failed to identify an association between antibiotic concentrations and clinical failure. CONCLUSIONS Subtherapeutic β-lactam concentrations are common in critically ill children and correlate with renal function. eGFR equations may be helpful in identifying patients who may require higher dosing. Future studies should focus on the impact of subtherapeutic concentrations on clinical outcome.
Collapse
|
33
|
Use available data to optimise antibiotic use in critically ill children. DRUGS & THERAPY PERSPECTIVES 2022. [DOI: 10.1007/s40267-022-00924-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
34
|
Hodiamont CJ, van den Broek AK, de Vroom SL, Prins JM, Mathôt RAA, van Hest RM. Clinical Pharmacokinetics of Gentamicin in Various Patient Populations and Consequences for Optimal Dosing for Gram-Negative Infections: An Updated Review. Clin Pharmacokinet 2022; 61:1075-1094. [PMID: 35754071 PMCID: PMC9349143 DOI: 10.1007/s40262-022-01143-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2022] [Indexed: 11/04/2022]
Abstract
Gentamicin is an aminoglycoside antibiotic with a small therapeutic window that is currently used primarily as part of short-term empirical combination therapy. Gentamicin dosing schemes still need refinement, especially for subpopulations where pharmacokinetics can differ from pharmacokinetics in the general adult population: obese patients, critically ill patients, paediatric patients, neonates, elderly patients and patients on dialysis. This review summarizes the clinical pharmacokinetics of gentamicin in these patient populations and the consequences for optimal dosing of gentamicin for infections caused by Gram-negative bacteria, highlighting new insights from the last 10 years. In this period, several new population pharmacokinetic studies have focused on these subpopulations, providing insights into the typical values of the most relevant pharmacokinetic parameters, the variability of these parameters and possible explanations for this variability, although unexplained variability often remains high. Both dosing schemes and pharmacokinetic/pharmacodynamic (PK/PD) targets varied widely between these studies. A gentamicin starting dose of 7 mg/kg based on total body weight (or on adjusted body weight in obese patients) appears to be the optimal strategy for increasing the probability of target attainment (PTA) after the first administration for the most commonly used PK/PD targets in adults and children older than 1 month, including critically ill patients. However, evidence that increasing the PTA results in higher efficacy is lacking; no studies were identified that show a correlation between estimated or predicted PK/PD target attainment and clinical success. Although it is unclear if performing therapeutic drug monitoring (TDM) for optimization of the PTA is of clinical value, it is recommended in patients with highly variable pharmacokinetics, including patients from all subpopulations that are critically ill (such as elderly, children and neonates) and patients on intermittent haemodialysis. In addition, TDM for optimization of the dosing interval, targeting a trough concentration of at least < 2 mg/L but preferably < 0.5–1 mg/L, has proven to reduce nephrotoxicity and is therefore recommended in all patients receiving more than one dose of gentamicin. The usefulness of the daily area under the plasma concentration–time curve for predicting nephrotoxicity should be further investigated. Additionally, more research is needed on the optimal PK/PD targets for efficacy in the clinical situations in which gentamicin is currently used, that is, as monotherapy for urinary tract infections or as part of short-term combination therapy.
Collapse
Affiliation(s)
- Caspar J Hodiamont
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Annemieke K van den Broek
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Suzanne L de Vroom
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan M Prins
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ron A A Mathôt
- Hospital Pharmacy and Clinical Pharmacology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Reinier M van Hest
- Hospital Pharmacy and Clinical Pharmacology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Yamada T, Emoto C, Fukuda T, Motomura Y, Inoue H, Ohga S, Ieiri I. Optimal Teicoplanin Dosing Regimen in Neonates and Children Developed by Leveraging Real-World Clinical Information. Ther Drug Monit 2022; 44:404-413. [PMID: 34629445 DOI: 10.1097/ftd.0000000000000930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Teicoplanin is a glycopeptide antibiotic used for the treatment of methicillin-resistant Staphylococcus aureus infections. To ensure successful target attainment, therapeutic drug monitoring-informed dosage adjustment is recommended. However, it relies on the experience of the clinician and the frequency of drug measurements. This study aimed to design a new optimal dosing regimen of teicoplanin with a maintenance dosing strategy for neonates and children based on their physiological characteristics. METHODS Data from teicoplanin-treated patients (n = 214) were collected from electronic medical records. Covariate analyses were performed using population pharmacokinetic (PK) modeling with 399 serum teicoplanin concentrations from 48 neonates and 166 children. Multiple PK simulations were conducted to explore optimal dosing regimens that would allow control of the trough concentration to the target of 15-30 mg/L quicker than the current standard regimen. RESULTS Allometrically scaled body weight, postmenstrual age (PMA), renal function, and serum albumin were implemented as substantial covariates for teicoplanin clearance in a two-compartment PK model. Covariate analyses and comprehensive simulation assessments recommended the following modifications to the current regimen: (1) decreased dose for premature babies (PMA ≤28 weeks), (2) decreased dose for children with renal dysfunction, and (3) increased dose for children (0.5-11 years) with an estimated glomerular filtration rate of ≥90 mL/min/1.73 m2. CONCLUSIONS This study leverages real-world clinical information and proposes new optimal dosing regimens for teicoplanin in neonates and children through PK modeling and simulation analyses, taking into account the age, including PMA, and renal function of patients.
Collapse
Affiliation(s)
- Takaaki Yamada
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Chie Emoto
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
| | - Tsuyoshi Fukuda
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University
- National Center for Child Health and Development, Tokyo, Japan; and
| | - Yoshitomo Motomura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirosuke Inoue
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
36
|
Al-Eyadhy A, Al-Jelaify MR. Suboptimal vancomycin levels in critically ill children with sickle cell disease and acute chest syndrome. J Infect Chemother 2022; 28:1304-1309. [DOI: 10.1016/j.jiac.2022.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/24/2022] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
|
37
|
Luxton T, King N, Wälti C, Jeuken L, Sandoe J. A systematic review of the effect of therapeutic drug monitoring on patient health outcomes during treatment with penicillins. J Antimicrob Chemother 2022; 77:1532-1541. [PMID: 35355067 PMCID: PMC9155611 DOI: 10.1093/jac/dkac101] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/03/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Dosing regimens guided by therapeutic drug monitoring (TDM) may be able to improve penicillin exposure in patients, which could result in improved patient health outcomes. OBJECTIVES This systematic review aims to describe the impact penicillin TDM has on health outcomes, including antimicrobial resistance (AMR). METHODS Studies measuring penicillins in patient samples that adjusted regimens according to the result, and reported health outcomes were selected. Study bias was assessed according to study type. Included study characteristics were tabulated and described by narrative synthesis. RESULTS Three randomized controlled trials (RCTs), 16 cohort studies, and 9 case studies were included. No RCTs showed statistically significant improvements in health outcomes. Five cohort studies showed improvement in at least one health outcome associated with target attainment. However, there was a high risk of bias in all studies for health outcomes. One study assessed the impact of penicillin TDM on AMR and found that improved target attainment was associated with suppression of resistance. No studies found a detrimental effect of penicillin TDM. CONCLUSIONS There is little evidence to suggest that TDM improves health outcomes, however neither health outcomes nor impact on AMR were adequately addressed. Variations in TDM implementation meant that a meta-analysis was not suitable. Penicillin TDM needs standardization, however there is currently no clear evidence of optimal conditions. Suitably powered studies are required to resolve the ambiguity surrounding the impact of TDM on clinical outcomes, including AMR. Further, standardized protocols and concentration targets need to be identified for TDM to be implemented successfully.
Collapse
Affiliation(s)
- Timothy Luxton
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Natalie King
- Leeds Institute of Health Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Lars Jeuken
- Leeds Institute of Health Sciences, University of Leeds, Leeds LS2 9JT, UK
- Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA, Leiden, The Netherlands
| | | |
Collapse
|
38
|
C/MIC > 4: A Potential Instrument to Predict the Efficacy of Meropenem. Antibiotics (Basel) 2022; 11:antibiotics11050670. [PMID: 35625314 PMCID: PMC9137711 DOI: 10.3390/antibiotics11050670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/01/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
This prospective study aimed to explore the determinants of meropenem trough concentration (Ctrough) in patients with bacterial pneumonia and to investigate the association between its concentration and efficacy. From January 2019 to December 2019, patients with pulmonary infections were prospectively enrolled from the intensive care unit. Factors affecting the meropenem trough concentration were analyzed, and a multiple linear regression model was constructed. Logistic regression analyses were used to investigate the relationship between Ctrough and clinical efficacy. A total of 64 patients were enrolled, in whom 210 meropenem concentrations were measured. Of the total, 60.9% (39/64) were considered clinically successful after treatment. Ctrough may increase with increased blood urea nitrogen, albumin, and concomitant antifungal use. By contrast, concentration may decrease with increased endogenous creatinine clearance rate. Six variables, including Ctrough/minimum inhibitory concentration (MIC) > 4, were associated with the efficacy of meropenem. There was an independent correlation between Ctrough/MIC > 4 and efficacy after fully adjusting for confounding factors. Based upon renal function indexes, it is possible to predict changes in meropenem concentration and adjust the dosage precisely and individually. Ctrough/MIC > 4 is a potential instrument to predict successful treatment with meropenem.
Collapse
|
39
|
Effect of Early Nutritional Assessment and Nutritional Support on Immune Function and Clinical Prognosis of Critically Ill Children. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7100238. [PMID: 35035853 PMCID: PMC8759854 DOI: 10.1155/2022/7100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 11/18/2022]
Abstract
The aim of this study was to study the effect of early nutritional assessment and nutritional support on immune function and clinical prognosis of critically ill children. 90 critically ill children at the same level of severity admitted to the pediatric intensive care unit (PICU) of our hospital (June 2019-June 2020) were chosen as the research objects and were equally separated into the experimental group and the control group by the random number table method. The children in the control group were admitted to the PICU according to the routine process, and the nutritional support was provided to the malnourished ones. After admission to the PICU, the children in the experimental group were given nutritional assessment, nutritional risk screening, and nutritional support according to the screening results. The PICU stay time and total hospitalization time of the experimental group were obviously shorter than those of the control group (P < 0.05), the hospitalization expenses of the experimental group were obviously lower than those of the control group (P < 0.05), the clinical outcomes and immune function of the experimental group were obviously better than those of the control group (P < 0.05), and the nutrition indicators of the experimental group were obviously higher than those of the control group (P < 0.05). Early nutritional assessment and nutritional support can effectively improve the immune function and reduce the incidence of adverse clinical outcomes of critically ill children, which are worthy of clinical application and promotion.
Collapse
|
40
|
Imburgia TA, Engdahl SR, Pettit RS. Evaluation of the safety of cefepime prolonged infusions in pediatric patients with cystic fibrosis. Pediatr Pulmonol 2022; 57:919-925. [PMID: 34989183 DOI: 10.1002/ppul.25817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pediatric cystic fibrosis (CF) patients possess unique pharmacokinetics and may benefit from prolonged beta-lactam infusions to optimize pharmacodynamics. This study compared adverse drug event (ADE) rates with cefepime prolonged (PI) and standard infusions (SI). METHODS This retrospective study included pediatric patients treated with cefepime for CF exacerbations between 2009 and 2019. One encounter per patient was analyzed with prioritization of SI encounters given sample size limitations. Baseline lab abnormalities, seizure disorders, and bleeding were exclusion criteria. The primary outcome was a composite safety endpoint (acute kidney injury [AKI], hepatotoxicity, hematologic toxicity, neurotoxicity, and hypersensitivity). RESULTS Of 188 patients, 135 received PI and 53 received SI. Baseline characteristics were similar between groups. More PI patients used CF transmembrane conductance regulator (CFTR) modulators (25% vs. 0%, p < 0.01) or had antibiotic allergies (62% vs. 38%, p = 0.02). Difference in rates of composite safety endpoint was not statistically significant between PI and SI (21 [15.6%] vs. 6 [11.3%] p = 0.46) nor was incidence of AKI (16 [11.8%] vs. 6 [11.3%], p = 0.92). Other ADEs were rarely observed. Length of stay (12.2 vs. 10.1 days, p = 0.06), change in discharge ppFEV1 from admission (13 vs. 12, p = 0.91) or from baseline (-4 vs. -6.5, p = 0.33), and time to next exacerbation (249.7 vs. 192.5 days, p = 0.93) were similar. CONCLUSIONS No difference in risk of ADEs including AKI was seen with cefepime PI in pediatric CF patients. Clinical outcomes were not significantly different between groups, but sample size may have limited comparison. PI cefepime may be considered in pediatric CF patients to optimize pharmacodynamics.
Collapse
Affiliation(s)
- Taylor A Imburgia
- Department of Pharmacy, West Virginia University Medicine Children's, Morgantown, West Virginia, USA
| | - Samantha R Engdahl
- Department of Pharmacy, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| | - Rebecca S Pettit
- Department of Pharmacy, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| |
Collapse
|
41
|
Imburgia TA, Kussin ML. A Review of Extended and Continuous Infusion Beta-Lactams in Pediatric Patients. J Pediatr Pharmacol Ther 2022; 27:214-227. [PMID: 35350159 DOI: 10.5863/1551-6776-27.3.214] [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: 05/13/2021] [Accepted: 10/29/2021] [Indexed: 11/11/2022]
Abstract
Intravenous beta-lactam antibiotics are the most prescribed antibiotic class in US hospitalized patients of all ages; therefore, optimizing their dosing is crucial. Bactericidal killing is best predicted by the time in which beta-lactam drug concentrations are maintained above the organism's minimum inhibitory concentration (MIC), rather than achievement of a high peak concentration. As such, administration of beta-lactam antibiotics via extended or continuous infusions over a minimum of 3 hours, rather than standard infusions over approximately 30 minutes, has been associated with improved achievement of pharmacodynamic targets and improved clinical outcomes in adult medical literature. This review summarizes the pediatric medical literature. Applicable studies include pharmacodynamic models, case series, retrospective analyses, and prospective studies on the use of extended infusion and continuous infusion penicillins, cephalosporins, carbapenems, and monobactams in neonates, infants, children, and adolescents. Specialized patient populations with unique pharmacokinetics and high-risk infections (neonates, critically ill, febrile neutropenia, cystic fibrosis) are also reviewed. While more studies are needed to confirm prospective clinical outcomes, the current body of evidence suggests extended and continuous infusions of beta-lactam antibiotics are well tolerated in children and improve achievement of pharmacokineticpharmacodynamic targets with similar or superior clinical outcomes, particularly in infections associated with high MICs.
Collapse
Affiliation(s)
- Taylor A Imburgia
- Department of Pharmacy (TAI), WVU Medicine Children's, Morgantown, WV
| | - Michelle L Kussin
- Department of Pharmacy (MLK), Riley Hospital for Children at Indiana University Health and Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
42
|
Xu C, Liu X, Cui Y, Huang X, Wang Y, Fan Y, Wu H, Li X, Guo B, Zhang J, Zhang Y. Case Report: Therapeutic Drug Monitoring of Polymyxin B During Continuous Renal Replacement Therapy in Two Pediatric Patients: Do Not Underestimate Extracorporeal Clearance. Front Pharmacol 2022; 13:822981. [PMID: 35401193 PMCID: PMC8988185 DOI: 10.3389/fphar.2022.822981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Polymyxin B has become the last choice for patient with carbapenem-resistant bacterial infection. However, the optimal dosing of polymyxin B in critically ill children receiving continuous renal replacement therapy (CRRT) remains unclear. Case Presentation: Two cases of critically ill pediatric patients (7 years old) with acute kidney injury requiring continuous renal replacement (CRRT) received polymyxin B treatment due to carbapenem-resistant organism bloodstream infections. Therapeutic drug monitoring (TDM) of polymyxin B was carried out by liquid chromatography tandem mass spectrometry (LC-MS/MS). The average steady-state plasma concentration (Css,avg) of 2–4 mg/L was set as the target level. Initial polymyxin B dose was 1 mg/kg every 12 h, and the Css,avg at 4–5th dosing were 1.76 and 1.06 mg/L for patient 1 and patient 2, respectively. TDM-guided polymyxin B dose was escalated to 2 mg/kg every 12 h for both patients, resulting in the Css,avg of 2.60 and 1.73 mg/L, and the infection was controlled subsequently. Css,avg of polymyxin B with the same dosing regimens and infusion length were different during CRRT and after termination of CRRT for both patients (2.60 mg/L vs. 4.94 mg/L with 2 mg/kg every 12 h in 2 h infusion for patient 1; and 1.73 mg/L vs. 3.53 mg/L with 2 mg/kg every 12 h in 2 h infusion for patient 2). The estimation of drug exposure (estimated by AUCss,12h at the same dose) during CRRT and cessation of CRRT showed that 45% and 51% of polymyxin B was cleared during CRRT. Conclusion: Our study showed high clearance of polymyxin B through CRRT, and supplanted dosing of polymyxin B is necessary in pediatric patients undergoing CRRT.
Collapse
Affiliation(s)
- Caifang Xu
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Yun Cui
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Yu Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Yaxin Fan
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Hailan Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Xin Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Beining Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- Phase I Clinical Trial Center, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yucai Zhang, ; Jing Zhang,
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yucai Zhang, ; Jing Zhang,
| |
Collapse
|
43
|
Cao G, Zhou P, Zhang H, Sun B, Tong X, Xing Y. Extended Infusion of Meropenem in Neonatal Sepsis: A Historical Cohort Study. Antibiotics (Basel) 2022; 11:341. [PMID: 35326804 PMCID: PMC8944670 DOI: 10.3390/antibiotics11030341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
This single-center historical cohort study investigated the effectiveness and safety of extended infusion (EI) compared with short-term infusion (STI) of meropenem in neonatal sepsis. Patient electronic health records from Peking University Third Hospital (1 December 2011−1 April 2021) were screened. Neonates diagnosed with sepsis and treated with meropenem in the neonatal intensive care unit were included (256 patients) as STI (0.5 h, 129 patients) and EI (2−3 h, 127 patients) groups. Three-day clinical effectiveness and three-day microbial clearance were considered the main outcomes. Univariate and multivariate analyses were performed. Baseline characteristics were similar in both groups. EI of meropenem was associated with a significantly higher 3-day clinical effectiveness rate (0.335 (0.180, 0.623), p = 0.001) and 3-day microbial clearance (4.127 (1.235, 13.784), p = 0.021) than STI, with comparable safety. Subgroup analyses showed that neonates with very low birth weight benefited from EI in terms of 3-day clinical effectiveness rate (75.6% versus 56.6%, p = 0.007), with no significant difference in the 3-day clinical effectiveness (85.1% versus 78.3%, p = 0.325) and microbial clearance (6% versus 5%, p > 0.999) rates between 3 h and 2 h infusions. Thus, EI of meropenem may be associated with better effectiveness and comparable safety in treating neonatal sepsis than STI. Nonetheless, historically analyzed safety evaluation might be biased, and these findings need confirmation in randomized controlled trials of larger sample sizes.
Collapse
Affiliation(s)
- Guangna Cao
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China;
| | - Pengxiang Zhou
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China;
- Peking University Health Science Center, Institute for Drug Evaluation, Beijing 100191, China
| | - Hua Zhang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China;
| | - Bangkai Sun
- Information Management and Big Data Center, Peking University Third Hospital, Beijing 100191, China;
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China;
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China;
| |
Collapse
|
44
|
Lu Q, He X, Fang J, Shi K, Hu F, Bian X, Wang X. Simultaneous determination of linezolid and voriconazole serum concentrations using liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2022; 212:114659. [PMID: 35182831 DOI: 10.1016/j.jpba.2022.114659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/25/2022]
Abstract
Linezolid and voriconazole are two antimicrobials used for severe infections in critically ill patients. Pharmacokinetics and pharmacodynamics are altered in critically ill patients. Therefore, standard dosing of anti-infective agents may not reach the optimal therapeutic targets. A rapid and simple liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for simultaneous determination of linezolid and voriconazole in human serum only 3 min after one-step protein precipitation pretreatment to monitor their concentrations. Multiple-reaction monitoring (MRM) mode was used for detection. The calibration curves were linear over the range of 0.5-100 μg/mL for both linezolid and voriconazole, with regression coefficients above 0.9900 for all analytes. The intra- and interday coefficients of variation were below 15% at all concentration levels (LLOQ/LQC/MQC/HQC). This method was successfully applied to routine therapeutic drug monitoring (TDM) for critically ill patients and other patients in need.
Collapse
Affiliation(s)
- Qiuya Lu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xiaoshuang He
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jie Fang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Kaifeng Shi
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Fengmei Hu
- Shanghai AB Sciex Analytical Instrument Trading Co., Ltd., Shanghai 200050, China
| | - Xiaolan Bian
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
45
|
Kato H, Hamada Y. Amikacin Therapy in Japanese Pediatric Patients: Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19041972. [PMID: 35206156 PMCID: PMC8871704 DOI: 10.3390/ijerph19041972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022]
Abstract
Children show a very wide range of physical development processes. These changes impact pharmacokinetic (PK) variability in pediatric patients. Most PK studies have been conducted on the Caucasian population. Therefore, whether current evidence of how developmental change affects PK and exposure-response relationships applies to Japanese pediatric patients remains unclear. This narrative review focuses on amikacin therapy in Japanese pediatric patients and shows the relationship between amikacin concentrations and efficacy/toxicity. Ten relevant articles were identified. Of these, nine articles were published in the 1980s. All studies reported a maximum concentration (Cmax) and minimum concentration (Cmin) of amikacin. Overall, articles reporting PK/pharmacodynamic (PD) indices and minimum inhibitory concentration (MIC) of isolated bacteria in Japanese pediatric patients is lacking, whereas all patients recovered from an infection state and showed negative cultures. Five of the included studies reported the association between Cmin and toxicity. The Cmin in three of four patients who developed toxicity was above 10 mg/L. This narrative review shows that further PK study of amikacin in Japanese pediatric patients is necessary. In particular, the pursuit of knowledge of Cmax/MIC ratio is vital. On the other hand, this review demonstrates that the optimal Cmin for Japanese pediatric patients is below 10 mg/L as a candidate concentration. However, it is noted that the number of patients who developed toxicity is very small.
Collapse
Affiliation(s)
- Hideo Kato
- Department of Pharmacy, Mie University Hospital, Tsu 514-8507, Japan
- Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
- Correspondence: or ; Tel.: +81-059-232-1111
| | - Yukihiro Hamada
- Department of Pharmacy, Tokyo Women’s Medical University Hospital, Tokyo 162-8666, Japan;
| |
Collapse
|
46
|
Hartman SJF, Upadhyay PJ, Mathôt RAA, van der Flier M, Schreuder MF, Brüggemann RJ, Knibbe CA, de Wildt SN. OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1725-1732. [PMID: 35383374 PMCID: PMC9155601 DOI: 10.1093/jac/dkac095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Stan J. F. Hartman
- Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Parth J. Upadhyay
- Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ron A. A. Mathôt
- Department of Clinical Pharmacology and Hospital Pharmacy - Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michiel van der Flier
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children's Hospital, and Section Pediatric Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Michiel F. Schreuder
- Department of Pediatrics, Division of Pediatric Nephrology, Amalia Children’s Hospital, Radboud Institute of Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | | | - Catherijne A. Knibbe
- Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Saskia N. de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboudumc, Nijmegen, The Netherlands
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Intensive Care Medicine, Radboud Institute of Health Sciences, Radboudumc, Nijmegen, The Netherlands
- Corresponding author. E-mail:
| |
Collapse
|
47
|
Schouwenburg S, Wildschut ED, de Hoog M, Koch BCP, Abdulla A. The Pharmacokinetics of Beta-Lactam Antibiotics Using Scavenged Samples in Pediatric Intensive Care Patients: The EXPAT Kids Study Protocol. Front Pharmacol 2021; 12:750080. [PMID: 34955824 PMCID: PMC8703159 DOI: 10.3389/fphar.2021.750080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022] Open
Abstract
Background: Emerging evidence supports the importance of optimized antibiotic exposure in pediatric intensive care unit (PICU) patients. Traditional antibiotic dosing is not designed for PICU patients, as the extreme pharmacokinetic (PK) behavior of drugs threatens the achievement of optimal antibiotic treatment outcomes. Scavenged sampling is a sampling strategy which may have positive implications for routine TDM and PK research, as well as monitoring other biomarkers. EXPAT Kids study was designed to analyze whether current empiric dosing regimens of frequently used beta-lactam antibiotics achieve defined therapeutic target concentrations in PICU patients. Methods: A mono-centre, exploratory pharmacokinetic and pharmacodynamic study was designed to assess target attainment of beta-lactam antibiotics. One hundred forty patients will be included within 24 months after start of inclusion. At various time points serum concentration of the study antibiotic (cefotaxime, ceftazidime, ceftriaxone, cefuroxime, flucloxacillin, and meropenem) are determined. In parallel with these sampling moments, residual material is collected to validate the use of blood of scavenged heparinized astrup syringes for the quantification of antibiotic exposure. The primary outcome is the time that the free (unbound) concentration of the study antibiotic remains above one to four the minimal inhibitory concentration during a dosing interval (100%ƒT > MIC and 100%ƒT>4xMIC). Other included outcomes are disease severity, safety, length of stay, and inflammatory biomarkers. Discussion: Potentially, scavenged sampling may enrich the EXPAT Kids dataset, and reduce additional blood sampling and workload for clinical personnel. The findings from the EXPAT Kids study will lead to new insights in the PK parameters of beta-lactams and consecutive effects on target attainment and clinical outcomes. Is there a need for more precision in dosing? Netherlands Trial Register Number: Trial NL9326.
Collapse
Affiliation(s)
- Stef Schouwenburg
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Enno D Wildschut
- Department of Pediatric Intensive Care, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M de Hoog
- Department of Pediatric Intensive Care, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Alan Abdulla
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Pediatric Intensive Care, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
48
|
Chen J, Huang X, Bu S, Chen X, Zhou J, Liu X, Guo X, Li L, Zhang J. The relationship between vancomycin AUC/MIC and trough concentration, age, dose, renal function in Chinese critically ill pediatric patients. Pharmacol Res Perspect 2021; 9:e00885. [PMID: 34664790 PMCID: PMC8525138 DOI: 10.1002/prp2.885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/02/2021] [Indexed: 12/16/2022] Open
Abstract
To assess the pharmacokinetic parameters of vancomycin in Chinese critically ill pediatric patients, children treated with vancomycin, hospitalized in the intensive care unit were included. Samples to determine peak and trough serum concentrations were obtained on the third day of treatment. Half-life was significantly longer in neonates and showed a decreasing trend in infants and children. In patients aged ≥1 month, AUC24 /MIC ≥400 was achieved in 31.8% at the dose of 40 mg/kg/d, and in 48.7% at the dose of 60 mg/kg/d with an assumed MIC of 1 mg/L. Augmented renal clearance (ARC) was present in 27.3% of children, which was associated with higher vancomycin clearance and lower AUC values. A good correlation was observed between trough concentration and AUC24 , and the trough concentration that correlated with AUC24 of 400 were varied according to the dosage regimens, 8.42 mg/L for 6-hintervals, and 6.63 mg/L for 8-h intervals. To conclude, vancomycin trough concentration that related to the AUC24 of 400 was much lower in critically ill children than that in adults. The dosage of 60 mg/kg/day did not enough for producing AUC24 in the range of 400-600 mg h/L in critically ill children, especially in those with ARC.
Collapse
Affiliation(s)
- Jihui Chen
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaohui Huang
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Shuhong Bu
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoxiao Chen
- Department of Hematology/OncologyShanghai Children’s Medical CenterSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jia Zhou
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xinzhu Liu
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaowen Guo
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Lixia Li
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jian Zhang
- Department of PharmacyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
49
|
Thomson KM, Dyer C, Liu F, Sands K, Portal E, Carvalho MJ, Barrell M, Boostrom I, Dunachie S, Farzana R, Ferreira A, Frayne F, Hassan B, Jones E, Jones L, Mathias J, Milton R, Rees J, Chan GJ, Bekele D, Mahlet A, Basu S, Nandy RK, Saha B, Iregbu K, Modibbo F, Uwaezuoke S, Zahra R, Shirazi H, Syed NU, Mazarati JB, Rucogoza A, Gaju L, Mehtar S, Bulabula ANH, Whitelaw A, van Hasselt JGC, Walsh TR, BARNARDS Group SahaSamirIslamMaksudaBin-AhmedZabedAhmedWazirBegumTaslimaChowdhuryMituSharminShailaRani DeyChumkiUttamMatinAbdulChakrabortySowmitra RanjanTasminSadiaRemaDipaKhatunRashidaNathLizaBalkachewNigatuBekeleDelayehuSchaughencyKatherineSolomonSemariaGebreyohanesZenebeAmbachewRozinaOdumadeOludareHaileselassieMisganaChanGraceRussoAbigailWorknehRedeatMetaferiaGesitAbaynehMahletMohammedYahya ZekariaBiteyeTeferaTekluAlulaGezahegnWendimagegnChakravortyPartha SarathiMukherjeeAnuradhaNandyRanjan KumarRoySamarpanSinhaAnuradhaNahaSharmiMalakarSukla SahaBoseSiddharthaMajhiMonakiSahooSubhasreeMukherjeePutulRoutaSumitra KumariNandiChaitaliBasuSulagnaSahaBijanChattopadhyayPinakiModibboFatima Zara IsaUwaezuokeStellaMeduekweDilichukwuMuhammadKhairiyyaNsudeQueenUkehIfeomaOkenuMary-JoeChinenyeAkpuluYakubuSamuelAsunugwoVivianAinaFolakeIssyIsibongAdekeyeDolapoEuniceAdieleAminaAbdulmlikOyewoleROlotonINnajiBCUmejiegoMAnokePNAdebayoSAbegunrinGOOmotoshoOBIbrahimRIgweBAbrokoMBalamiKBayemLAnyanwuCHarunaHOkikeJGorohKBoi-SundayMUgaforAugustaMakamaMaryamNdukweKanibaOdamaAnastesiaYusufHadizaWachukwuPatienceYahayaKachallaKalade ColsonsTitusKuraMercyOrebiyiDamilolaIregbuKenneth C.MmaduekeChukwuemekaAuduLamidiIdrisNuraGamboSafiyaIbrahimJamilaPreciousEdwinHassanAshiruGwadabeShamsuddenAdeleye FalolaAdeolaAliyuMuhammadIbrahimAminaMukaddasAisha SaniKhalidRashida YakubuAlkaliFatima IbrahimMuhammadMaryam YahayaTukurFatima MohammadMuhammadSurayya MustaphaShittuAdeolaBelloMurjanatuHassanMuhammad AbubakarSa adFatima HabibKassimAishatuShiraziHaiderMuhammadAdilZahraRabaabUllahSyed NajeebHilal JanMuhammadKamranRubinaSajanaSaeedJazbaMaqsoodNoreenZafarMariaSadiqSaraeenAhsanSumbleTariqMadihaSajidSidraMustafaHasmaRehmanAnees-urMuhammadAtifMehmoodGahssanNisarMahnoorAkifShermeenYasmeenTahiraNawazSabirAttaAnam ShanalLaiq-ur-RehmanMianKousarRobinaBibiKalsoomWaheedKosarMajeedZainabJalilAyeshaKajibwamiEspoirRucogozaAnicethNzabahimanaInnocentJean-BaptisteMazaratiGajuLucieRizikiKankundiyeUwamahoroBrigetteUweraRachelNyiratuzaEugenieMuzunguKumwamiUwitonzeVioletteHoranimpunduMarie CNzeyimanaFrancineMitimaPrinceDramowskiAngelaWhitelawAndrewPatersonLaurenFransMaryJohnsonMarvinaSwanepoelEvelineBojanaZolekadu PreezMiemeMehtarShaheenBulabulaAndreLiuFeiyanvan HasseltJohan GCWalshTimothySandsKirstyCarvalhoMariaMiltonRebeccaThomsonKathrynPortalEdwardMathiasJordanDyerCalieFerreiraAnaAndrewsRobertWatkinsJohnGillespieDavidHoodKerryTaiyaiKatieKirbyNigelNietoMariaHenderThomasHoganPatrickSaifHabibaHassanBrekhnaJonesEllisBarrellMatthewBoostromIanFrayneFrancisReesJessicaJonesLimDunachieSusannaSpillerBradParkhillJulian. Effects of antibiotic resistance, drug target attainment, bacterial pathogenicity and virulence, and antibiotic access and affordability on outcomes in neonatal sepsis: an international microbiology and drug evaluation prospective substudy (BARNARDS). THE LANCET. INFECTIOUS DISEASES 2021; 21:1677-1688. [PMID: 34384533 PMCID: PMC8612937 DOI: 10.1016/s1473-3099(21)00050-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 12/01/2020] [Accepted: 01/22/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Sepsis is a major contributor to neonatal mortality, particularly in low-income and middle-income countries (LMICs). WHO advocates ampicillin-gentamicin as first-line therapy for the management of neonatal sepsis. In the BARNARDS observational cohort study of neonatal sepsis and antimicrobial resistance in LMICs, common sepsis pathogens were characterised via whole genome sequencing (WGS) and antimicrobial resistance profiles. In this substudy of BARNARDS, we aimed to assess the use and efficacy of empirical antibiotic therapies commonly used in LMICs for neonatal sepsis. METHODS In BARNARDS, consenting mother-neonates aged 0-60 days dyads were enrolled on delivery or neonatal presentation with suspected sepsis at 12 BARNARDS clinical sites in Bangladesh, Ethiopia, India, Pakistan, Nigeria, Rwanda, and South Africa. Stillborn babies were excluded from the study. Blood samples were collected from neonates presenting with clinical signs of sepsis, and WGS and minimum inhibitory concentrations for antibiotic treatment were determined for bacterial isolates from culture-confirmed sepsis. Neonatal outcome data were collected following enrolment until 60 days of life. Antibiotic usage and neonatal outcome data were assessed. Survival analyses were adjusted to take into account potential clinical confounding variables related to the birth and pathogen. Additionally, resistance profiles, pharmacokinetic-pharmacodynamic probability of target attainment, and frequency of resistance (ie, resistance defined by in-vitro growth of isolates when challenged by antibiotics) were assessed. Questionnaires on health structures and antibiotic costs evaluated accessibility and affordability. FINDINGS Between Nov 12, 2015, and Feb 1, 2018, 36 285 neonates were enrolled into the main BARNARDS study, of whom 9874 had clinically diagnosed sepsis and 5749 had available antibiotic data. The four most commonly prescribed antibiotic combinations given to 4451 neonates (77·42%) of 5749 were ampicillin-gentamicin, ceftazidime-amikacin, piperacillin-tazobactam-amikacin, and amoxicillin clavulanate-amikacin. This dataset assessed 476 prescriptions for 442 neonates treated with one of these antibiotic combinations with WGS data (all BARNARDS countries were represented in this subset except India). Multiple pathogens were isolated, totalling 457 isolates. Reported mortality was lower for neonates treated with ceftazidime-amikacin than for neonates treated with ampicillin-gentamicin (hazard ratio [adjusted for clinical variables considered potential confounders to outcomes] 0·32, 95% CI 0·14-0·72; p=0·0060). Of 390 Gram-negative isolates, 379 (97·2%) were resistant to ampicillin and 274 (70·3%) were resistant to gentamicin. Susceptibility of Gram-negative isolates to at least one antibiotic in a treatment combination was noted in 111 (28·5%) to ampicillin-gentamicin; 286 (73·3%) to amoxicillin clavulanate-amikacin; 301 (77·2%) to ceftazidime-amikacin; and 312 (80·0%) to piperacillin-tazobactam-amikacin. A probability of target attainment of 80% or more was noted in 26 neonates (33·7% [SD 0·59]) of 78 with ampicillin-gentamicin; 15 (68·0% [3·84]) of 27 with amoxicillin clavulanate-amikacin; 93 (92·7% [0·24]) of 109 with ceftazidime-amikacin; and 70 (85·3% [0·47]) of 76 with piperacillin-tazobactam-amikacin. However, antibiotic and country effects could not be distinguished. Frequency of resistance was recorded most frequently with fosfomycin (in 78 isolates [68·4%] of 114), followed by colistin (55 isolates [57·3%] of 96), and gentamicin (62 isolates [53·0%] of 117). Sites in six of the seven countries (excluding South Africa) stated that the cost of antibiotics would influence treatment of neonatal sepsis. INTERPRETATION Our data raise questions about the empirical use of combined ampicillin-gentamicin for neonatal sepsis in LMICs because of its high resistance and high rates of frequency of resistance and low probability of target attainment. Accessibility and affordability need to be considered when advocating antibiotic treatments with variance in economic health structures across LMICs. FUNDING The Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Kathryn M Thomson
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, UK,Correspondence to: Kathryn M Thomson, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK and Ineos Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Calie Dyer
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Feiyan Liu
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Kirsty Sands
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, UK
| | - Edward Portal
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Maria J Carvalho
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Matthew Barrell
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Ian Boostrom
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Susanna Dunachie
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK,Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Refath Farzana
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, UK
| | - Ana Ferreira
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Francis Frayne
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Brekhna Hassan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Ellis Jones
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Lim Jones
- Public Health Wales Microbiology, University Hospital of Wales, Cardiff, UK
| | - Jordan Mathias
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Rebecca Milton
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Jessica Rees
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Grace J Chan
- Division of Medicine Critical Care, Boston Children's Hospital, Boston, MA, USA,Department of Epidemiology, Harvard T H Chan School of Public Health, Boston, MA, USA,Department of Paediatrics, St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Delayehu Bekele
- Department of Obstetrics and Gynecology, St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Abayneh Mahlet
- Department of Paediatrics, St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Sulagna Basu
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases Beliaghata, Kolkata, India
| | - Ranjan K Nandy
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases Beliaghata, Kolkata, India
| | - Bijan Saha
- Department of Neonatology, Institute of Postgraduate Medical Education & Research, Kolkata, India
| | | | | | | | | | - Haider Shirazi
- Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | | | | | - Aniceth Rucogoza
- University Teaching Hospital, Kigali, Rwanda,National Reference Laboratory, Rwanda Biomedical Center, Kigali, Rwanda
| | - Lucie Gaju
- University Teaching Hospital, Kigali, Rwanda
| | - Shaheen Mehtar
- Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | - Andre N H Bulabula
- Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | - Andrew Whitelaw
- Division of Medical Microbiology, Stellenbosch University, Cape Town, South Africa,National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| | | | - Timothy R Walsh
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,Ineos Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, UK,Prof Timothy R Walsh, Ineos Oxford Institute of Antimicrobial Research, Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | | |
Collapse
|
50
|
Akunne OO, Mugabo P, Argent AC. Pharmacokinetics of Vancomycin in Critically Ill Children: A Systematic Review. Eur J Drug Metab Pharmacokinet 2021; 47:31-48. [PMID: 34750740 PMCID: PMC8574943 DOI: 10.1007/s13318-021-00730-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2021] [Indexed: 11/26/2022]
Abstract
Background and Objective Vancomycin is often used in the ICU for the treatment of Gram-positive bacterial infection. In critically ill children, there are pathophysiologic changes that affect the pharmacokinetics of vancomycin. A systematic review of vancomycin pharmacokinetics and pharmacodynamics in critically ill children was performed. Methods Pharmacokinetic studies of vancomycin in critically ill children published up to May 2021 were included in the review provided they included children aged > 1 month. Studies including neonates were excluded. A search was performed using the PubMed, Scopus, and Google Scholar databases. The Risk of Bias Assessment Tool for Systematic Reviews (ROBIS) was used to check for quality and reduce bias. Data on study characteristics, patient demographics, clinical parameters, pharmacokinetic parameters, outcomes, and study limitations were collected. Results Thirteen studies were included in this review. A wide variety of dosing and sampling strategies were used in the studies. Methods for estimating vancomycin pharmacokinetics, especially the area under the curve over 24 h, varied. Vancomycin doses of 20–60 mg/kg were given daily. This resulted in high variability in pharmacokinetic parameters. Vancomycin trough level was less than 15 μg/mL in most of the studies. Vancomycin clearance ranged from 0.05 to 0.38 L/h/kg. Volume of distribution ranged from 0.1 to 1.16 L/kg. Half-life was between 2.4 and 23.6 h. Patients in the study receiving continuous vancomycin infusion had AUC24 < 400 µg·h/mL. Conclusion There is large variability in the pharmacokinetics of vancomycin among critically ill patients. Studies to assess the factors responsible for this variability in vancomycin pharmacokinetics are needed.
Collapse
Affiliation(s)
- Onyinye Onyeka Akunne
- Discipline of Pharmacology, School of Pharmacy, University of the Western Cape, Bellville, Cape Town, 7535 South Africa
| | - Pierre Mugabo
- Discipline of Pharmacology, School of Pharmacy, University of the Western Cape, Bellville, Cape Town, 7535 South Africa
| | - Andrew C Argent
- Paediatrics and Child Health, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
- Paediatric Intensive Care Unit, Red Cross War Memorial Children Hospital, Rondebosch, Cape Town, 7700 South Africa
| |
Collapse
|