1
|
Itohara K, Yamamoto K, Fujinaka S, Hashimoto M, Tamura N, Kitahiro Y, Omura T, Fujioka K, Yano I. Pharmacokinetic and Pharmacodynamic Assessment of Valganciclovir in Infants With Congenital Cytomegalovirus Infection. Ther Drug Monit 2025; 47:393-399. [PMID: 40344019 DOI: 10.1097/ftd.0000000000001257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Valganciclovir (VGCV) is administered at a dose of 16 mg/kg 2 times daily for 6 months to treat symptomatic congenital cytomegalovirus (CMV) infections. During the treatment period, approximately 20% of the patients developed grade 3 or higher neutropenia. Currently, information on the pharmacokinetics and pharmacodynamics of ganciclovir, an active metabolite of VGCV, in infants is limited. In the current study, the relationship between ganciclovir concentration and neutropenia was investigated, and a population pharmacokinetic (PPK) model of ganciclovir in infants with symptomatic congenital CMV infection was developed. METHODS Japanese infants who were prescribed oral VGCV for symptomatic congenital CMV infections between July 2017 and January 2021 were included. The relationship between the observed trough ganciclovir concentrations and neutrophil counts was examined. PPK analysis was performed to evaluate the covariates affecting the pharmacokinetics of ganciclovir. RESULTS Twenty-seven ganciclovir serum samples from 8 patients were analyzed. A moderate negative correlation was observed between the observed trough ganciclovir concentration and neutrophil count. PPK model analysis showed that postmenstrual age (PMA) affected the total body clearance of ganciclovir after correcting for the empirical allometric scaling of body weight. Based on PMA and body weight, a nomogram to achieve the target area under the concentration-time curve from 0 to 24 hours of 40-60 mcg·h·mL -1 of ganciclovir was calculated. CONCLUSIONS The relationship between neutrophil count and ganciclovir trough concentration in infants was clarified. The PPK model showed that the dose of VGCV should be reduced in patients with a low PMA to achieve target exposure.
Collapse
Affiliation(s)
| | | | | | | | - Naoki Tamura
- Department of Pharmacy, Kobe University Hospital
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University; and
| | | | | | - Kazumichi Fujioka
- Department of Pediatrics, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Ikuko Yano
- Department of Pharmacy, Kobe University Hospital
| |
Collapse
|
2
|
Burgmaier K, Kilian S, Arbeiter K, Atmis B, Boyer O, Buescher A, Dursun I, Erger F, Fila M, Galiano M, Gokce I, Haeffner K, Haffner D, Hooman N, Klaus G, König J, Lange-Sperandio B, Marlais M, Massella L, Mekahli D, Miklaszewska M, Miloševski-Lomić G, Obrycki L, Ranchin B, Seitz B, Stabouli S, Tabel Y, Taranta-Janusz K, Weber LT, Weitz M, Wühl E, Yilmaz A, Dötsch J, Schaefer F, Liebau MC. A risk score to predict kidney survival in patients with autosomal recessive polycystic kidney disease at the age of two months. Kidney Int 2025; 107:903-915. [PMID: 39922379 DOI: 10.1016/j.kint.2025.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 02/10/2025]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is a severe hepatorenal fibrocystic disorder. Its rareness and the variability of disease courses have been major obstacles for the establishment of clinical trials on treatment of kidney disease in ARPKD. In this observational study we characterized kidney disease progression in a very large cohort of up to 658 patients with the clinical diagnosis of ARPKD and identified risk factors associated with rapid kidney disease progression. The estimated probability of kidney failure by the age of 20 years was 50.1% (95% confidence interval 42.2%‒57.0%), with earlier kidney failure in specific subgroups. Mean yearly estimated glomerular filtration rate decline after the first year of life was 1.3 ml/min per 1.73 m2 during childhood and adolescence in the overall cohort, ranging from 0.5 to 2.2 ml/min per 1.73 m2 in various subgroups. Furthermore, we developed prediction models for the relative risk of early kidney failure to be applied at the age of two months in daily clinical life. The finally chosen predictor set for a score based on a Cox model encompassed five factors: gestational age at oligo- or anhydramnios, gestational age at birth, functional genotype, serum creatinine (mg/dl) as well as documentation of arterial hypertension at the age of two months. The derived simple prognostic score showed good prediction performance, especially in the first three years of life. It reliably identified patients who are not at risk of early kidney failure and may be helpful to identify patients at risk of more rapid disease progression that could benefit from novel therapeutic interventions.
Collapse
Affiliation(s)
- Kathrin Burgmaier
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany; Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Samuel Kilian
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Klaus Arbeiter
- Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Bahriye Atmis
- Department of Pediatric Nephrology, Cukurova University Faculty of Medicine, Adana, Turkiye
| | - Olivia Boyer
- Pediatric Nephrology and Kidney Transplantation, Hôpital Necker Enfants Malades, MARHEA, Institut Imagine, Université Paris Cité, Paris, France
| | - Anja Buescher
- Department of Pediatrics II, University Hospital Essen, Essen, Germany
| | - Ismail Dursun
- Department of Pediatric Nephrology, Erciyes University, Faculty of Medicine, Kayseri, Turkiye
| | - Florian Erger
- Institute of Human Genetics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Marc Fila
- Pediatric Nephrology Unit, Centre Hospitalier Universitaire Arnaud de Villeneuve-Université de Montpellier, Montpellier, France
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ibrahim Gokce
- Division of Pediatric Nephrology, Research and Training Hospital, Marmara University, Istanbul, Turkiye
| | - Karsten Haeffner
- Department of Internal Medicine IV, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Nakysa Hooman
- Department of Pediatric Nephrology, Ali-Asghar Children Hospital, Ali-Asghar Clinical Research Development Center (AACRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Guenter Klaus
- KfH Center of Paediatric Nephrology and Department of Pediatric Nephrology, Marburg Kidney Research Center, Philipps University, Marburg, Germany
| | - Jens König
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Bärbel Lange-Sperandio
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universitaet, Munich, Germany
| | - Matko Marlais
- UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Laura Massella
- Division of Nephrology, Bambino Gesú Children's Hospital-IRCCS, Rome, Italy
| | - Djalila Mekahli
- PKD Research Group, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | | | - Lukasz Obrycki
- Department of Nephrology, Kidney Transplantation and Hypertension, the Children's Memorial Health Institute, Warsaw, Poland
| | - Bruno Ranchin
- Pediatric Nephrology Unit, Centre de référence maladies rénales rares, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Barbara Seitz
- KfH Center of Pediatric Nephrology, Children's Hospital Munich Schwabing, Munich, Germany
| | - Stella Stabouli
- First Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokratio Hospital, Thessaloniki, Greece
| | - Yilmaz Tabel
- Department of Pediatric Nephrology, Faculty of Medicine, İnönü University, Malatya, Turkey
| | | | - Lutz Thorsten Weber
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Marcus Weitz
- Department of General Pediatrics and Haematology/Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Elke Wühl
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Alev Yilmaz
- Pediatric Nephrology Department, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkiye
| | - Jörg Dötsch
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Family Health, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Max Christoph Liebau
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University Hospital Cologne and University of Cologne, Cologne, Germany; Center for Family Health, University Hospital Cologne and University of Cologne, Cologne, Germany; West German Center for Child and Adolescent Health (WZKJ), Partner site Cologne, Department of Pediatrics, Cologne, Germany.
| |
Collapse
|
3
|
Yaliniz A, El Hassani M, Blanchard AC, Marsot A. Anti-infectives in Pediatric Patients with Cystic Fibrosis: A Comprehensive Review of Population Pharmacokinetic Analyses. Clin Pharmacokinet 2025; 64:631-653. [PMID: 40254714 DOI: 10.1007/s40262-025-01505-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Pulmonary complications are the leading cause of morbidity and mortality in pediatric patients with cystic fibrosis. Altered pharmacokinetic parameters in this population, as well as high inter- and intra-individual variability, complicate the optimization of anti-infective treatments. In this review, we aim to summarize and describe all anti-infective population pharmacokinetic (popPK) models applied to pediatric populations with cystic fibrosis. Our objectives were to identify the most-reported structural models and retained covariates and to compare the dosing regimens used in clinical routine with those recommended in literature and guidelines. A literature search was done through the PubMed database from inception to August 2024. Studies were retained only if they complied with the inclusion and exclusion criteria. The review included 21 popPK models covering the pharmacokinetic profiles of eight different molecules. Among these, five are recommended antibiotics for treating pulmonary infections in patients with cystic fibrosis. All models incorporated body composition and/or renal function measures as covariates in their pharmacokinetic parameter equations. Standard dosing regimens in the studies were consistent with guidelines and literature recommendations. This is the first review summarizing and describing all anti-infective popPK models in pediatric patients with cystic fibrosis. Improved estimation of pharmacokinetic parameters and a clearer understanding of variability sources will enhance the optimization of antibiotic treatment in clinical practice. Finally, the impact of new targeted therapies on the management of this population will have to be closely monitored in the years ahead.
Collapse
Affiliation(s)
- Aysenur Yaliniz
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculty of Pharmacy, Université de Montréal, 2940 chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada.
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.
| | - Mehdi El Hassani
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculty of Pharmacy, Université de Montréal, 2940 chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - Ana C Blanchard
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Amélie Marsot
- Laboratoire de Suivi Thérapeutique Pharmacologique et Pharmacocinétique, Faculty of Pharmacy, Université de Montréal, 2940 chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| |
Collapse
|
4
|
Gotta V, Csajka C, Glauser A, Berger C, Pfister M, Paioni P. Risk of Potentially Neurotoxic Exposure in Infants Under High-Dose Cefepime Treatment-A Pharmacometric Simulation Study. Pharmaceutics 2025; 17:544. [PMID: 40430837 PMCID: PMC12114997 DOI: 10.3390/pharmaceutics17050544] [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: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Optimal dosing of cefepime in infants 1-2 months remains undefined. Objectives: We aimed to quantify the risk of potentially neurotoxic exposure with high-dose cefepime (50 mg/kg/8 h) in infants 1-2 months of age, as compared to adjacent age groups (neonates, infants 2-12 months) and lower dose treatment (50 mg/kg/12 h). Methods: Pharmacometric simulations were performed using two published population pharmacokinetic models combined with demographic data, including serum creatinine, for neonates and infants ≤ 12 months. Adult-derived safety thresholds for potential neurotoxicity were defined as steady-state trough concentration (Ctrough) > 20 or > 35 mg/L, respectively. The corresponding probability of target attainment (PTA) was calculated as free concentration, 50% of the time during the dosing interval above the minimal inhibitory concentration (MIC) breakpoint of 8 mg/L (Pseudomonas spp.) (50% fT>MIC8mg/L). Results: The predicted risk of Ctrough > 20 (>35) mg/L under high-dose cefepime was 40-54% (12-22%) in infants 1-2 months while providing high PTA (100%). It was predicted to be 1.3-1.7 fold higher in neonates (model 1), and reduced 1.8-2.4 fold in infants 2-12 months (model 1), or to be similar (model 2), respectively. Both models predicted approximately 2-4 fold reduced risk using lower dose treatments while maintaining high PTA (≥97%). Conclusions: The risk of potential neurotoxic concentrations in infants > 1 month treated with cefepime 50 mg/kg/8 h is high if defined by adult safety thresholds. Lower dose cefepime in infants 1-2 months could be a safe option without compromising PTA, if defined as 50% fT>MIC8mg/L. Achievement of 100% fT>MIC8mg/L may require prolonged infusion time even under high-dose treatment. Future research is required to evaluate potentially age-dependent safety thresholds.
Collapse
Affiliation(s)
- Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children’s Hospital, 4031 Basel, Switzerland
- Pediatric Clinical Pharmacy, University of Basel Children’s Hospital, 4031 Basel, Switzerland
| | - Chantal Csajka
- Center for Research and Innovation, University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | | | - Christoph Berger
- SwissPedDose, 8008 Zurich, Switzerland
- Division of Infectious Diseases, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children’s Hospital, 4031 Basel, Switzerland
| | - Paolo Paioni
- Division of Infectious Diseases, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| |
Collapse
|
5
|
Matcha S, Dilli Batcha JS, Raju AP, Chaudhari BB, Moorkoth S, Mallayasamy S, Lewis LE. Precision dosing of amikacin in term neonates using pharmacometric approach. Pediatr Res 2025:10.1038/s41390-025-04044-7. [PMID: 40210955 DOI: 10.1038/s41390-025-04044-7] [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] [Received: 08/08/2024] [Revised: 01/16/2025] [Accepted: 02/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Maintaining amikacin concentrations within a specific therapeutic window is crucial to avoid sub-therapeutic or toxic levels. This study aimed to design a dosing nomogram for amikacin in neonates using a Population Pharmacokinetic (PopPK) modeling approach. METHODS PopPK model was developed using 101 amikacin concentrations from 80 neonates and validated using model diagnostics, and empirical Bayesian forecasting was performed. Pharmacokinetic profiles were simulated for virtual subjects with a range of covariates to identify suitable dosage regimens. Dosage regimens with the highest probability for the target group were selected to design the dosing nomogram. RESULTS A two-compartment PK model best described the study data. Body weight (WT), serum creatinine (SCR), and post-natal age (PNA) affected the clearance of amikacin. The model predictions are with less than 15% absolute prediction error. WT and SCR were divided into five groups each, with each group repeated for every week of PNA for four weeks for dosing nomogram development. CONCLUSION A PopPK model was developed and successfully-predicted concentrations in the study population. This model was used to develop a nomogram considering significant covariates like WT, SCR, and PNA. The proposed dosing nomogram can assist clinicians in developing individualized dosage regimens. IMPACT Population pharmacokinetic (PopPK) models for amikacin in term neonates were developed using clinical data from an Indian clinical setting and successfully-predicted the amikacin concentrations for the study population. Pharmacokinetic simulations with virtual subjects were used to calculate the probability of target attainment for different dosing regimens. The proposed dosing nomogram can potentially assist clinicians in designing optimal amikacin dosage regimens for neonates.
Collapse
Affiliation(s)
- Saikumar Matcha
- Research Scholar, Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
- Titus Family Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, USA
| | - Jaya Shree Dilli Batcha
- Research Scholar, Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
- Center for Pharmacometrics, Manipal Academy of Higher Education, Manipal, India
| | - Arun Prasath Raju
- Research Scholar, Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
- Center for Pharmacometrics, Manipal Academy of Higher Education, Manipal, India
| | - Bhim Bahadur Chaudhari
- Research Scholar, Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sudheer Moorkoth
- Professor & Head, Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Surulivelrajan Mallayasamy
- Center for Pharmacometrics, Manipal Academy of Higher Education, Manipal, India.
- Professor & Head, Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India.
| | - Leslie E Lewis
- Professor & Head, Department of Pediatrics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
6
|
Xu L, Xu J, Mao H, Qian W, Ma Z, Zhang Y, Zhu Y, Zhu X, Xu Y. Prevalence and Associated Explanatory Factors for Augmented Renal Clearance in Early Sepsis: Single-Center, Retrospective PICU Cohort in China, 2022-2023. Pediatr Crit Care Med 2025:00130478-990000000-00463. [PMID: 40111116 DOI: 10.1097/pcc.0000000000003727] [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: 03/22/2025]
Abstract
OBJECTIVES We aimed to 1) evaluate the prevalence of augmented renal clearance (ARC) in pediatric sepsis patients; 2) analyze potential explanatory factors associated with ARC out of hemodynamic, oxygenation, and inflammatory parameters; and 3) assess ARC outcomes. DESIGN Retrospective, single-center, cohort from January 2022 to June 2023. SETTING PICU at a tertiary care hospital in China. PATIENTS Children 28 days to 16 years of age admitted with sepsis defined using Phoenix Sepsis Criteria. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Among 69 patients, 34 (49.3%) were categorized as having ARC. Designation as having ARC, vs. not, was associated with being younger (median 2.4 vs. 7.2 years, p < 0.001), hemodynamic and intrarenal flow changes, and higher C-reactive protein levels (31.0 vs. 12.0 mg/L, p < 0.05). None of the 34 patients with ARC developed acute kidney injury, but 22 of 35 non-ARC patients did. ARC was associated with shorter PICU stays (median 7 vs. 11 days, p < 0.05). Univariate regression analyses identified fluid balance, cardiac function parameters, renal resistive index, and inflammatory markers as explanatory factors associated with ARC. CONCLUSIONS In this retrospective cohort of pediatric sepsis patients admitted to the PICU, the prevalence of early-onset ARC is around 50%, and younger patients may be at risk. The presence of ARC is associated with hemodynamic and inflammatory responses. Taken together, more prospective work is needed, with an emphasis on drug-level targeting and a better understanding of interactions with intrarenal pathophysiology.
Collapse
Affiliation(s)
- Lili Xu
- All authors: Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Boer-Pérez FS, Lima-Rogel V, Mejía-Elizondo AR, Medellín-Garibay SE, Rodríguez-Báez AS, Rodríguez-Pinal CJ, Milán-Segovia RDC, Romano-Moreno S. External Evaluation of Population Pharmacokinetic Models of Piperacillin in Preterm and Term Patients from Neonatal Intensive Care. Eur J Drug Metab Pharmacokinet 2024; 49:595-607. [PMID: 38951408 DOI: 10.1007/s13318-024-00906-3] [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] [Accepted: 06/10/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND AND OBJECTIVES Piperacillin/tazobactam is extensively used off-label to treat late-onset neonatal sepsis, but safety and pharmacokinetic data in this population are limited. Additionally, the organic immaturity of the newborns contributes to a high piperacillin pharmacokinetic variability. This affects the clinical efficacy of the antibiotic treatment and increases the probability of developing drug resistance. This study aimed to evaluate the predictive performance of reported piperacillin population pharmacokinetic models for their application in a model-informed precision dosing strategy in preterm and term Mexican neonatal intensive care patients. METHODS Published population pharmacokinetic models for piperacillin which included neonates in their study population were identified. From the reference models, structured models, population pharmacokinetic parameters, and interindividual and residual variability data were extracted to be replicated in pharmacokinetic software (NONMEM® version 7.4). For the clinical study, a sampling schedule was designed, and 2-3 blood samples of 250 µL were taken from neonates who met the inclusion criteria. Piperacillin plasma concentrations were determined by liquid chromatography/tandem mass spectrometry. The clinical treatment data were collected, and piperacillin plasma concentrations were estimated using reference pharmacokinetic models for an a priori or Bayesian approach. Statistical methods were used in terms of bias and precision to evaluate the differences between observed and estimated neonatal piperacillin plasma concentrations with the different approaches and to identify the pharmacokinetic model that best fits the neonatal data. RESULTS A total of 70 plasma samples were collected from 25 neonatal patients, of which 15 were preterm neonates. The overall median value (range) postnatal age, gestational age, body weight, and serum creatinine at the sampling collecting day were 12 (3-26) days, 34.2 (26-41.1) weeks, 1.78 (0.08-3.90) Kg, 0.47 (0.20-0.90) mg/dL, respectively. Three population pharmacokinetic models for piperacillin in infants up to 2 months were identified, and their predictive performance in neonatal data was evaluated. No pharmacokinetic model was suitable for our population using an a priori approach. The model published by Cohen-Wolkowiez et al. in 2014 with a Bayesian approach showed the best performance of the pharmacokinetic models evaluated in our neonatal data. The procedure requires two blood samples (predose and postdose), and, when applied, it predicted 66.6% of the observations with a relative median absolute predicted error of less than 30%. CONCLUSIONS The population pharmacokinetic model developed by Cohen-Wolkowiez et al. in 2014 demonstrated superior performance in predicting the plasma concentration of piperacillin in preterm and term Mexican neonatal intensive care patients. The Bayesian approach, including two different piperacillin plasma concentrations, was clinically acceptable regarding bias and precision. Its application for model-informed precision dosing can be an option to optimize the piperacillin dosage in our population.
Collapse
Affiliation(s)
- Frida S Boer-Pérez
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México
| | - Victoria Lima-Rogel
- Neonatal Intensive Care Unit, Hospital Central "Dr. Ignacio Morones Prieto", San Luis Potosí, México
| | - Ana R Mejía-Elizondo
- Neonatal Intensive Care Unit, Hospital Central "Dr. Ignacio Morones Prieto", San Luis Potosí, México
| | - Susanna E Medellín-Garibay
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México
| | - Ana S Rodríguez-Báez
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México
| | - Cristian J Rodríguez-Pinal
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México
| | - Rosa Del C Milán-Segovia
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México
| | - Silvia Romano-Moreno
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, #6, Dr. Manuel Nava Martinez, S.L.P. PO Box 78210, San Luis Potosí, México.
| |
Collapse
|
8
|
Vidal E, Ray PE. Acute kidney injury during the first week of life: time for an update? Pediatr Nephrol 2024; 39:2543-2547. [PMID: 38332124 DOI: 10.1007/s00467-024-06310-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Affiliation(s)
- Enrico Vidal
- Pediatric Nephrology Unit, Department for Women's and Children's Health, University-Hospital of Padua, Padua, Italy.
- Department of Medicine (DMED), University of Udine, Udine, Italy.
| | - Patricio E Ray
- Child Health Research Center and Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
9
|
Cheng Y, Zhang Y, Zhang Y, Liu M, Zhao L. Population pharmacokinetic analyses of methotrexate in pediatric patients: a systematic review. Eur J Clin Pharmacol 2024; 80:965-982. [PMID: 38498098 DOI: 10.1007/s00228-024-03665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Methotrexate is widely utilized in the chemotherapy of malignant tumors and autoimmune diseases in the pediatric population, but dosing can be challenging. Several population pharmacokinetic models were developed to characterize factors influencing variability and improve individualization of dosing regimens. However, significant covariates included varied across studies. The primary objective of this review was to summarize and discuss population pharmacokinetic models of methotrexate and covariates that influence pharmacokinetic variability in pediatric patients. METHODS Systematic searches were conducted in the PubMed and EMBASE databases from inception to 7 July 2023. Reporting Quality was evaluated based on a checklist with 31 items. The characteristics of studies and information for model construction and validation were extracted, summarized, and discussed. RESULTS Eighteen studies (four prospective studies and fourteen retrospective studies with sample sizes of 14 to 772 patients and 2.7 to 93.1 samples per patient) were included in this study. Two-compartment models were the commonly used structural models for methotrexate, and the clearance range of methotrexate ranged from 2.32 to 19.03 L/h (median: 6.86 L/h). Body size and renal function were found to significantly affect the clearance of methotrexate for pediatric patients. There were limited reports on the role of other covariates, such as gene polymorphisms and co-medications, in the pharmacokinetic parameters of methotrexate pediatric patients. Internal and external evaluations were used to assess the performance of the population pharmacokinetic models. CONCLUSION A more rigorous external evaluation needs to be performed before routine clinical use to select the appropriate PopPK model. Further research is necessary to incorporate larger cohorts or pool analyses in specific susceptible pediatric populations to improve the understanding of predicted exposure profiles and covariate identification.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pharmacy, Shengjing Hospital Affiliated to China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Yujia Zhang
- Department of Pharmacy, Shengjing Hospital Affiliated to China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Ying Zhang
- Department of Pharmacy, Shengjing Hospital Affiliated to China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Rd, Gulou, Fuzhou, 350001, Fujian Province, People's Republic of China.
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital Affiliated to China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
10
|
Ferreira Menoni SM, Leon LL, de Lima RG, Lutaif ACGDB, Prates LC, Palma LMP, Costa SCB, Belangero VMS, Bonon SHA. Characterization of Herpesviridae Family Members, BK Virus, and Adenovirus in Children and Adolescents with Nephrotic Syndrome. Viruses 2024; 16:1017. [PMID: 39066180 PMCID: PMC11281385 DOI: 10.3390/v16071017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 07/28/2024] Open
Abstract
Since the significance of viral infections in children and adolescents with nephrotic syndrome (NS) is yet to be defined, this study intended to estimate the occurrence, pattern, and outcomes of some DNA viral infections in children with NS. METHODS A prospective study was conducted to determine the genome identification of the viruses Epstein-Barr (EBV), human cytomegalovirus (HCMV), human herpesvirus 6 (HHV-6 type A and type B) and 7 (HHV-7), polyomavirus (BKV), and human adenovirus (HAdV) in plasma and urine samples of pediatric patients with NS. RESULTS A total of 35 patients aged 1 to 18 years with NS and under immunosuppressant drugs participated in the study. Plasma and urine samples were collected at regular intervals during a median follow-up of 266 days (range 133-595), and DNA was analyzed to detect the selected DNA viruses. Eleven patients (31.4%) had active virus infections, and patterns were classified as coinfection, recurrent, and consecutive. Of these, six patients (54.5%) presented viral coinfection, six (54.5%) viral recurrence, and seven patients (63.3%) had viral consecutive infection. Ten of the eleven patients with active infection had a proteinuria relapse (91%) and eight (72.7%) were hospitalized (p = 0.0022). Active HCMV infection was the most frequent infection and was observed in six patients (54.5%), three of the eleven patients (27.2%) had suspected HCMV disease in the gastrointestinal tract, and one had HHV-7 coinfection. The frequency of other infections was: 9% for HHV-6, 45.5% for BKV, 27.3% for HHV-7, 18.2% for EBV, and 18.2% for HAdV. CONCLUSION viral infections, especially HCMV, can be an important cause of morbidity and nephrotic syndrome relapse in children.
Collapse
Affiliation(s)
- Silvia Mendonça Ferreira Menoni
- Laboratory of Virology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (S.M.F.M.); (L.L.L.); (R.G.d.L.); (S.C.B.C.)
| | - Lucas Lopes Leon
- Laboratory of Virology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (S.M.F.M.); (L.L.L.); (R.G.d.L.); (S.C.B.C.)
| | - Rodrigo Gonçalves de Lima
- Laboratory of Virology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (S.M.F.M.); (L.L.L.); (R.G.d.L.); (S.C.B.C.)
| | - Anna Cristina Gervásio de Brito Lutaif
- Integrated Nephrology Center Unit, Pediatric Nephrology, Department of Pediatrics, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (A.C.G.d.B.L.); (L.C.P.); (L.M.P.P.); (V.M.S.B.)
| | - Liliane Cury Prates
- Integrated Nephrology Center Unit, Pediatric Nephrology, Department of Pediatrics, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (A.C.G.d.B.L.); (L.C.P.); (L.M.P.P.); (V.M.S.B.)
| | - Lilian Monteiro Pereira Palma
- Integrated Nephrology Center Unit, Pediatric Nephrology, Department of Pediatrics, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (A.C.G.d.B.L.); (L.C.P.); (L.M.P.P.); (V.M.S.B.)
| | - Sandra Cecília Botelho Costa
- Laboratory of Virology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (S.M.F.M.); (L.L.L.); (R.G.d.L.); (S.C.B.C.)
| | - Vera Maria Santoro Belangero
- Integrated Nephrology Center Unit, Pediatric Nephrology, Department of Pediatrics, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (A.C.G.d.B.L.); (L.C.P.); (L.M.P.P.); (V.M.S.B.)
| | - Sandra Helena Alves Bonon
- Laboratory of Virology, School of Medical Sciences, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-887, Brazil; (S.M.F.M.); (L.L.L.); (R.G.d.L.); (S.C.B.C.)
| |
Collapse
|
11
|
Pokorná P, Michaličková D, Tibboel D, Berner J. Meropenem Disposition in Neonatal and Pediatric Extracorporeal Membrane Oxygenation and Continuous Renal Replacement Therapy. Antibiotics (Basel) 2024; 13:419. [PMID: 38786147 PMCID: PMC11117356 DOI: 10.3390/antibiotics13050419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
This study aimed to characterize the impact of extracorporeal membrane oxygenation (ECMO) on the pharmacokinetics (PK) of meropenem in neonates and children and to provide recommendations for meropenem dosing in this specific population of patients. Therapeutic drug monitoring (152 meropenem plasma concentrations) data from 45 patients (38 received ECMO) with a body weight (BW) of 7.88 (3.62-11.97) kg (median (interquartile range)) and postnatal age of 3 (0-465) days were collected. The population PK analysis was performed using NONMEM V7.3.0. Monte Carlo simulations were performed to assess the probability of target achievement (PTA) for 40% of time the free drug remained above the minimum inhibitory concentration (fT > MIC) and 100% fT > MIC. BW was found to be a significant covariate for the volume of distribution (Vd) and clearance (CL). Additionally, continuous renal replacement therapy (CRRT) was associated with a two-fold increase in Vd. In the final model, the CL and Vd for a typical patient with a median BW of 7.88 kg that was off CRRT were 1.09 L/h (RSE = 8%) and 3.98 L (14%), respectively. ECMO did not affect meropenem PK, while superimposed CRRT significantly increased Vd. We concluded that current dosing regimens provide acceptably high PTA for MIC ≤ 4 mg/L for 40% fT > MIC, but individual dose adjustments are needed for 100% fT > MIC.
Collapse
Affiliation(s)
- Pavla Pokorná
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, 128 00 Prague, Czech Republic
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 128 00 Prague, Czech Republic
- Department of Physiology and Pharmacology, Karolinska Institute and Karolinska University Hospital, 171 77 Stockholm, Sweden
- Department of Pediatric Surgery, Erasmus Medical Center Sophia Children’s Hospital, 3062 PA Rotterdam, The Netherlands
| | - Danica Michaličková
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, 128 00 Prague, Czech Republic
| | - Dick Tibboel
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 128 00 Prague, Czech Republic
- Department of Pediatric Surgery, Erasmus Medical Center Sophia Children’s Hospital, 3062 PA Rotterdam, The Netherlands
| | - Jonas Berner
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 128 00 Prague, Czech Republic
- Department of Physiology and Pharmacology, Karolinska Institute and Karolinska University Hospital, 171 77 Stockholm, Sweden
- Pediatric Perioperative Medicine and Intensive Care, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
12
|
Natraj R, Bhaskaran AK, Rola P, Haycock K, Siuba MTT, Ranjit S. Venous Congestion Assessed by Venous Excess Ultrasound (VExUS) and Acute Kidney Injury in Children with Right Ventricular Dysfunction. Indian J Crit Care Med 2024; 28:447-452. [PMID: 38738193 PMCID: PMC11080090 DOI: 10.5005/jp-journals-10071-24705] [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: 02/26/2024] [Accepted: 04/03/2024] [Indexed: 05/14/2024] Open
Abstract
Background Right ventricular dysfunction (RVD) is a complication following congenital cardiac surgery in children and can lead to systemic venous congestion, low cardiac output, and organ dysfunction. Venous congestion can be transmitted backwards and adversely affect encapsulated organs such as the kidneys. Primary objective To investigate the association between systemic venous congestion, as estimated by Venous Excess Ultrasound (VExUS), and the occurrence of acute kidney injury (AKI) in children with RVD following congenital heart surgery. Secondary objectives included comparing changes in VExUS scores after initiating treatment for RVD and venous congestion. Methods and results This was a prospective observational study in children with RVD. The VExUS study was performed on day 1, day 2, and day 3 and categorized as VExUS-1, VExUS-2, and VExUS-3. Among 43 patients with RVD and dilated inferior vena cava, 19/43 (44%), 10/43 (23%), and 12/43 (28%) were VExUS-2 and VExUS-3, respectively. There was an association between severe RVD and elevated pulmonary artery systolic pressures and a VExUS score >2. A significant association was observed between central venous pressure (CVP) measurements and VExUS. Among 31 patients with a high VExUS score >2, 18 (58%) had AKI. Additionally, improvement in CVP and fluid balance was associated with improving VExUS scores following targeted treatment for RVD. Conclusion VExUS serves as a valuable bedside tool for diagnosing and grading venous congestion through ultrasound Doppler. An elevated VExUS score was associated with the occurrence of AKI, and among the components of VExUS, portal vein pulsatility may be useful as a predictor of AKI. How to cite this article Natraj R, Bhaskaran AK, Rola P, Haycock K, Siuba MTT, Ranjit S. Venous Congestion Assessed by Venous Excess Ultrasound (VExUS) and Acute Kidney Injury in Children with Right Ventricular Dysfunction. Indian J Crit Care Med 2024;28(5):447-452.
Collapse
Affiliation(s)
- Rajeswari Natraj
- Pediatric Intensive Care Unit and Cardiothoracic Services, Apollo Children's Hospital, Chennai, Tamil Nadu, India
| | | | - Philippe Rola
- Intensive Care Unit, Santa Cabrini Hospital, CEMTL; Intensive Care Unit, University of Montreal, Montreal, Canada
| | - Korbin Haycock
- Emergency Department, Riverside University Health System Medical Center, Moreno Valley, California, Loma Linda, United States
| | - Matthew TT Siuba
- Intensive Care Unit, Department of Critical Care Medicine, Integrated Hospital Care, Cleveland, Ohio, United States
| | - Suchitra Ranjit
- Department of Pediatric Intensive Care Unit, Apollo Children's Hospital, Chennai, Tamil Nadu, India
| |
Collapse
|
13
|
Hollander SA, Chung S, Reddy S, Zook N, Yang J, Vella T, Navaratnam M, Price E, Sutherland SM, Algaze CA. Intraoperative and Postoperative Hemodynamic Predictors of Acute Kidney Injury in Pediatric Heart Transplant Recipients. J Pediatr Intensive Care 2024; 13:37-45. [PMID: 38571984 PMCID: PMC10987224 DOI: 10.1055/s-0041-1736336] [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: 08/05/2021] [Accepted: 08/28/2021] [Indexed: 10/20/2022] Open
Abstract
Acute kidney injury (AKI) is common after pediatric heart transplantation (HT) and is associated with inferior patient outcomes. Hemodynamic risk factors for pediatric heart transplant recipients who experience AKI are not well described. We performed a retrospective review of 99 pediatric heart transplant patients at Lucile Packard Children's Hospital Stanford from January 1, 2015, to December 31, 2019, in which clinical and demographic characteristics, intraoperative perfusion data, and hemodynamic measurements in the first 48 postoperative hours were analyzed as risk factors for severe AKI (Kidney Disease: Improving Global Outcomes [KDIGO] stage ≥ 2). Univariate analysis was conducted using Fisher's exact test, Chi-square test, and the Wilcoxon rank-sum test, as appropriate. Multivariable analysis was conducted using logistic regression. Thirty-five patients (35%) experienced severe AKI which was associated with lower intraoperative cardiac index ( p = 0.001), higher hematocrit ( p < 0.001), lower body temperature ( p < 0.001), lower renal near-infrared spectroscopy ( p = 0.001), lower postoperative mean arterial blood pressure (MAP: p = 0.001), and higher central venous pressure (CVP; p < 0.001). In multivariable analysis, postoperative CVP >12 mm Hg (odds ratio [OR] = 4.27; 95% confidence interval [CI]: 1.48-12.3, p = 0.007) and MAP <65 mm Hg (OR = 4.9; 95% CI: 1.07-22.5, p = 0.04) were associated with early severe AKI. Children with severe AKI experienced longer ventilator, intensive care, and posttransplant hospital days and inferior survival ( p = 0.01). Lower MAP and higher CVP are associated with severe AKI in pediatric HT recipients. Patients, who experienced AKI, experienced increased intensive care unit (ICU) morbidity and inferior survival. These data may guide the development of perioperative renal protective management strategies to reduce AKI incidence and improve patient outcomes.
Collapse
Affiliation(s)
- Seth A. Hollander
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, California, United States
| | - Sukyung Chung
- Quantitative Sciences Unit, Stanford University, Stanford, California, United States
| | - Sushma Reddy
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, California, United States
| | - Nina Zook
- Department of Pediatrics, Stanford University, Stanford, California, United States
| | - Jeffrey Yang
- Department of Pediatrics, Stanford University, Stanford, California, United States
| | - Tristan Vella
- Perfusion Services, Lucile Packard Children's Hospital Stanford, Palo Alto, California, United States
| | - Manchula Navaratnam
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California, United States
| | - Elizabeth Price
- Patient Care Services, Cardiovascular Intensive Care Unit, Lucile Packard Children's Hospital Stanford, Palo Alto, California, United States
| | - Scott M. Sutherland
- Department of Pediatrics (Nephrology), Scott M Sutherland, Stanford University School of Medicine, Stanford, California, United States
| | - Claudia A. Algaze
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, California, United States
- Center for Pediatric and Maternal Value, Stanford University, Palo Alto, California, Unites States
| |
Collapse
|
14
|
Chen Y, Lan J, Zhu L, Dong M, Wang Y, Li Z. Is the current therapeutic dosage of nadroparin adequate for neonates and infants under 8 months with thromboembolic disease? a population pharmacokinetic study from a national children's medical center. Front Pharmacol 2024; 15:1331673. [PMID: 38357306 PMCID: PMC10864485 DOI: 10.3389/fphar.2024.1331673] [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: 11/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
Objectives: Nadroparin, a low-molecular-weight-heparin is commonly used off-label in neonates and infants for thromboembolic events prevention. However, the recommended dosing regimen often fails to achieve therapeutic target ranges. This study aimed to develop a population pharmacokinetic (PK) model of nadroparin to determine an appropriate dosing regimen for neonates and infants less than 8 months. Methods: A retrospective chart review was conducted on patients treated with nadroparin at Children's Hospital of Fudan University between July 2021 and December 2023. A population PK model was developed using anti-Xa levels, and its predictive performance was evaluated internally. Monte Carlo simulations were performed to design an initial dosing schedule targeting anti-Xa levels between 0.5 and 1 IU/mL. Results: A total of 40 neonates and infants aged less than 8 months with gestational age ranging from 25 to 41 weeks treated with nadroparin were enrolled in the study for analysis. A one-compartment PK model with first order absorption and elimination was adequately fitted to the data. Creatinine clearance was identified as a significant factor contributing to inter-individual variability in clearance. The typical population parameter estimates of clearance, distribution volume and absorption rate in this population were 0.211 L/h, 1.55 L and 0.495 h-1, respectively. Our findings suggest that current therapeutic doses of nadroparin (150-200 IU/kg q12 h) may result in subtherapeutic exposure, thus higher doses might be required. Conclusion: The present study offers the first estimation of PK parameters for nadroparin in preterm or term neonates and infants less than 8 months utilizing the model. Our findings have potential implications for recommending initial personalized dosages, particularly among patient populations exhibiting similar characteristics.
Collapse
Affiliation(s)
- Yewei Chen
- Department of Pharmacy, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Jianger Lan
- Department of Pharmacy, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Lin Zhu
- Department of Pharmacy, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Yi Wang
- Department of Neurology, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Zhiping Li
- Department of Pharmacy, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
15
|
Bradley JS, Makieieva N, Tøndel C, Roilides E, Kelly MS, Patel M, Vaddady P, Maniar A, Zhang Y, Paschke A, Chen LF. Pharmacokinetics, Safety, and Tolerability of Imipenem/Cilastatin/Relebactam in Children with Confirmed or Suspected Gram-Negative Bacterial Infections: A Phase 1b, Open-Label, Single-Dose Clinical Trial. J Clin Pharmacol 2023; 63:1387-1397. [PMID: 37562063 DOI: 10.1002/jcph.2334] [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: 04/18/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Imipenem/cilastatin/relebactam is approved for the treatment of serious gram-negative bacterial infections in adults. This study assessed the pharmacokinetics (PK), safety, and tolerability of a single dose of imipenem/cilastatin/relebactam (with a fixed 2:1 ratio of imipenem/cilastatin to relebactam, and with a maximum dose of 15 mg/kg imipenem and 15 mg/kg cilastatin [≤500 mg imipenem and ≤500 mg cilastatin] and 7.5 mg/kg relebactam [≤250 mg relebactam]) in children with confirmed/suspected gram-negative bacterial infections receiving standard-of-care antibacterial therapy. In this phase 1, noncomparative study (ClinicalTrials.gov identifier, NCT03230916), PK parameters from 46 children were analyzed using both population modeling and noncompartmental analysis. The PK/pharmacodynamic (PD) target for imipenem was percent time of the dosing interval that unbound plasma concentration exceeded the minimum inhibitory concentration (%fT>MIC) of ≥30% (MIC = 2 mcg/mL). For relebactam, the PK/PD target was a free drug area under the plasma concentration-time curve (AUC) normalized to MIC (at 2 mcg/mL) of ≥8.0 (equivalent to an AUC from time zero extrapolated to infinity of ≥20.52 mcg·h/mL). Safety was assessed up to 14 days after drug infusion. For imipenem, the ranges for the geometric mean %fT>MIC and maximum concentration (Cmax ) across age cohorts were 56.5%-93.7% and 32.2-38.2 mcg/mL, respectively. For relebactam, the ranges of the geometric mean Cmax and AUC from 0 to 6 hours across age cohorts were 16.9-21.3 mcg/mL and 26.1-55.3 mcg·h/mL, respectively. In total, 8/46 (17%) children experienced ≥1 adverse events (AEs) and 2/46 (4%) children experienced nonserious AEs that were deemed drug related by the investigator. Imipenem and relebactam exceeded plasma PK/PD targets; single doses of imipenem/cilastatin/relebactam were well tolerated with no significant safety concerns identified. These results informed imipenem/cilastatin/relebactam dose selection for further pediatric clinical evaluation.
Collapse
Affiliation(s)
- John S Bradley
- Department of Pediatrics, University of California San Diego School of Medicine and Rady Children's Hospital of San Diego, San Diego, CA, USA
| | - Nataliia Makieieva
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, and Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Emmanuel Roilides
- Third Department of Pediatrics, Infectious Diseases Unit, School of Medicine, Aristotle University and Hippokration General Hospital, Thessaloniki, Greece
| | - Matthew S Kelly
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | - Pavan Vaddady
- Merck & Co. Inc, Rahway, NJ, USA
- Daiichi Sankyo, Inc., Basking Ridge, NJ, USA
| | | | | | | | | |
Collapse
|
16
|
Yan R, Zhang C, Wang C, Sun Z, Peng X. Evaluation of glomerular filtration rate estimation equations based on serum creatinine in healthy Chinese children and adolescents: a nationwide cross-sectional study. BMJ Paediatr Open 2023; 7:e002132. [PMID: 37827805 PMCID: PMC10582894 DOI: 10.1136/bmjpo-2023-002132] [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: 06/13/2023] [Accepted: 08/29/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Several equations for glomerular filtration rate (GFR) estimation based on serum creatinine (SCr) have been proposed for children, but most were developed among patients with kidney disease. The association between SCr and GFR may be distorted by kidney dysfunction and thus not applicable to healthy children. This study aimed to evaluate the applicability of existing SCr-based GFR estimation equations in healthy Chinese children. METHODS GFR estimation equations that developed in healthy children were mainly analysed, including the Flanders Metadata (FM), simple height-independent (Simple), full age spectrum (FAS) and FAS-height equations. The FM equation assumed that GFR is proportional to the ratio of height to SCr. The Simple, FAS and FAS-height equations assumed that the ratio of GFR to population mean is equal to the reciprocal ratio of SCr to population mean (denoted by Q). Estimated GFR were calculated using data of SCr, age, sex and height collected from 12 208 healthy Chinese children aged 3 months to <20 years. The performance of GFR estimation equations was evaluated by the sex and age distribution of the estimated GFR and the deviation from the measured GFR reported by other literatures. RESULTS The FM and Simple equations performed well in their applicable age of 1 month to 14 years, but presented undesirable sex difference after adolescence. The FAS and FAS-height equations showed reasonable development trend of estimated GFR throughout childhood, and the FAS equation had higher consistency than the FAS-height equation compared with measured GFR in healthy children. The GFR estimated by the FAS equation increased with age before 2 years, and reached the adult level thereafter without important sex difference. CONCLUSIONS The FAS equation is applicable to healthy Chinese children.
Collapse
Affiliation(s)
- Ruohua Yan
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Chao Zhang
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Chen Wang
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Zimo Sun
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Xiaoxia Peng
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| |
Collapse
|
17
|
Matsell DG, Catapang M, Becknell B. Predicting outcomes in children with congenital anomalies of the kidney and urinary tract. Pediatr Nephrol 2023; 38:3407-3415. [PMID: 37133803 PMCID: PMC10962402 DOI: 10.1007/s00467-023-05992-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Congenital anomalies of the kidney and urinary tract (CAKUT) are the most frequent causes of childhood chronic kidney disease (CKD). Using a large CAKUT cohort, we sought to identify the predictors of CKD and to develop a prediction model that informs a risk-stratified clinical pathway. METHODS This was a retrospective cohort study including cases with multicystic dysplastic kidneys (MCDK), unilateral kidney agenesis (UKA), kidney hypoplasia (KH), and posterior urethral valves (PUV). We identified risk factors for CKD (estimated glomerular filtration rate [eGFR] <60 ml/min/1.73 m2) and tested their performance in an adjusted multivariate binary regression model. Prediction probability scores for CKD were used to separate cases likely to develop complications from those not needing specialist follow-up. RESULTS We identified 452 eligible cases of CAKUT with 22% developing CKD. Strongest associations with CKD included primary diagnosis (OR 3.5, 95% CI 2.6-4.6), preterm delivery (OR 2.3, 95% CI 1.2-4.4), non-kidney anomalies (OR 1.8, 95% CI 1.1-3), first eGFR<90 (OR 8.9, 95% CI 4.4-18.1), small kidney size (OR 9, 95% CI 4.9-16.6), and additional kidney anomalies (OR 1.6, 95% CI 1.2-2.8). PUV (OR 4.7, 95% CI 1.5-15.3), first eGFR <90 (OR 4.4, 95% CI 2-9.7), and kidney length to body length ratio <7.9 (OR 4.2, 95% CI 1.9-9.2) were independent predictors of CKD. The regression model had a prediction accuracy of 80% and a prediction probability c-statistic of 0.81. CONCLUSION Using a large combined CAKUT cohort we identified risk factors for CKD. Our prediction model provides the first steps towards a risk-stratified clinical pathway. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Douglas G Matsell
- University of British Columbia, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
| | - Marisa Catapang
- University of British Columbia, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Brian Becknell
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
18
|
Li QY, van den Anker J, Wu YE, Hao GX, Zhao W. Optimizing ganciclovir and valganciclovir dosing regimens in pediatric patients with cytomegalovirus infection: a spotlight on therapeutic drug monitoring. Expert Rev Clin Pharmacol 2023; 16:727-739. [PMID: 36794592 DOI: 10.1080/17512433.2023.2181161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Infants and immunocompromised children with cytomegalovirus (CMV) infection have significant morbidity and mortality. Ganciclovir (GCV) and its oral prodrug valganciclovir (VGCV) are the major antiviral options of choice for the prophylaxis and treatment of CMV infection. However, with the currently recommended dosing regimens used in pediatric patients, large intra- and inter-individual variability of pharmacokinetic (PK) parameters and exposure are observed. AREAS COVERED This review describes the PK and pharmacodynamic (PD) characteristics of GCV and VGCV in pediatrics. Moreover, the role of therapeutic drug monitoring (TDM) and current clinical practice for GCV and VGCV dosing regimens optimization in pediatrics are discussed. EXPERT OPINION GCV/VGCV TDM has shown the potential value to improve the benefit/risk ratio in pediatrics when using the therapeutic ranges derived from adults. However, well-designed studies are required to evaluate the relationship of TDM with clinical outcomes. Furthermore, studies to explore the children-specific dose-response-effect relationships will be helpful to facilitate the TDM practice. In the clinical setting, optimal sampling methods such as limited sampling strategies for pediatrics can be used in TDM and intracellular ganciclovir triphosphate may be used as an alternative TDM marker.
Collapse
Affiliation(s)
- Qiu-Yue Li
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Medical Center, Washington, DC, USA
- Departments of Pediatrics, Pharmacology & Physiology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
- Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Basel, Switzerland
| | - Yue-E Wu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| |
Collapse
|
19
|
Matcha S, Dillibatcha J, Raju AP, Chaudhari BB, Moorkoth S, Lewis LE, Mallayasamy S. Predictive Performance of Population Pharmacokinetic Models for Amikacin in Term Neonates. Paediatr Drugs 2023; 25:365-375. [PMID: 36943583 PMCID: PMC10097735 DOI: 10.1007/s40272-023-00564-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND AND OBJECTIVE Amikacin is preferred in treating Gram-negative infections in neonates and it has a narrow therapeutic window. The population pharmacokinetic modeling approach can aid in designing optimal dosage regimens for amikacin in neonates. In this study, we attempted to identify the suitable population pharmacokinetic model from the published reports for the study population from an Indian setting. METHODS Published population pharmacokinetic studies for amikacin in neonates were identified. Data on structural models and typical pharmacokinetic parameters were extracted from the studies. For the clinical study, neonates who met the inclusion criteria were enrolled in the study from the NICU, Kasturba Medical College, Manipal, during Jan 2020 to March 2022. Drug concentrations were estimated, and demographic and clinical data were collected. Identified population pharmacokinetic models were used to predict the amikacin concentrations in neonates. Predicted concentrations were compared against the observed concentrations. Differences between predicted and observed concentrations were quantified using statistical measures. The population pharmacokinetic model, which was able to predict the data well, is considered a suitable model for the study population. Dosing regimens were suggested for neonates using the pharmacometric simulation approach generated by the selected model. RESULTS A total of 43 plasma samples were collected from 31 neonates. Twelve population pharmacokinetic models were found for amikacin in neonates. The predictive performance of the 12 studies was performed using clinical data. A two-compartment model reported by Illamola et al. predicted the amikacin concentrations better than other models. Illamola et al. reported creatinine clearance and body weight as the significant covariates impacting the pharmacokinetic parameters of amikacin. This model was able to predict the clinical data with 29.97% and 0.686 of relative median absolute prediction error and relative root mean square error, respectively, which is the best among the published models. The Illamola et al. model was selected as the final model to perform pharmacometric simulations for the subjects with different combinations of creatinine clearance and body weight. Dosage regimens were designed to attain target therapeutic concentrations for the virtual subjects and a nomogram was developed. CONCLUSIONS The population pharmacokinetic model reported by the Illamola et al. model was selected as the final model to explain the clinical data with the lowest relative median absolute prediction error and relative root mean square error when compared with other models. An amikacin nomogram was developed for the neonates whose creatinine clearance and body weight ranged between 10 and 90 mL/min and between 2 and 4 kg, respectively. A developed nomogram can assist clinicians to design an optimal dosage regimen of amikacin for term neonates.
Collapse
Affiliation(s)
- Saikumar Matcha
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jayashree Dillibatcha
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Arun Prasath Raju
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bhim Bahadur Chaudhari
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sudheer Moorkoth
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Leslie E Lewis
- Department of Pediatrics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| | - Surulivelrajan Mallayasamy
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
- Centre for Pharmacometrics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
20
|
Vandenberghe W, De Loor J, Francois K, Vandekerckhove K, Herck I, Vande Walle J, Peperstraete H, Bové T, De Wolf D, Nuytinck L, De Waele JJ, Meyer E, Hoste EAJ. Potential of Urine Biomarkers CHI3L1, NGAL, TIMP-2, IGFBP7, and Combinations as Complementary Diagnostic Tools for Acute Kidney Injury after Pediatric Cardiac Surgery: A Prospective Cohort Study. Diagnostics (Basel) 2023; 13:diagnostics13061047. [PMID: 36980354 PMCID: PMC10047361 DOI: 10.3390/diagnostics13061047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Acute kidney injury (AKI) is common after pediatric cardiac surgery (CS). Several urine biomarkers have been validated to detect AKI earlier. The objective of this study was to evaluate urine CHI3L1, NGAL, TIMP-2, IGFBP7, and NephroCheck® as predictors for AKI ≥ 1 in pediatric CS after 48 h and AKI ≥ 2 after 12 h. Pediatric patients (age < 18 year; body weight ≥ 2 kg) requiring CS were prospectively included. Urine CHI3L1, NGAL, TIMP-2, IGFBP7, and NephroCheck® were measured during surgery and intensive care unit (ICU) stay and corrected for urine dilution. One hundred and one pediatric patients were included. AKI ≥ 1 within 48 h after ICU admission occurred in 62.4% and AKI ≥ 2 within 12 h in 30.7%. All damage biomarkers predicted AKI ≥ 1 within 48 h after ICU admission, when corrected for urine dilution: CHI3L1 (AUC-ROC: 0.642 (95% CI, 0.535–0.741)), NGAL (0.765 (0.664–0.848)), TIMP-2 (0.778 (0.662–0.868)), IGFBP7 (0.796 (0.682–0.883)), NephroCheck® (0.734 (0.614–0.832)). Similarly, AKI ≥ 2 within 12 h was predicted by all damage biomarkers when corrected for urine dilution: uCHI3L1 (AUC-ROC: 0.686 (95% CI, 0.580–0.780)), NGAL (0.714 (0.609–0.804)), TIMP-2 (0.830 (0.722–0.909)), IGFBP7 (0.834 (0.725–0.912)), NephroCheck® (0.774 (0.658–0.865)). After pediatric cardiac surgery, the damage biomarkers urine CHI3L1, NGAL, TIMP-2, IGFBP7, and NephroCheck® reliably predict AKI after correction for urine dilution.
Collapse
Affiliation(s)
- Wim Vandenberghe
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-933-20802; Fax: +32-9-332-4995
| | - Jorien De Loor
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Katrien Francois
- Department of Cardiac Surgery, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Kristof Vandekerckhove
- Department of Pediatric Cardiology, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Ingrid Herck
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Johan Vande Walle
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pediatric Nephrology, ERKNET Centre, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Harlinde Peperstraete
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Thierry Bové
- Department of Cardiac Surgery, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Daniël De Wolf
- Department of Pediatric Cardiology, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lieve Nuytinck
- Health, Innovation and Research Institute UZ Gent, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jan J. De Waele
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, 9000 Ghent, Belgium
| | - Eric A. J. Hoste
- Department of Intensive Care Medicine, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
21
|
Smeets NJL, Hartmann O, Schulte J, Schreuder MF, de Wildt SN. Proenkephalin A as a marker for glomerular filtration rate in critically ill children: validation against gold standard iohexol GFR measurements. Clin Chem Lab Med 2023; 61:104-111. [PMID: 36283061 DOI: 10.1515/cclm-2022-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/07/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Accurate determination of glomerular filtration rate (GFR) is important. Several endogenous biomarkers exist for estimating GFR, yet, they have limited accuracy, especially in the paediatric population. Proenkephalin A 119-159 (PENK) is a novel and promising GFR marker, but its relation with age in children remains unknown. Also, the value of PENK has never been validated against measured GFR (mGFR) in children when compared to traditional GFR markers including serum creatinine (SCr), SCr-based estimated GFR (eGFR) and cystatin C (cysC). METHODS Critically ill children and term-born neonates were included in this single-centre, prospective study. Iohexol-based mGFR, SCr, and cysC were determined in each patient. eGFR was calculated using the bedside Schwartz equation, incorporating SCr and height. Spearman correlation coefficients were calculated to determine the correlation between mGFR and PENK, SCr, cysC and eGFR. RESULTS For 97 patients (56 children and 41 neonates), mGFR, SCr, cysC and PENK levels were available. PENK levels were higher in young children and decreased to adult PENK reference values around two years of age. PENK levels were highly correlated with mGFR (ρ=-0.88, p<0.001), and similar to mGFR-eGFR correlation (ρ=-0.87, p<0.001). For cysC and SCr the correlation with mGFR was lower (ρ=-0.77 and ρ=-0.46, respectively. Both p<0.001). CONCLUSIONS The correlation of PENK with mGFR was as good as SCr-based eGFR-mGFR correlation. To determine the added value of PENK in paediatric clinical care and prior to implementation, PENK reference values are needed and the development and validation of a paediatric PENK-based eGFR equation is necessary.
Collapse
Affiliation(s)
- Nori J L Smeets
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.,Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | - Michiel F Schreuder
- Department of Pediatrics, Division of Pediatric Nephrology, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.,Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Shiraishi C, Kato H, Imai H, Iwamoto T. Impact of Extracorporeal Membrane Oxygenation in an Infant Treated with Vancomycin: A Case Report. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1839. [PMID: 36767208 PMCID: PMC9915104 DOI: 10.3390/ijerph20031839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Vancomycin is a glycopeptide antibiotic used for prophylaxis and treatment of infections caused by methicillin-resistant Staphylococcus aureus. Although major organ sizes and functions mature during infancy, pharmacokinetic studies, especially those focused on infants, are limited. Changes in extracorporeal membrane oxygenation-related drug disposition largely contribute to changes in pharmacokinetics. Here, pharmacokinetic profiles of vancomycin in an infant receiving extracorporeal membrane oxygenation therapy are presented. A two-month-old Japanese infant with moderately decreased renal function was started on 12.0 mg/kg vancomycin every 8 h from day X for prophylaxis of pneumonia during extracorporeal membrane oxygenation therapy. As the trough concentration of vancomycin observed on day X+3 was 27.1 μg/mL, vancomycin was then discontinued. The trough concentration decreased to 18.6 μg/mL 24 h after discontinuation, and 9.0 mg/kg vancomycin every 12 h was restarted from day X+5. On day X+6, the trough concentration increased to 36.1 μg/mL, and vancomycin therapy was again discontinued. On day X+7, the trough concentration decreased to 22.4 μg/mL. The pharmacokinetic profiles of vancomycin based on first-order conditional estimation in this infant were as follows: plasma clearance = 0.053 L/kg/hour, distribution volume = 2.19 L/kg, and half-life = 29.5 h. This research reported the prolonged half-life of vancomycin during extracorporeal membrane oxygenation in infants with moderately decreased renal function.
Collapse
Affiliation(s)
- Chihiro Shiraishi
- 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
| | - 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
| | - Hiroshi Imai
- Emergency and Critical Care Center, 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
|
23
|
Leow EH, Lee JH, Hornik CP, Ng YH, Hays T, Clark RH, Tolia VN, Greenberg RG. Congenital anomalies of the kidney and urinary tract (CAKUT) in critically ill infants: a multicenter cohort study. Pediatr Nephrol 2023; 38:161-172. [PMID: 35467155 DOI: 10.1007/s00467-022-05542-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/02/2022] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of the study was to determine the prevalence of congenital anomalies of the kidney and urinary tract (CAKUT) in the neonatal intensive care unit (NICU) and to evaluate risk factors associated with worse outcomes. We hypothesized that infants with CAKUT with extra-renal manifestations have higher mortality. METHODS This is a cohort study of all inborn infants who were diagnosed with any form of CAKUT discharged from NICUs managed by the Pediatrix Medical Group from 1997 to 2018. Logistic and linear regression models were used to analyze risk factors associated with in-hospital mortality. RESULTS The prevalence of CAKUT was 1.5% among infants hospitalized in 419 NICUs. Among the 13,383 infants with CAKUT analyzed, median gestational age was 35 (interquartile range [IQR] 31-38) weeks and median birth weight was 2.34 (IQR 1.54-3.08) kg. Overall in-hospital mortality for infants with CAKUT was 6.8%. Oligohydramnios (adjusted odds ratio [aOR] 4.5, 95% confidence interval [CI] 2.2-9.1, p < 0.001), extra-renal anomalies (aOR 2.5, 95% CI 2.0-3.1, p < 0.001), peak SCr (aOR 1.02, 95% CI 1.01-1.03, p < 0.001) and exposure to nephrotoxic medications (aOR 1.4, 95% CI 1.1-1.7, p = 0.01) were associated with increased mortality, while a history of urological surgery or intervention was associated with lower mortality (aOR 0.6, 95% CI 0.4-0.7, p < 0.001). CONCLUSIONS Infants hospitalized in the NICU who have CAKUT and the independent risk factors for mortality (e.g., oligohydramnios and presence of extra-renal anomalies) require close monitoring, minimizing of exposure to nephrotoxic drugs, and timely urological surgery or intervention. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Esther Huimin Leow
- Paediatric Nephrology, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore.
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Christoph P Hornik
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Yong Hong Ng
- Paediatric Nephrology, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, 229899, Singapore
| | - Thomas Hays
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York City, NY, USA
| | - Reese H Clark
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- The MEDNAX Center for Research, Education, Quality and Safety, Sunrise, FL, USA
| | - Veeral N Tolia
- The MEDNAX Center for Research, Education, Quality and Safety, Sunrise, FL, USA
- Department of Neonatology, Baylor University Medical Center and Pediatrix Medical Group, Dallas, TX, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
24
|
Glomerular filtration rate in critically ill neonates and children: creatinine-based estimations versus iohexol-based measurements. Pediatr Nephrol 2023; 38:1087-1097. [PMID: 35916956 PMCID: PMC9925555 DOI: 10.1007/s00467-022-05651-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) and augmented renal clearance (ARC), both alterations of the glomerular filtration rate (GFR), are prevalent in critically ill children and neonates. AKI and ARC prevalence estimates are based on estimation of GFR (eGFR) using serum creatinine (SCr), which is known to be inaccurate. We aimed to test our hypothesis that AKI prevalence will be higher and ARC prevalence will be lower in critically ill children when using iohexol-based measured GFR (mGFR), rather than using eGFR. Additionally, we aimed to investigate the performance of different SCr-based eGFR methods. METHODS In this single-center prospective study, critically ill term-born neonates and children were included. mGFR was calculated using a plasma disappearance curve after parenteral administration of iohexol. AKI diagnosis was based on the KDIGO criteria, SCr-based eGFR, and creatinine clearance (CrCL). Differences between eGFR and mGFR were determined using Wilcoxon signed-rank tests and by calculating bias and accuracy (percentage of eGFR values within 30% of mGFR values). RESULTS One hundred five children, including 43 neonates, were included. AKI prevalence was higher based on mGFR (48%), than with KDIGO or eGFR (11-40%). ARC prevalence was lower with mGFR (24%) compared to eGFR (38-51%). eGFR equations significantly overestimated mGFR (60-71 versus 41 ml/min/1.73 m2, p < 0.001-0.002). Accuracy was highest with eGFR equations based on age- and sex-dependent equations (up to 59%). CONCLUSION Iohexol-based AKI prevalence was higher and ARC prevalence lower compared to standard SCr-based eGFR methods. Age- and sex-dependent equations for eGFR (eGFR-Smeets for neonates and eGFR-Pierce for children) best approached measured GFR and should preferably be used to optimize diagnosis of AKI and ARC in this population. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
|
25
|
Forero-Delgadillo J, Ochoa V, Restrepo JM, Torres-Canchala L, Nieto-Aristizábal I, Ruiz-Ordoñez I, Sánchez A, Barrera MC, Jimenez CA, Tobón GJ. B-cell activating factor (BAFF) and its receptors' expression in pediatric nephrotic syndrome is associated with worse prognosis. PLoS One 2022; 17:e0277800. [PMID: 36399480 PMCID: PMC9674176 DOI: 10.1371/journal.pone.0277800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
AIM Immune pathogenesis of nephrotic syndrome (NS) is not completely understood. We aimed to evaluate the expression of B-cell activating factor (BAFF) and its receptors in renal samples from pediatric NS patients and its relationship with renal function survival. MATERIALS AND METHODS We conducted an ambispective study on 33 patients with pediatric NS. Immunohistochemistry for BAFF, TACI, BCMA and BR3 was performed. Markers were evaluated on podocytes and interstitial inflammatory infiltrates (III). We performed Kaplan-Meier curves to describe renal function survival according to markers' expression. RESULTS Thirty-three NS patients were included. Minimal change disease was seen in 21 (63.6%) patients, and focal segmental glomerulosclerosis in 12 (36.4%). BAFF was found in podocytes (18.2% of samples) and III (36.4% of samples), BAFF-R in one sample, TACI in 4 (podocytes and III), and BCMA in 5 samples of podocytes and 7 of III. BAFF on podocytes and III was associated with worst renal function at follow-up; those patients had 25% probability of having GFR >90 mL/min/1.73m2, versus 84.9% when absent (p = 0.0067). Patients with BAFF in III had 42.9% probability of having GFR>90 mL/min/1.73 m2, versus 94.1% when absent (p = 0.0063). CONCLUSION BAFF expression in renal biopsies could be a prognostic factor for renal function.
Collapse
Affiliation(s)
| | - Vanessa Ochoa
- Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Jaime M Restrepo
- Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
- Servicio de Nefrología Pediátrica, Fundación Valle del Lili, Cali, Colombia
| | | | | | - Ingrid Ruiz-Ordoñez
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, Colombia
| | - Aura Sánchez
- Servicio de Patología, Fundación Valle del Lili, Cali, Colombia
| | - María Claudia Barrera
- Universidad Icesi, CIRAT: Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
| | | | - Gabriel J Tobón
- Universidad Icesi, CIRAT: Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Laboratorio de Inmunología, Fundación Valle del Lili, Cali, Colombia
| |
Collapse
|
26
|
Mohr Lytsen R, Taageby Nielsen S, Kongsgaard Hansen M, Strandkjær N, Juul Rasmussen I, Axelsson Raja A, Vøgg RO, Sillesen AS, Kamstrup PR, Schmidt IM, Iversen K, Bundgaard H, Frikke-Schmidt R. Markers of Kidney Function in Early Childhood and Association With Maternal Comorbidity. JAMA Netw Open 2022; 5:e2243146. [PMID: 36409493 PMCID: PMC9679880 DOI: 10.1001/jamanetworkopen.2022.43146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
IMPORTANCE Kidney functional capacity is low at birth but doubles during the first 2 weeks of life and reaches near-adult levels at age 1 to 2 years. Existing reference intervals for markers of kidney function in newborns are mostly based on preterm newborns, newborns with illness, or small cohorts of term newborns, and the consequences of maternal comorbidities for newborn kidney function are sparsely described. OBJECTIVE To establish robust reference intervals for creatinine and urea in healthy children in early childhood and to assess whether maternal comorbidity is associated with newborn creatinine and urea concentrations. DESIGN, SETTING, AND PARTICIPANTS This multicenter, prospective, population-based cohort study assessed data and umbilical cord blood samples from participants in the Copenhagen Baby Heart Study (CBHS) who were born between April 1, 2016, and October 31, 2018, and venous blood samples from a subsample of CBHS participants who were enrolled in the COMPARE study between May 3, 2017, and November 4, 2018. Cord blood samples of 13 354 newborns from the CBHS and corresponding venous blood samples of 444 of those newborns from the COMPARE study were included. Blood samples were collected at birth, age 2 months, and age 14 to 16 months, with follow-up completed on February 12, 2020. Healthy nonadmitted term newborns from maternity wards at 3 hospitals in the Capital Region of Denmark were included. EXPOSURES Maternal comorbidity. MAIN OUTCOMES AND MEASURES Creatinine and urea concentrations. RESULTS Among 13 354 newborns in the CBHS cohort, characteristics of 12 938 children were stratified by sex and gestational age (GA). Of those, 6567 children (50.8%) were male; 5259 children (40.6%) were born at 37 to 39 weeks' GA, and 7679 children (59.4%) were born at 40 to 42 weeks' GA. Compared with children born at 40 to 42 weeks' GA, those born at 37 to 39 weeks' GA had lower birth weight, Apgar scores at 5 minutes, placental weight, and placental-fetal weight ratio. Children born at 37 to 39 weeks' GA vs those born at 40 to 42 weeks' GA were more frequently small for GA at birth and more likely to have placental insufficiency and exposure to maternal preeclampsia, maternal diabetes, maternal kidney disease, and maternal hypertension. Among children born at 37 to 39 weeks' GA, reference intervals were 0.54 to 1.08 mg/dL for creatinine and 5.32 to 14.67 mg/dL for urea; among children born at 40 to 42 weeks' GA, reference intervals were 0.57 to 1.19 mg/dL for creatinine and 5.60 to 14.85 mg/dL for urea. At birth, multifactorially adjusted odds ratios among children exposed to preeclampsia were 9.40 (95% CI, 1.68-52.54) for a venous creatinine concentration higher than the upper reference limit, 4.29 (95% CI, 1.32-13.93) for a venous creatinine concentration higher than the 90th percentile, and 3.10 (95% CI, 1.14-8.46) for a venous creatinine concentration higher than the 80th percentile. CONCLUSIONS AND RELEVANCE In this study, improved reference intervals for creatinine and urea concentrations were generated. Preeclampsia was associated with an increased risk of high newborn creatinine concentrations, suggesting that newborns of mothers with preeclampsia need closer observation of their kidney function.
Collapse
Affiliation(s)
- Rikke Mohr Lytsen
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Sofie Taageby Nielsen
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Malene Kongsgaard Hansen
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - Nina Strandkjær
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - Ida Juul Rasmussen
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Anna Axelsson Raja
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - R. Ottilia Vøgg
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - Anne-Sophie Sillesen
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - Pia R. Kamstrup
- Department of Clinical Biochemistry, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
| | - Ida Maria Schmidt
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Copenhagen University Hospital–Herlev-Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Laeer S, Cawello W, Burckhardt BB, Ablonczy L, Bajcetic M, Breur JMPJ, Dalinghaus M, Male C, de Wildt SN, Breitkreutz J, Faisal M, Keatley-Clarke A, Klingmann I, Lagler FB. Enalapril and Enalaprilat Pharmacokinetics in Children with Heart Failure Due to Dilated Cardiomyopathy and Congestive Heart Failure after Administration of an Orodispersible Enalapril Minitablet (LENA-Studies). Pharmaceutics 2022; 14:pharmaceutics14061163. [PMID: 35745735 PMCID: PMC9228797 DOI: 10.3390/pharmaceutics14061163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/29/2022] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACEI), such as enalapril, are a cornerstone of treatment for pediatric heart failure which is still used off-label. Using a novel age-appropriate formulation of enalapril orodispersible minitablets (ODMTs), phase II/III open-label, multicenter pharmacokinetic (PK) bridging studies were performed in pediatric patients with heart failure due to dilated cardiomyopathy (DCM) and congenital heart disease (CHD) in five participating European countries. Children were treated for 8 weeks with ODMTs according to an age-appropriate dosing schedule. The primary objective was to describe PK parameters (area under the curve (AUC), maximal concentration (Cmax), time to reach maximal concentration (t-max)) of enalapril and its active metabolite enalaprilat. Of 102 patients, 89 patients (n = 26, DCM; n = 63 CHD) were included in the primary PK endpoint analysis. Rate and extent of enalapril and its active metabolite enalaprilat were described and etiology and age could be identified as potential PK modifying factors. The dosing schedule appeared to be tolerated well and did not result in any significant drug-related serious adverse events. The PK analysis and the lack of severe safety events supports the applied age-appropriate dosing schedule for the enalapril ODMTs.
Collapse
Affiliation(s)
- Stephanie Laeer
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
- Correspondence: ; Tel.: +49-211-8110740
| | - Willi Cawello
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | - Bjoern B. Burckhardt
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | - László Ablonczy
- Goettsegen György Hungarian Institute of Cardiology (HPHC), 1450 Budapest, Hungary;
| | - Milica Bajcetic
- Univerzitetska Dečja Klinika (UDK), University Children Hospital, School of Medicine, University of Belgrade, 11129 Belgrade, Serbia;
| | - Johannes M. P. J. Breur
- University Medical Center Utrecht, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands;
| | - Michiel Dalinghaus
- Division of Pediatric Cardiology, Erasmus MC Sophia Children’s Hospital, 3000 CA Rotterdam, The Netherlands;
| | - Christoph Male
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Saskia N. de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, 3015 GJ Rotterdam, The Netherlands;
- Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | | | - Muhammed Faisal
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine-Universitaet Düsseldorf, 40225 Duesseldorf, Germany; (W.C.); (B.B.B.); (M.F.)
| | | | | | | |
Collapse
|
28
|
Sun ZM, Zhou N, Peng XX, Wang H, Shen Y. Insufficient application of estimation equations of glomerular filtration rate: a survey of 1009 Chinese pediatricians. World J Pediatr 2022; 18:368-372. [PMID: 35267183 DOI: 10.1007/s12519-021-00509-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Zi-Mo Sun
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Nan Zhou
- Department of Nephrology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xiao-Xia Peng
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Hui Wang
- Department of Nephrology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Ying Shen
- Department of Nephrology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| |
Collapse
|
29
|
Doctor P, Aggarwal S, Garcia R. Is there an association of near-infrared spectroscopy with low cardiac output and adverse outcomes in single-ventricle patients after stage 1 palliation? Ann Pediatr Cardiol 2022; 15:249-256. [PMID: 36589644 PMCID: PMC9802614 DOI: 10.4103/apc.apc_234_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/22/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Our primary objective study was to evaluate the association between near-infrared spectroscopy (NIRS) and low cardiac output (LCO) in patients with single-ventricle physiology after stage 1 palliation. Methods In this retrospective study, infants ≤6 months of age with single-ventricle physiology who underwent stage 1 palliation were included. Cerebral and renal NIRS values at various time intervals after surgery were compared between patients with low and normal cardiac output. LCO within the first 48 after surgery was defined as per the pediatric cardiac critical care consortium database. NIRS values were also compared with other adverse outcomes such as cardiac arrest, need for extracorporeal membrane oxygenation and mortality. The receiver operative characteristic curve was generated to determine an optimal cut-off NIRS value for detecting LCO. Results Ninety-one patients with median (Interquartile range) age of 10 days (6-26) and weight of 3.3 kg (3-3.5) were included in the study. Cerebral NIRS at 1 h (41.2 vs. 49.5; P = 0.002), 6 h (44 vs. 52.2; P = 0.001), and 12 h (51.8 vs. 56; P = 0.025) was significantly lower in the grouP with LCO compared to no LCO. Cerebral NIRS at 6 h was independently associated with LCO (P = 0.018), and cerebral NIRS at 6 h ≤57% had 91% sensitivity and 72% specificity to detect LCO. Conclusions Cerebral NIRS ≤57% at 6 h after surgery detected LCO after stage 1 palliation in single-ventricle patients. Cerebral or renal NIRS was not associated with adverse outcomes and therefore, may not be useful in predicting adverse outcomes in this population.
Collapse
Affiliation(s)
- Pezad Doctor
- Department of Pediatrics, Division of Cardiology, Children's Medical Center, University of Texas Southwestern Medical Center, Medical District, Dallas, Texas, USA
| | - Sanjeev Aggarwal
- Department of Pediatrics, Division of Cardiology, Children's Hospital of Michigan, Central Michigan University College of Medicine, Beaubien Blvd, Detroit, MI, USA
| | - Richard Garcia
- Department of Pediatrics, Division of Critical Care Medicine, Children's Hospital of Michigan, Central Michigan University College of Medicine, Beaubien Blvd, Detroit, MI, USA
| |
Collapse
|
30
|
Smeets N, IntHout J, van der Burgh M, Schwartz G, Schreuder M, de Wildt S. Maturation of Glomerular Filtration Rate in Term-Born Neonates: An Individual Participant Data Meta-Analysis. J Am Soc Nephrol 2022; 33:1277-1292. [PMID: 35474022 PMCID: PMC9257816 DOI: 10.1681/asn.2021101326] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/28/2022] [Indexed: 11/03/2022] Open
Abstract
Background: The evidence from individual studies to support the maturational pattern of glomerular filtration rate (GFR) in healthy term-born neonates is inconclusive. We performed an individual participant data (IPD) meta-analysis of reported measured GFR (mGFR) data aimed to establish neonatal GFR reference values. Furthermore, we aimed to optimise neonatal creatinine-based GFR estimations Methods: We identified studies reporting mGFR measured by exogenous markers or creatinine clearance (CrCL) in healthy term-born neonates. The relationship between postnatal age and clearance was investigated using cubic splines with generalized additive linear mixed models. From our reference values, we estimated an updated coefficient for the Schwartz equation (eGFR(ml/min/1.73m2)=(k*height (cm))/serum creatinine(mg/dl)). Results: Forty-eight out of 1521 screened articles reported mGFR in healthy term-born neonates, and 978 mGFR values from 881 neonates were analysed. IPD were available for 367 neonates and the other 514 neonates were represented by 41 aggregated data points as means/medians per group. GFR doubled in the first five days after birth from 19.6 (95%CI 14.7;24.6) ml/min/1.73m2 to 40.6 (95%CI 36.7;44.5) ml/min/1.73m2, then more gradually increased to 59.4 (95%CI 45.9;72.9) ml/min/1.73m2 by four weeks of age. A coefficient of 0.31 to estimate GFR best fitted the data. Conclusions: These reference values for healthy term-born neonates show a biphasic increase in GFR with the largest increase between days 1 and 5. Together with the re-examined Schwartz equation, this can help identify altered GFR in term-born neonates. To enable widespread implementation of our proposed eGFR equation, validation in a large cohort of neonates is required.
Collapse
Affiliation(s)
- Nori Smeets
- N Smeets, Department of Pharmacology and Toxicology, Radboudumc Radboud Institute for Health Sciences, Nijmegen, Netherlands
| | - Joanna IntHout
- J IntHout, Department for Health Evidence, Section Biostatistics, Radboudumc, Nijmegen, Netherlands
| | - Maurice van der Burgh
- M van der Burgh, Department of Pharmacology and Toxicology, Radboudumc Radboud Institute for Health Sciences, Nijmegen, Netherlands
| | - George Schwartz
- G Schwartz, Department of Pediatrics, Pediatric Nephrology, University of Rochester Medical Center, Rochester, United States
| | - Michiel Schreuder
- M Schreuder, Department of Pediatrics, division of Pediatric Nephrology, Radboudumc, Nijmegen, Netherlands
| | - Saskia de Wildt
- S de Wildt, Department of Pharmacology and Toxicology, Radboudumc Radboud Institute for Health Sciences, Nijmegen, Netherlands
| |
Collapse
|
31
|
Blood pressure variability during pediatric cardiac surgery is associated with acute kidney injury. Pediatr Nephrol 2022; 37:871-879. [PMID: 34436673 DOI: 10.1007/s00467-021-05234-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Blood pressure variability (BPV), defined as the degree of variation between discrete blood pressure readings, is associated with poor outcomes in acute care settings. Acute kidney injury (AKI) is a common and serious postoperative complication of cardiac surgery with cardiopulmonary bypass (CPB) in children. No studies have yet assessed the association between intraoperative BPV during cardiac surgery with CPB and the development of AKI in children. METHODS A retrospective chart review of children undergoing cardiac surgery with CPB was performed. Intraoperative BPV was calculated using average real variability (ARV) and standard deviation (SD). Multiple regression models were used to examine the association between BPV and outcomes of AKI, hospital and intensive care unit (PICU) length of stay, and length of mechanical ventilation. RESULTS Among 231 patients (58% males, median age 8.6 months) reviewed, 51.5% developed AKI (47.9% Stage I, 41.2% Stage II, 10.9% Stage III). In adjusted models, systolic and diastolic ARV were associated with development of any stage AKI (OR 1.40, 95% CI 1.08-1.8 and OR 1.4, 95% CI 1.05-1.8, respectively). Greater diastolic SD was associated with longer PICU length of stay (β 0.94, 95% CI 0.62-1.2). When stratified by age, greater systolic ARV and SD were associated with AKI in infants ≤ 12 months, but there was no relationship in children > 12 months. CONCLUSIONS Greater BPV during cardiac surgery with CPB was associated with development of postoperative AKI in infants, suggesting that BPV is a potentially modifiable risk factor for AKI in this high-risk population.
Collapse
|
32
|
Algaze CA, Margetson TD, Sutherland SM, Kwiatkowski DM, Maeda K, Navaratnam M, Samreth SP, Price EP, Zook NB, Yang JK, Hollander SA. Impact of a clinical pathway on acute kidney injury in patients undergoing heart transplant. Pediatr Transplant 2022; 26:e14166. [PMID: 34727417 DOI: 10.1111/petr.14166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND To evaluate the impact of a clinical pathway on the incidence and severity of acute kidney injury in patients undergoing heart transplant. METHODS This was a 2.5-year retrospective evaluation using 3 years of historical controls within a cardiac intensive care unit in an academic children's hospital. Patients undergoing heart transplant between May 27, 2014, and April 5, 2017 (pre-pathway) and May 1, 2017, and November 30, 2019 (pathway) were included. The clinical pathway focused on supporting renal perfusion through hemodynamic management, avoiding or delaying nephrotoxic medications, and providing pharmacoprophylaxis against AKI. RESULTS There were 57 consecutive patients included. There was an unadjusted 20% reduction in incidence of any acute kidney injury (p = .05) and a 17% reduction in Stage 2/3 acute kidney injury (p = .09). In multivariable adjusted analysis, avoidance of Stage 2/3 acute kidney injury was independently associated with the clinical pathway era (AOR -1.3 [95% CI -2.5 to -0.2]; p = .03), achieving a central venous pressure of or less than 12 mmHg (AOR -1.3 [95% CI -2.4 to -0.2]; p = .03) and mean arterial pressure above 60 mmHg (AOR -1.6 [95% CI -3.1 to -0.01]; p = .05) in the first 48 h post-transplant, and older age at transplant (AOR - 0.2 [95% CI -0.2 to -0.06]; p = .002). CONCLUSIONS This report describes a renal protection clinical pathway associated with a reduction in perioperative acute kidney injury in patients undergoing heart transplant and highlights the importance of normalizing perioperative central venous pressure and mean arterial blood pressure to support optimal renal perfusion.
Collapse
Affiliation(s)
- Claudia A Algaze
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA.,Center for Pediatric and Maternal Value, Stanford University School of Medicine, Palo Alto, California, USA
| | - Tristan D Margetson
- Department of Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Scott M Sutherland
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - David M Kwiatkowski
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Katsuhide Maeda
- Department of Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Manchula Navaratnam
- Department of Anesthesia, Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, California, USA
| | - Sarah P Samreth
- Center for Pediatric and Maternal Value, Stanford University School of Medicine, Palo Alto, California, USA
| | - Elizabeth P Price
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Nina B Zook
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jeffrey K Yang
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Seth A Hollander
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
33
|
Ozcanoglu HD, Öztürk E, Tanıdır İC, Şahin GT, Ozalp S, Yıldız O, Özcan FG, Hatemi A. The comparison of three different acute kidney injury classification systems after congenital heart surgery. Pediatr Int 2022; 64:e15270. [PMID: 36239168 DOI: 10.1111/ped.15270] [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] [Received: 11/10/2021] [Revised: 04/09/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND We aimed to compare the frequency of acute kidney injury (AKI) and its effects on mortality and morbidity with different classification systems in pediatric patients who had surgery under cardiopulmonary bypass for congenital heart disease. METHODS This study included children younger than 18 years old who were followed up in the pediatric cardiac intensive care unit between September 1 and December 1, 2020, after congenital heart surgery with cardiopulmonary bypass. Each case was categorized postoperatively in terms of AKI using Pediatric-Modified Risk, Injury, Failure, Loss, and End-Stage (pRIFLE), Acute Kidney Injury Network (AKIN), and Kidney Disease: Improving Global Outcomes (KDIGO). Hospital mortality (developed within the first 30 days postoperatively) and morbidity (longer than 7 days intensive care unit stay) were compared by three model classes. Results were evaluated statistically. RESULTS One hundred patients were included in the study. The median age was 3 months (1 day-180 months). Acute kidney injury was diagnosed in 49% of the cases according to the pRIFLE classification. It was diagnosed in 31% of the patients by AKIN classification. It was diagnosed in 41% of the patients with the KDIGO criteria. Morbidity was observed in 25% (n = 25) of all cases. The morbidity predictor was 0.800 for pRIFLE, 0.747 for AKIN and 0.853 for KDIGO by receiver operating characteristics analysis. All three categories predicted morbidity significantly (P < 0.001). Mortality was 10% (n = 10) for all groups. The mortality predictor was 0.783 for pRIFLE, 0.717 for AKIN and 0.794 for KDIGO by receiver operating characteristics analysis, and all three categories predicted mortality significantly (P < 0.001). CONCLUSIONS Regardless of the three methods used, AKI was commonly detected in pediatric patients undergoing congenital heart surgery. pRIFLE classification diagnosed more patients with AKI than AKIN and KDIGO. The KDIGO and pRIFLE classifications were better in predicting hospital mortality.
Collapse
Affiliation(s)
- Hatice Dilek Ozcanoglu
- Department of Anaesthesiology and Reanimation, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Erkut Öztürk
- Department of Pediatric Cardiology, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - İbrahim Cansaran Tanıdır
- Department of Pediatric Cardiology, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Gulhan Tunca Şahin
- Department of Pediatric Cardiology, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Serife Ozalp
- Department of Anaesthesiology and Reanimation, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Okan Yıldız
- Department of Cardiovascular Surgery, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Funda Gümüş Özcan
- Department of Anaesthesiology and Reanimation, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| | - Alican Hatemi
- Department of Cardiovascular Surgery, Istanbul Saglik Bilimleri University Basaksehir Cam and Sakura Hospital, Istanbul, Turkey
| |
Collapse
|
34
|
Chen CY, Montez-Rath ME, May LJ, Maeda K, Hollander SA, Rosenthal DN, Krawczeski CD, Sutherland SM. Hemodynamic Predictors of Renal Function After Pediatric Left Ventricular Assist Device Implantation. ASAIO J 2021; 67:1335-1341. [PMID: 34860188 PMCID: PMC8647769 DOI: 10.1097/mat.0000000000001460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Although renal function often improves after pediatric left ventricular assist device (LVAD) implantation, recovery is inconsistent. We aimed to identify hemodynamic parameters associated with improved renal function after pediatric LVAD placement. A single-center retrospective cohort study was conducted in patients less than 21 years who underwent LVAD placement between June 2004 and December 2015. The relationship between hemodynamic parameters and estimated glomerular filtration rate (eGFR) was assessed using univariate and multivariate modeling. Among 54 patients, higher preoperative central venous pressure (CVP) was associated with eGFR improvement after implantation (p = 0.012). However, 48 hours postimplantation, an increase in CVP from baseline was associated with eGFR decline over time (p = 0.01). In subgroup analysis, these associations were significant only for those with normal pre-ventricular assist device renal function (p = 0.026). In patients with preexisting renal dysfunction, higher absolute CVP values 48 and 72 hours after implantation predicted better renal outcome (p = 0.005). Our results illustrate a complex relationship between ventricular function, volume status, and renal function. Additionally, they highlight the challenge of using CVP to guide management of renal dysfunction in pediatric heart failure. Better methods for evaluating right heart function and volume status are needed to improve our understanding of how hemodynamics impact renal function in this population.
Collapse
Affiliation(s)
- Chiu-Yu Chen
- From the Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - Maria E Montez-Rath
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Lindsay J May
- Division of Pediatric Cardiology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Katsuhide Maeda
- Department of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Seth A Hollander
- From the Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - David N Rosenthal
- From the Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - Catherine D Krawczeski
- Division of Pediatric Cardiology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - Scott M Sutherland
- Division of Pediatric Nephrology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
35
|
Outcomes of solitary functioning kidneys-renal agenesis is different than multicystic dysplastic kidney disease. Pediatr Nephrol 2021; 36:3673-3680. [PMID: 33954810 DOI: 10.1007/s00467-021-05064-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Multicystic dysplastic kidney (MCDK) disease and unilateral renal agenesis (URA) are well-known causes of a solitary functioning kidney (SFK) and are associated with long-term kidney injury. The aims of this study were to characterize the natural history of SFK at our center, define the risk factors associated with chronic kidney injury, and identify distinguishing features between URA and MCDK that predict outcome. METHODS This was a retrospective cohort study of 230 SFK patients. We compared MCDK (n=160) and URA (n=70) according to clinical features at diagnosis and kidney outcomes over follow-up. Univariate and multivariate binary regression analysis was used to determine independent risk factors for chronic kidney injury, defined as the composite outcome of hypertension, proteinuria, or chronic kidney disease (eGFR <60 mL/min/1.73m2). RESULTS URA had a higher prevalence of comorbid genetic syndromes (15 vs. 6%, p=0.04), non-renal anomalies (39 vs. 11%, p<0.001), and congenital anomalies of the kidney and urinary tract (CAKUT) (51 vs. 26%, p<0.001) than MCDK. Over follow-up, URA experienced more hypertension (19 vs. 3%, p=0.002), proteinuria (12 vs. 3%, p=0.03), and the composite outcome (19 vs. 6%, p=0.003) than MCDK. Independent risk factors for chronic kidney injury included CAKUT (OR 5.01, p=0.002) and URA (OR 2.71, p=0.04). CONCLUSIONS In our population, URA was more likely to have associated syndromes or anomalies, and to have worse outcomes over time than MCDK. URA diagnosis was an independent risk factor for chronic kidney injury. Our results will be used to develop a standardized clinical pathway for SFK management. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
|
36
|
Lu H, Thurnherr E, Meaney CJ, Fusco NM. Incidence and Risk Factors for Acute Kidney Injury in Hospitalized Children Receiving Piperacillin-Tazobactam. J Pediatr Pharmacol Ther 2021; 26:597-602. [PMID: 34421409 DOI: 10.5863/1551-6776-26.6.597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/30/2020] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Drug-induced kidney injury contributes to morbidity and mortality in hospitalized children. Antibiotics such as TZP have been implicated in the development of acute kidney injury (AKI) in adults; however, data are limited in children. The purpose of this study was to determine the incidence of AKI in hospitalized children receiving TZP. METHODS This was a retrospective cohort study of hospitalized children between 2 months and 19 years of age who received TZP for at least 48 hours. Acute kidney injury was defined as a 50% increase from the initial serum creatinine (SCr) prior to TZP initiation. Serum creatinine values were adjusted for fluid balance using a validated approach. Severity of AKI was characterized using the Pediatric Risk, Injury, Failure, Loss, End-Stage Renal Disease (pRIFLE) criteria. Descriptive and inferential statistics were used to describe the incidence and risk factors of AKI, with an alpha = 0.05. RESULTS A total of 65 subjects were included. Twenty-five (38.5%) required PICU admission. The incidence of AKI was 7.7% (n = 5) using adjusted SCr (13.37 cases/1000 patient-days). According to pRIFLE, 6.15% (n = 4) subjects met criteria for Risk (n = 3) or Injury (n = 1), and none developed Failure, Loss, or End-Stage (10.70 cases/1000 patient-days for Risk and Injury categories). No risk factors were identified. Hospital length of stay was longer in subjects who experienced AKI compared with those who did not (p = 0.04). CONCLUSIONS The incidence of AKI in hospitalized children exposed to TZP was low. In those who did develop AKI, peak SCr occurred approximately 1 week after TZP initiation.
Collapse
|
37
|
Mishra OP, Verma AK, Abhinay A, Singh A, Singh A, Prasad R. Risk factors for mortality in critically ill infants with acute kidney injury: A resource-limited setting experience. Ther Apher Dial 2021; 26:297-305. [PMID: 34296516 DOI: 10.1111/1744-9987.13712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 11/28/2022]
Abstract
Infants with acute kidney injury (AKI) who are critically ill often will have multiorgan dysfunctions. Objective of the present study was to find out mortality, recovery of kidney function at discharge and at 3 months, and to determine risk factors for mortality. Fifty-two infants (24 newborns and 28 postneonatal) with AKI were included. Staging was done as per Kidney Disease Improving Global Outcomes classification. Patients were subjected to medical treatment and peritoneal dialysis (PD), wherever indicated. Kidney function tests were performed at admission, discharge, and at 3 months follow-up. Median age of neonates was 8 days and postneonatal infants were 4.5 months. Stage 1, 2, and 3 AKI were present in 14 (26.9%), 16 (30.7%), and 22 (42.3%) cases, respectively. PD was required in 22 (42.3%) infants, and significantly higher in postneonatal than in neonates (57.1% vs. 25%, p < 0.05). Significant recovery of kidney function occurred at discharge and cases had normal parameters at 3 months. Mortality was 17.3%. Patients had significantly higher risk of mortality, if they had metabolic acidosis (OR 13.22, CI 2.33-74.94, p = 0.002) and needed ventilation (OR 14.93, 95% CI 1.7-130.97, p = 0.006) and PD (OR 6.53, 95% CI 1.20-35.48, p = 0.026). In logistic regression analysis, fluid overload (p < 001), hypotension (p < 0.01), and higher PRISM-III score (p < 0.05) were found as significant risk factors for mortality. Medical management including PD led to good recovery of kidney function. Presence of fluid overload, hypotension, and higher PRISM-III score adversely affected the outcome.
Collapse
Affiliation(s)
- Om P Mishra
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Avdhesh Kumar Verma
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Abhishek Abhinay
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Akanksha Singh
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ankur Singh
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajniti Prasad
- Division of Pediatric Nephrology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
38
|
Iacobelli S, Guignard JP. Maturation of glomerular filtration rate in neonates and infants: an overview. Pediatr Nephrol 2021; 36:1439-1446. [PMID: 32529323 DOI: 10.1007/s00467-020-04632-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/16/2020] [Accepted: 05/27/2020] [Indexed: 11/25/2022]
Abstract
Glomerular filtration rate (GFR) increases progressively throughout fetal life, matures rapidly after birth according to gestational and post-menstrual age, and reaches adult values by 1-year post-natal age. GFR is considered the best marker of kidney function, and in clinical practice, estimated GFR is useful to anticipate complications, establish prognosis, and facilitate treatment decisions. This review article summarizes the maturation of glomerular filtration and the factors and conditions that modulate and impair developing glomerular filtration, and discusses the techniques available to assess GFR in neonates and infants. We focused on simple, reliable, easily available, and cheap techniques to estimate GFR, which may provide valuable information on the renal aspects of the clinical care of this group of patients.
Collapse
Affiliation(s)
- Silvia Iacobelli
- Néonatologie, Réanimation Néonatale et Pédiatrique, CHU La Réunion, Site Sud, Saint Pierre, France. .,Centre d'Etudes Périnatales de l'Océan Indien, EA 7388, CHU La Réunion, Site Sud, Saint Pierre et Université de la Réunion, Réunion, France.
| | | |
Collapse
|
39
|
Chapron BD, Chapron A, Leeder JS. Recent advances in the ontogeny of drug disposition. Br J Clin Pharmacol 2021; 88:4267-4284. [PMID: 33733546 DOI: 10.1111/bcp.14821] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Developmental changes that occur throughout childhood have long been known to impact drug disposition. However, pharmacokinetic studies in the paediatric population have historically been limited due to ethical concerns arising from incorporating children into clinical trials. As such, much of the early work in the field of developmental pharmacology was reliant on difficult-to-interpret in vitro and in vivo animal studies. Over the last 2 decades, our understanding of the mechanistic processes underlying age-related changes in drug disposition has advanced considerably. Progress has largely been driven by technological advances in mass spectrometry-based methods for quantifying proteins implicated in drug disposition, and in silico tools that leverage these data to predict age-related changes in pharmacokinetics. This review summarizes our current understanding of the impact of childhood development on drug disposition, particularly focusing on research of the past 20 years, but also highlighting select examples of earlier foundational research. Equally important to the studies reviewed herein are the areas that we cannot currently describe due to the lack of research evidence; these gaps provide a map of drug disposition pathways for which developmental trends still need to be characterized.
Collapse
Affiliation(s)
- Brian D Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Alenka Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA.,Schools of Medicine and Pharmacy, University of Missouri-Kansas City, MO, USA
| |
Collapse
|
40
|
Lee SM, Yang S, Kang S, Chang MJ. Population pharmacokinetics and dose optimization of vancomycin in neonates. Sci Rep 2021; 11:6168. [PMID: 33731764 PMCID: PMC7969932 DOI: 10.1038/s41598-021-85529-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/26/2021] [Indexed: 01/12/2023] Open
Abstract
The pharmacokinetics of vancomycin vary among neonates, and we aimed to conduct population pharmacokinetic analysis to determine the optimal dosage of vancomycin in Korean neonates. From a retrospective chart review, neonates treated with vancomycin from 2008 to 2017 in a neonatal intensive care unit (NICU) were included. Vancomycin concentrations were collected based on therapeutic drug monitoring, and other patient characteristics were gathered through electronic medical records. We applied nonlinear mixed-effect modeling to build the population pharmacokinetic model. One- and two-compartment models with first-order elimination were evaluated as potential structural pharmacokinetic models. Allometric and isometric scaling was applied to standardize pharmacokinetic parameters for clearance and volume of distribution, respectively, using fixed powers (0.75 and 1, respectively, for clearance and volume). The predictive performance of the final model was developed, and dosing strategies were explored using Monte Carlo simulations with AUC0–24 targets 400–600. The patient cohort included 207 neonates, and 900 vancomycin concentrations were analyzed. Only 37.4% of the analyzed concentrations were within trough concentrations 5–15 µg/mL. A one-compartment model with first-order elimination best described the vancomycin pharmacokinetics in neonates. Postmenstrual age (PMA) and creatinine clearance (CLcr) affected the clearance of vancomycin, and model evaluation confirmed the robustness of the final model. Population pharmacokinetic modeling and dose optimization of vancomycin in Korean neonates showed that vancomycin clearance was related to PMA and CLcr, as well as body weight. A higher dosage regimen than the typical recommendation is suggested.
Collapse
Affiliation(s)
- Soon Min Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Seungwon Yang
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Soyoung Kang
- Department of Pharmaceutical Medicine and Regulatory Science, Yonsei University, Veritas Hall D #214, Yonsei University International Campus, Songdogwahak-ro 85, Yeonsu-gu, Incheon, Korea
| | - Min Jung Chang
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea. .,Department of Pharmaceutical Medicine and Regulatory Science, Yonsei University, Veritas Hall D #214, Yonsei University International Campus, Songdogwahak-ro 85, Yeonsu-gu, Incheon, Korea.
| |
Collapse
|
41
|
Liu YX, Wen H, Niu WJ, Li JJ, Li ZL, Jiao Z. External Evaluation of Vancomycin Population Pharmacokinetic Models at Two Clinical Centers. Front Pharmacol 2021; 12:623907. [PMID: 33897418 PMCID: PMC8058705 DOI: 10.3389/fphar.2021.623907] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/15/2021] [Indexed: 01/19/2023] Open
Abstract
Background: Numerous vancomycin population pharmacokinetic models in neonates have been published; however, their predictive performances remain unknown. This study aims to evaluate their external predictability and explore the factors that might affect model performance. Methods: Published population pharmacokinetic models in neonates were identified from the literature and evaluated using datasets from two clinical centers, including 171 neonates with a total of 319 measurements of vancomycin levels. Predictive performance was assessed by prediction- and simulation-based diagnostics and Bayesian forecasting. Furthermore, the effect of model structure and a number of identified covariates was also investigated. Results: Eighteen published pharmacokinetic models of vancomycin were identified after a systematic literature search. Using prediction-based diagnostics, no model had a median prediction error of ≤ ± 15%, a median absolute prediction error of ≤30%, and a percentage of prediction error that fell within ±30% of >50%. A simulation-based visual predictive check of most models showed there were large deviations between observations and simulations. After Bayesian forecasting with one or two prior observations, the predicted performance improved significantly. Weight, age, and serum creatinine were identified as the most important covariates. Moreover, employing a maturation model based on weight and age as well as nonlinear model to incorporate serum creatinine level significantly improved predictive performance. Conclusion: The predictability of the pharmacokinetic models for vancomycin is closely related to the approach used for modeling covariates. Bayesian forecasting can significantly improve the predictive performance of models.
Collapse
Affiliation(s)
- Yi-Xi Liu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haini Wen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wan-Jie Niu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing-Jing Li
- Department of Pharmacy, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Incidence and impact of acute kidney injury in patients with hypoplastic left heart syndrome following the hybrid stage 1 palliation. Cardiol Young 2021; 31:414-420. [PMID: 33261689 DOI: 10.1017/s1047951120004199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Acute kidney injury leads to worse outcomes following paediatric cardiac surgery. There is a lack of literature focusing on acute kidney injury after the Hybrid stage 1 palliation for single ventricle physiology. Patients undergoing the Hybrid Stage 1, as a primary option, may have a lower incidence of kidney injury than previously reported. When present, kidney injury may increase the risk of post-operative morbidity and mortality. METHODS A retrospective, single centre review was conducted in patients with hypoplastic left heart syndrome who underwent Hybrid Stage 1 from 2008 to 2018. Acute kidney injury was defined as a dichotomous yes (meeting any injury criteria) or no (no injury) utilising two different criteria utilised in paediatrics. The impact of kidney injury on perioperative characteristics and 30-day mortality was analysed. RESULTS The incidence of acute kidney injury is 13.4-20.7%, with a severe injury rate of 2.4%. Patients without a prenatal diagnosis of hypoplastic left heart syndrome have a higher incidence of kidney injury than those prenatally diagnosed, (40% versus 14.5%, p = 0.024). Patients with acute kidney injury have a significantly higher incidence of 30-day mortality, 27.3%, compared to without, 5.6% (p = 0.047). DISCUSSION The incidence of severe acute kidney injury after the Hybrid Stage 1 palliation is low. A prenatal diagnosis may be associated with a lower incidence of kidney injury following the Hybrid Stage 1. Though uncommon, severe acute kidney injury following Hybrid Stage 1 may be associated with higher 30-day mortality.
Collapse
|
43
|
Matsuoka D, Hirabayashi K, Murase T, Saito S, Hidaka Y, Nakazawa Y. Assessment of kidney function using inulin-based and estimated glomerular filtration rates before and after allogeneic hematopoietic stem cell transplantation in pediatric patients. Pediatr Blood Cancer 2020; 67:e28733. [PMID: 33001557 DOI: 10.1002/pbc.28733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/10/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Accurate evaluation of kidney function before and after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is important for both informed decision making and detection of chronic kidney disease. However, to the best of our knowledge, no report has evaluated the glomerular filtration rate (GFR) in pediatric patients who underwent HSCT using the gold standard GFR measurement, as well as inulin-based GFR (iGFR). METHODS We assessed iGFR before and after allo-HSCT to evaluate the impact of allo-HSCT on GFR in a prospective cohort study of 17 pediatric patients. We also assessed the accuracy and bias of the values of estimated GFR (eGFR) calculated using serum creatinine (Cr), cystatin C (CysC), beta-2 microglobulin (β2 MG), 24-h creatinine clearance (24hCcr), and the full chronic kidney disease in children (CKiD) index that combines Cr, CysC, and blood urea nitrogen-based equations with iGFR as a reference to identify the most reliable equation for GFR. RESULTS There was no significant difference between the values before and after allo-HSCT. CKiD CysC-, 24hCcr-, and full CKiD-based values showed good within 30% (P30) accuracy (80.6%, 79.3%, and 80.6%, respectively), but only 24hCcr and full CKiD had good mean bias (8.5% and 8.9%, respectively) and narrow 95% limits of agreement (-32.2 to 52.7 mL/min/1.73 m2 and -29.3 to 47.4 mL/min/1.73 m2 , respectively) compared with the corresponding iGFR. CONCLUSION There was no significant impact of allo-HSCT on GFR in our cohort. The most reliable equations for pediatric patients with allo-HSCT were eGFR-24hCcr and eGFR-full CKiD.
Collapse
Affiliation(s)
- Daisuke Matsuoka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Koichi Hirabayashi
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tsubasa Murase
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shoji Saito
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshihiko Hidaka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
44
|
Çetin B, Dönmez Mİ, Erdem S, Ziylan O, Oktar T. Renal, Bladder and Sexual Outcomes in Adult Men with History of Posterior Urethral Valves Treated in Childhood. Urology 2020; 153:301-306. [PMID: 33188791 DOI: 10.1016/j.urology.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To determine the adulthood outcomes of bladder, kidney, and sexual functions of posterior urethral valve (PUV) patients. MATERIALS AND METHODS The records of patients who were treated for PUV between 1980, and 2001 and aged ≥18 years by the end of 2019 were retrospectively reviewed. Patients with complete adulthood data were included in the study. Renal, bladder, and sexual functions were assessed. Adulthood uroflowmetry results of 22 patients were also evaluated. RESULTS Thirty-nine patients with complete adulthood data out of 89 were included. The median follow-up time was 22.7 years (15-33 years). Median age at initial surgery for PUV was 36 months (1-168 months), and median age at last follow-up was 26 years (18-46 years). Lower urinary tract dysfunction was noted in 15 (38%) patients. In 22 patients (56%) with uroflowmetric analyses, median values of Qmax, voided volume, and post voiding residual urine volume were 20.5 mL/sec (7-43 mL/sec), 389 mL (154-1750 mL), and 18.5 mL (range 0-190 mL), respectively. Nineteen patients (48.7%) had normal glomerular filtration rate, 2 patients (5.1%) had chronic kidney disease, and the remaining 18 patients (46.1%) had end stage renal disease, 14 (35.9%) of whom underwent renal transplantation. Thirty-two patients (82%) had sexual function evaluation, none of which reported erectile dysfunction. However, 4 patients (12.5%) reported slow ejaculation. Five patients fathered a child whereas infertility was observed in 2 patients. CONCLUSION After 2 decades, glomerular filtration rate was normal almost in half of the PUV patients while lower urinary tract dysfunction was detected in 38%. Furthermore, erectile dysfunction was rare.
Collapse
Affiliation(s)
- Bilal Çetin
- Department of Urology, Division of Pediatric Urology, İstanbul University İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Muhammet İrfan Dönmez
- Department of Urology, Division of Pediatric Urology, İstanbul University İstanbul Faculty of Medicine, İstanbul, Turkey.
| | - Selçuk Erdem
- Department of Urology, İstanbul University İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Orhan Ziylan
- Department of Urology, Division of Pediatric Urology, İstanbul University İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Tayfun Oktar
- Department of Urology, Division of Pediatric Urology, İstanbul University İstanbul Faculty of Medicine, İstanbul, Turkey; Department of Urology, Koç University Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
45
|
Lee A, Concepcion W, Gonzales S, Sutherland SM, Hollander SA. Acute kidney injury and chronic kidney disease after combined heart-liver transplant in patients with congenital heart disease: A retrospective case series. Pediatr Transplant 2020; 24:e13833. [PMID: 32985770 DOI: 10.1111/petr.13833] [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] [Received: 06/23/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 11/29/2022]
Abstract
Although it is known that children undergoing heart transplantation are at increased risk for both AKI and CKD, renal function following CHLT remains understudied. All pediatric CHLT patients from 2006 to 2019 were included. The prevalence of AKI in the first 7 post-operative days, renal recovery at 30 post-operative days, and CKD were ascertained. AKI was defined as an increase in creatinine greater than 1.5 times baseline, and CKD, as an eCrCl less than 90 mL/min/1.73 m2 . The need for RRT was also analyzed. 10 patients were included, with an average age of 20 years and an average listing time of 130 days. Preoperatively, the median eCrCl was 91.12 mL/min/m2 (IQR 70.51, 127.75 min/mL/m2 ). 5 (50%) patients had CKD, with 4 at stage 2 and 1 at stage 3. AKI occurred post-operatively in 3 of 9 (33%) patients: 2 at stage 1 and 1 at stage 2. 2 (67%) resolved by 7 days. Of the 5 patients who reached their 1-year follow-up, 1 (20%) had stage 3 CKD. Among 2 patients, neither had CKD at 5 years. One patient required RRT 2 weeks after CHLT. Despite an increased prevalence of preoperative CKD, patients undergoing CHLT have a lower AKI prevalence than those receiving an isolated heart or liver transplant. Of those with AKI, early renal recovery is common, although at 1 year CKD remains present in 20%. Among long-term survivors, normal renal function is achievable.
Collapse
Affiliation(s)
- Angela Lee
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Waldo Concepcion
- Division of Abdominal Transplantation, Department of General Surgery, Stanford University, Stanford, California, USA
| | - Selena Gonzales
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Scott M Sutherland
- Division of Nephrology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Seth A Hollander
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
46
|
Perazzo S, Revenis M, Massaro A, Short BL, Ray PE. A New Approach to Recognize Neonatal Impaired Kidney Function. Kidney Int Rep 2020; 5:2301-2312. [PMID: 33305124 PMCID: PMC7710891 DOI: 10.1016/j.ekir.2020.09.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/27/2020] [Accepted: 09/22/2020] [Indexed: 01/01/2023] Open
Abstract
Introduction Previous studies in term newborns with hypoxic ischemic encephalopathy showed that the rate of serum creatinine (SCr) decline during the first week of life could be used to identify newborns with impaired kidney function (IKF) who are missed by standard definitions of neonatal acute kidney injury (nAKI). Methods Retrospective review of the medical records of 329 critically ill newborns ≥27 weeks of gestational age (GA) admitted to a level 4 neonatal intensive care unit (NICU). We tested the hypothesis that the rate of SCr decline combined with SCr thresholds provides a sensitive approach to identify term and preterm newborns with IKF during the first week of life. Results Excluding neonates with nAKI, an SCr decline <31% by the seventh day of life, combined with an SCr threshold ≥0.7 mg/dl, recognized newborns of 40 to 31 weeks of GA with IKF. An SCr decline <21% combined with an SCr threshold ≥0.8 mg/dl identified newborns of 30 to 27 weeks of GA with IKF. Neonates with IKF (∼17%), like those with nAKI (7%), showed a more prolonged hospital stay and required more days of mechanical ventilation, vasoactive drugs, and diuretics, when compared with the controls. Changes in urine output did not distinguish newborns with IKF. Conclusion The rate of SCr decline combined with SCr thresholds identifies newborns with IKF during the first week of life. This distinctive group of newborns that is missed by standard definitions of nAKI, warrants close monitoring in the NICU to prevent further renal complications.
Collapse
Affiliation(s)
- Sofia Perazzo
- Division of Neonatology, Children's National Hospital, Washington, DC, USA
| | - Mary Revenis
- Division of Neonatology, Children's National Hospital, Washington, DC, USA.,Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - An Massaro
- Division of Neonatology, Children's National Hospital, Washington, DC, USA.,Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Billie L Short
- Division of Neonatology, Children's National Hospital, Washington, DC, USA.,Department of Pediatrics, The George Washington University, Washington, DC, USA
| | - Patricio E Ray
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
47
|
Idkaidek N, Hamadi S, Bani-Domi R, Al-Adham I, Alsmadi M, Awaysheh F, Aqrabawi H, Al-Ghazawi A, Rabayah A. Saliva versus Plasma Therapeutic Drug Monitoring of Gentamicin in Jordanian Preterm Infants. Development of a Physiologically-Based Pharmacokinetic (PBPK) Model and Validation of Class II Drugs of Salivary Excretion Classification System. Drug Res (Stuttg) 2020; 70:455-462. [PMID: 32877949 DOI: 10.1055/a-1233-3582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Gentamicin has proven to be a very successful treatment for bacterial infection, but it also can cause adverse effects, especially ototoxicity, which is irreversible. Therapeutic drug monitoring (TDM) in saliva is a more convenient non-invasive alternative compared to plasma. A physiologically-based pharmacokinetic (PBPK) model of gentamicin was built and validated using previously-published plasma and saliva data. The validated model was then used to predict experimentally-observed plasma and saliva gentamicin TDM data in Jordanian pediatric preterm infant patients measured using sensitive LCMS/MS method. A correlation was established between plasma and saliva exposures. The developed PBPK model predicted previously reported gentamicin levels in plasma, saliva and those observed in the current study. A good correlation was found between plasma and saliva exposures. The PBPK model predicted that gentamicin in saliva is 5-7 times that in plasma, which is in agreement with observed results. Saliva can be used as an alternative for TDM of gentamicin in preterm infant patients. Exposure to gentamicin in plasma and saliva can reliably be predicted using the developed PBPK model in patients.
Collapse
Affiliation(s)
| | - Salim Hamadi
- College of Pharmacy, University of Petra, Amman, Jordan
| | | | | | - Motasem Alsmadi
- College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Faten Awaysheh
- Royal Medical Services, Queen Rania Children Hospital, Amman, Jordan
| | - Hisham Aqrabawi
- Royal Medical Services, Queen Rania Children Hospital, Amman, Jordan
| | | | | |
Collapse
|
48
|
Zhang Y, Mehta N, Muhari-Stark E, Burckart GJ, van den Anker J, Wang J. Pediatric Renal Ontogeny and Applications in Drug Development. J Clin Pharmacol 2020; 59 Suppl 1:S9-S20. [PMID: 31502684 DOI: 10.1002/jcph.1490] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
The clinical applications of renal ontogeny mainly include renal function evaluation and optimal dosing of renally eliminated drugs in pediatric patients, which rely on pharmacometric models and/or bedside estimated glomerular filtration rate equations. However, these applications in drug development are based on an understanding of renal function development, especially when considering premature infants, and the renal biomarkers that can be used for renal function assessment. This review provides a general overview on (1) renal function development, (2) the biomarkers that are used to assess renal function, and (3) the practical application of this knowledge to drug dosing for renally eliminated drugs during pediatric development. While pharmacometric approaches for estimating renal function during development have improved considerably, the number of drug development programs that have studied premature infants is small and suggests that caution should be taken in estimating doses for renally eliminated drugs during periods of rapid change in renal function.
Collapse
Affiliation(s)
- Yifei Zhang
- Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Neha Mehta
- Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Gilbert J Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA.,Pediatric Pharmacology and Pharmacometrics Research Center, University of Basel Children's Hospital, Basel, Switzerland
| | - Jian Wang
- Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
49
|
Crass RL, Pai MP. Optimizing Estimated Glomerular Filtration Rate to Support Adult to Pediatric Pharmacokinetic Bridging Studies in Patients with Cystic Fibrosis. Clin Pharmacokinet 2020; 58:1323-1332. [PMID: 30972695 DOI: 10.1007/s40262-019-00761-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The estimated glomerular filtration rate (eGFR) is often used to model drug clearance (CL) and scale doses across age and body size. Over their lifetime, patients with cystic fibrosis (CF) receive repeated courses of tobramycin, an antibiotic with eGFR-dependent CL, for the treatment of pulmonary exacerbations. Tobramycin population pharmacokinetic (PK) modeling can be used to decipher the best approach to define eGFR for pediatric bridging studies. METHODS Inpatients with CF who received intravenous tobramycin between 1 January 2006 and 30 May 2018 were eligible for inclusion. Encounters without tobramycin concentration measurement or missing covariate data were excluded. Population PK analysis was performed using NONMEM.Covariate models were built following identification of the base model, with specific emphasis on the effect of different methods of estimating renal function as a covariate of tobramycin CL. RESULTS A total of 296 CF patients contributed 1029 care encounters (420 pediatric, 609 adult) and 4352 tobramycin concentrations to this analysis. The median (minimum, maximum) age at encounter was 19 years (0.2, 60), with serum creatinine of 0.60 mg/dL (0.10, 3.41). A two-compartment model best described the observed data, with height and eGFR as significant covariates of tobramycin CL. eGFR was best modeled using a combination of the modified Schwartz and Chronic Kidney Disease Epidemiology Collaboration (CKDEPI) equations expressed in absolute units. CONCLUSIONS The CKDEPI equation bridges PK data generated in adults to adolescents with CF better than the current regulatory standard. The eGFR should be expressed in absolute units (mL/min) for PK analyses.
Collapse
Affiliation(s)
- Ryan L Crass
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI, 48109, USA
| | - Manjunath P Pai
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
50
|
Zhang Y, Sherwin CM, Gonzalez D, Zhang Q, Khurana M, Fisher J, Burckart GJ, Wang Y, Yao LP, Ganley CJ, Wang J. Creatinine-Based Renal Function Assessment in Pediatric Drug Development: An Analysis Using Clinical Data for Renally Eliminated Drugs. Clin Pharmacol Ther 2020; 109:263-269. [PMID: 32696977 DOI: 10.1002/cpt.1991] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/30/2020] [Indexed: 11/09/2022]
Abstract
The estimated glomerular filtration rate (eGFR) equations based on serum creatinine (SCR) have been used for pediatric dose adjustment in drug labeling. This study evaluated the performance of those equations in estimating individual clearance of drugs that are predominantly eliminated by glomerular filtration, using clinical data from the renally eliminated drugs gadobutrol, gadoterate, amikacin, and vancomycin. The eGFR was compared with the observed drug clearance (CL) in 352 pediatric patients from birth to 12 years of age. Multiple eGFR equations overestimated the drug CL on average, including the original and bedside Schwartz equations, which showed an average eGFR/CL ratio between 1 and 3. Further analysis with bedside Schwartz equation showed a higher eGFR/CL ratio in the subjects with a lower SCR or CL. Supraphysiological eGFR as high as 380 mL/min/1.73 m2 was obtained using the bedside Schwartz equation for some of the subjects, most of whom are children < 2 years of age with SCR < 0.2 mg/dL. Excluding the subjects with supraphysiological eGFR from the analysis did not change the overall trend of overestimation. In conclusion, Schwartz equations led to an overestimation of drug clearance for the drugs evaluated. When greater precision is required in predicting eGFR for pediatric patients, such as in drug dosing, revised k constants for the Schwartz equation or new methods of glomerular filtration rate estimation may be necessary.
Collapse
Affiliation(s)
- Yifei Zhang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Catherine M Sherwin
- Department of Pediatrics, Wright State University School of Medicine, Dayton Children's Hospital, Dayton, Ohio, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Qunshu Zhang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mona Khurana
- Division of Pediatric and Maternal Health, Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffrey Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lynne P Yao
- Division of Pediatric and Maternal Health, Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Charles J Ganley
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jian Wang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|