1
|
Ariceta G, Ardissino G. A better future for children with STEC-hemolytic uremic syndrome: news from Argentina. Pediatr Nephrol 2025; 40:1-2. [PMID: 38954038 DOI: 10.1007/s00467-024-06429-y] [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: 05/19/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024]
Affiliation(s)
- Gema Ariceta
- Department of Pediatric Nephrology, Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain.
| | - Gianluigi Ardissino
- Centro per la Cura e lo Studio della Sindrome Emolitico-Uremica, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
2
|
Reidy KJ, Guillet R, Selewski DT, Defreitas M, Stone S, Starr MC, Harer MW, Todurkar N, Vuong KT, Gogcu S, Askenazi D, Tipple TE, Charlton JR. Advocating for the inclusion of kidney health outcomes in neonatal research: best practice recommendations by the Neonatal Kidney Collaborative. J Perinatol 2024; 44:1863-1873. [PMID: 38969825 PMCID: PMC11606916 DOI: 10.1038/s41372-024-02030-1] [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: 04/15/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
Acute kidney injury (AKI) occurs in nearly 30% of sick neonates. Chronic kidney disease (CKD) can be detected in certain populations of sick neonates as early as 2 years. AKI is often part of a multisystem syndrome that negatively impacts developing organs resulting in short- and long-term pulmonary, neurodevelopmental, and cardiovascular morbidities. It is critical to incorporate kidney-related data into neonatal clinical trials in a uniform manner to better understand how neonatal AKI or CKD could affect an outcome of interest. Here, we provide expert opinion recommendations and rationales to support the inclusion of short- and long-term neonatal kidney outcomes using a tiered approach based on study design: (1) observational studies (prospective or retrospective) limited to data available within a center's standard practice, (2) observational studies involving prospective data collection where prespecified kidney outcomes are included in the design, (3) interventional studies with non-nephrotoxic agents, and (4) interventional studies with known nephrotoxic agents. We also provide recommendations for biospecimen collection to facilitate ancillary kidney specific research initiatives. This approach balances the costs of AKI and CKD ascertainment with knowledge gained. We advocate that kidney outcomes be included routinely in neonatal clinical study design. Consistent incorporation of kidney outcomes across studies will increase our knowledge of neonatal morbidity.
Collapse
Affiliation(s)
- Kimberly J Reidy
- Division of Nephrology, Department of Pediatrics, Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY, 10467, USA
| | - Ronnie Guillet
- Division of Neonatology, Golisano Children's Hospital, University of Rochester, Rochester, NY, USA
| | - David T Selewski
- Division of Nephrology, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Marissa Defreitas
- Division of Nephrology, Department of Pediatrics, University of Miami/Holtz Children's Hospital, Miami, FL, USA
| | - Sadie Stone
- Department of Pharmacy, Children's of Alabama, Birmingham, AL, UK
| | - Michelle C Starr
- Division of Pediatric Nephrology, Division of Child Health Service Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew W Harer
- Division of Neonatology, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Namrata Todurkar
- Division of Neonatal Perinatal Medicine, Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kim T Vuong
- Division of Pediatric Nephrology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Semsa Gogcu
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - David Askenazi
- Division of Nephrology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Trent E Tipple
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer R Charlton
- Division of Nephrology, Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA, 22903, USA.
| |
Collapse
|
3
|
Mohamed T, Alexander R, Davidson B, Klamer B, Gehred A, Starr MC, Slagle C, Krawczeski C, Harer MW. Urinary Neutrophil Gelatinase-Associated Lipocalin Values in Preterm Neonates: A Systematic Review and Meta-analysis. Am J Perinatol 2024. [PMID: 39437994 DOI: 10.1055/a-2417-4087] [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: 10/25/2024]
Abstract
OBJECTIVE Acute kidney injury (AKI) is common in hospitalized preterm neonates. Urinary neutrophil gelatinase-associated lipocalin (uNGAL) is a promising noninvasive AKI biomarker. However, normal values of uNGAL in preterm neonates without AKI are poorly characterized. The objective of this study was to evaluate the current literature to determine normal uNGAL values for preterm neonates without AKI. STUDY DESIGN Systematic review and meta-analysis of all articles published before November 2021 evaluating uNGAL values in preterm neonates without AKI. RESULTS Of 1,607 studies evaluated for eligibility, 11 were included in the final meta-analysis (210 males, 202 females). uNGAL values were higher in the preterm neonates <29 weeks and ranged between 20.7 and 782.65 ng/mL. Meta mean estimates of gestational age (GA), birthweight, and neutrophil gelatinase-associated lipocalin were 29.4 weeks (95% confidence interval [CI]: 28.8-30.0), 1,241 g (95% CI: 1,111-1,372), and 148.9 ng/mL (95% CI: 48-231), respectively. CONCLUSION In limited studies, a wide range of uNGAL values in preterm neonates without AKI are reported. Future studies should identify normal uNGAL values in preterm neonates using larger cohorts by GA and birthweight. KEY POINTS · Urinary NGAL is a promising noninvasive biomarker of neonatal AKI.. · A wide range of uNGAL is reported in preterm neonates but the baseline values are not well defined.. · Urine NGAL values are higher in extremely preterm compared with preterm neonates..
Collapse
Affiliation(s)
- Tahagod Mohamed
- Division of Nephrology and Hypertension, Department of Pediatric, Nationwide Children's Hospital, Columbus, Ohio
- The Kidney and Urinary Tract Center, Department of Pediatric, Nationwide Children's Hospital, Columbus, Ohio
| | - Robin Alexander
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | | | - Brett Klamer
- The Kidney and Urinary Tract Center, Department of Pediatric, Nationwide Children's Hospital, Columbus, Ohio
| | | | - Michelle C Starr
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Cara Slagle
- Division of Neonatal-Perinatal Medicine, Riley Children's Hospital, Indiana University, Indianapolis, Indiana
| | - Catherine Krawczeski
- Division of Pediatric Cardiology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - Matthew W Harer
- Division of Neonatology, Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
4
|
Vander Elst Z, Laenen A, Deberdt J, Delemarre L, Vermeersch P, Frans G, Naulaers G, Gijsen M, Dreesen E, Spriet I, Allegaert K, Smits A. Human serum albumin: prediction model and reference values for preterm and term neonates. Pediatr Res 2024:10.1038/s41390-024-03634-1. [PMID: 39394426 DOI: 10.1038/s41390-024-03634-1] [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: 11/22/2023] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Human serum albumin (HSA) concentrations may alter HSA-bound drug distribution. This study aims to describe longitudinal real-world HSA trends, and to develop a prediction model for HSA concentrations using a large neonatal cohort. METHODS Patients admitted to the neonatal intensive care unit of the University Hospitals Leuven (postnatal age (PNA) ≤28days) were retrospectively included. Using linear mixed models, covariate effects on HSA were explored. A multivariable prediction model was developed (backward model selection procedure, 1% significance level). RESULTS In total, 848 neonates were included [median(interquartile range) gestational age (GA) 35(32-38)weeks, birth weight (BW) 2400(1640-3130)grams]. Median HSA concentration was 32.3(28.7-35.6)g/L. Longitudinal analyses demonstrated increasing HSA concentrations with PNA and GA for most GA groups. Univariable analyses revealed significant associations of HSA with PNA, GA, BW, current weight, total and direct bilirubin, total plasma proteins, respiratory support, mechanical ventilation, sepsis, ibuprofen use, and C-reactive protein (p-values < 0.05). A high-performance (R2 = 76.3%) multivariable HSA prediction model was developed, and PNA- and GA-dependent HSA centiles were provided. CONCLUSION Population-specific HSA centiles and an accurate neonatal HSA prediction model were developed, incorporating both maturational and non-maturational covariates. These results can enhance future clinical care and pharmacokinetic analyses to improve pharmacotherapy of HSA-bound drugs in neonates, respectively. IMPACT To improve future pharmacokinetic modeling initiatives, a high-performance human serum albumin (HSA) prediction model was developed for (pre)term neonates, using a large, single-center cohort of real-world data. This prediction model integrates both maturational and non-maturational covariates, resulting in accurate HSA predictions in neonates. Additionally, HSA centiles based on postnatal and gestational age were developed, which can be easily applied in clinical practice when interpreting HSA concentrations of neonates. In general, unbound drug fractions are higher in neonates compared to older populations. To improve pharmacotherapy of HSA-bound drugs in neonates, the obtained results can be integrated in future pharmacokinetic-pharmacodynamic analyses.
Collapse
Affiliation(s)
- Zoë Vander Elst
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-Biostat), KU Leuven, Leuven, Belgium
| | | | | | - Pieter Vermeersch
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Glynis Frans
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Gunnar Naulaers
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Gijsen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Pharmacy Department, UZ Leuven, Leuven, Belgium
| | - Erwin Dreesen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Pharmacy Department, UZ Leuven, Leuven, Belgium
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Bekker A, Capparelli EV, Mirochnick M, Clarke DF, Cotton MF, Shapiro R, McCarthy K, Moye J, Violari A, Chokephaibulkit K, Abrams E, Penazzato M, Ruel TD, Cressey TR. Lamivudine dosing for preterm infants exposed to HIV: a population pharmacokinetic modelling and simulation study. J Antimicrob Chemother 2024; 79:2570-2574. [PMID: 39092932 PMCID: PMC11441998 DOI: 10.1093/jac/dkae259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVES To develop a pragmatic twice daily lamivudine dosing strategy for preterm infants from 24 to 37 completed weeks of gestation. METHODS Data were combined from eight pharmacokinetic studies in neonates and infants receiving lamivudine oral solution. A population pharmacokinetic model was developed using non-linear mixed effects regression. Different lamivudine dosing strategies, stratified by gestational age at birth (GA) bands, were simulated in a virtual population of preterm infants, aimed at maintaining lamivudine drug exposures (AUC0-12) within a reference target range of 2.95 to 13.25 µg·h/mL, prior to switching to WHO-weight band doses when ≥4 weeks of age and weighing ≥3 kg. RESULTS A total of 154 infants (59% female) contributed 858 lamivudine plasma concentrations. Median (range) GA at birth was 38 (27-41) weeks. At the time of first pharmacokinetic sampling infants were older with median postnatal age (PNA) of 6.3 (0.52-26.6) weeks. Lamivudine concentrations were described by a one-compartment model, with CL/F and V/F allometrically scaled to weight. Maturation of CL/F was described using an Emax model based on PNA. CL/F was also adjusted on GA to allow extrapolation for extreme prematurity. Simulations predicted an optimal lamivudine dosing for infants GA ≥24 to <30 weeks of 2 mg/kg twice daily from birth until weighing 3 kg; and for GA ≥30 to <37 weeks, 2 mg/kg twice daily for the first 4 weeks of life, followed by 4 mg/kg twice daily until weighing 3 kg. CONCLUSIONS Model-based predictions support twice daily pragmatic GA band dosing of lamivudine for preterm infants, but clinical validation is warranted.
Collapse
Affiliation(s)
- Adrie Bekker
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Edmund V Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, CA, USA
| | - Mark Mirochnick
- Division of Neonatology, Department of Pediatrics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Diana F Clarke
- Section of Pediatric Infectious Diseases, Boston Medical Center, Boston, MA, USA
| | - Mark F Cotton
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Roger Shapiro
- Botswana-Harvard AIDS Institute Partnership for HIV Research and Education, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Jack Moye
- Division of Extramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Avy Violari
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Kulkanya Chokephaibulkit
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Elaine Abrams
- ICAP at Columbia University, Mailman School of Public Health, and Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Martina Penazzato
- Research for Health Department, Science Division, World Health Organization, Geneva, Switzerland
| | - Theodore D Ruel
- Division of Pediatric Infectious Diseases and Global Health, University of California, San Francisco, San Francisco, CA, USA
| | - Tim R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
6
|
Ye P, Shi J, Guo Z, Yang X, Li Q, Chen K, Zhao F, Zhou H, Zhang Y, van den Anker J, Song L, Zhao W. Piperacillin/tazobactam treatment in children: evidence of subtherapeutic concentrations. Front Pharmacol 2024; 15:1254005. [PMID: 39027331 PMCID: PMC11255394 DOI: 10.3389/fphar.2024.1254005] [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: 11/27/2023] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Objective Piperacillin/tazobactam (PIP/TAZ) is used for the treatment of lower respiratory tract bacterial infections in children. This study was performed to evaluate if the current dosing regimen results in therapeutic drug concentrations. Patients and methods Patients suspected or proven to have lower respiratory tract bacterial infection and administrated PIP/TAZ intravenously for a duration of no less than 0.5 h, q6h-q12h daily, were enrolled. Blood samples were collected, and PIP concentrations were determined by high-performance liquid chromatography. The individual predicted concentration of PIP was evaluated using the individual empirical Bayesian estimate method. The evaluated PK/PD targets included (1) 70% time when the predicted free drug concentration exceeds the minimum inhibitory concentration (fT > MIC) and (2) 50% fT > 4× MIC. Probability of target attainment (PTA) was assessed by the proportion of patients who reached the PK/PD targets. The PIP concentrations between different groups of patients were compared. Results A total of 57 samples were collected from 57 patients with a median age of 2.26 years (0.17-12.58). For the PK/PD targets of 70% fT > MIC and 50% fT > 4× MIC for Pseudomonas aeruginosa and Klebsiella pneumoniae, the PTA was all 0. The median Cmin of PIP was significantly higher in infants than in children, and the median Cmin after administration in q8h was significantly higher than that after administration in q12h. Conclusion The current dose regimen of PIP/TAZ leads to extremely low plasma concentrations in most children with lower respiratory tract bacterial infections. More optimized dosing regimens or better alternative therapies need to be further explored.
Collapse
Affiliation(s)
- Panpan Ye
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Jinyi Shi
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Zixuan Guo
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Xinmei Yang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Qian Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Keguang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Furong Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Haiyan Zhou
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Yehui Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC, United States
- Departments of Pediatrics, Pharmacology and Physiology, Genomics and Precision Medicine, the George Washington University School of Medicine and Health Sciences, Washington, DC, United States
- Department of Paediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Linlin Song
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
7
|
Fataar A, Pillay-Fuentes Lorente V, Decloedt EH, van Eck A, Reddy K, Dramowski A, Bekker A. A Retrospective Study Evaluating Neonatal Vancomycin Loading Doses to Achieve a Therapeutic Target. Ther Drug Monit 2024:00007691-990000000-00238. [PMID: 38858809 DOI: 10.1097/ftd.0000000000001234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Vancomycin is a glycopeptide antibiotic that has been used to treat hospital-acquired gram-positive infections for more than 5 decades. However, the literature is divided regarding the therapeutic advantages of vancomycin loading doses in neonates. OBJECTIVES This study aimed to investigate the effect of vancomycin loading doses on therapeutic target attainment in neonates with sepsis. METHODS A retrospective cohort study was conducted to compare the vancomycin target attainment (area under the curve 0-24 hours/minimum inhibitory concentration ≥400) in neonates before and after the 2019 change in vancomycin prescription guidelines at a neonatal unit in Cape Town, South Africa. As the standard of care, Bayesian modelling software was used to compute the area under the curve from the trough concentrations. RESULTS Two hundred ten neonates were included. Multivariate regression analysis showed a 2-fold increase in the odds of target attainment among neonates receiving a loading dose of vancomycin. Early target attainment (within 8-12 hours of treatment initiation) was significantly higher in the loading dose group compared with the no loading dose group [97/105 (92.4%) versus 64/105 (61.0%); P < 0.001]. However, the overall proportion of neonates achieving target attainment at 24 hours was similar between groups [73/105 (69.5%) in the loading dose group versus 62/105 (59.0%) in the no loading dose group; P = 0.110]. The nephrotoxicity rates were low [2/105 (1.9%) in the loading dose group and 2/105 (1.9%) in the no loading dose group]. CONCLUSIONS The addition of a vancomycin loading dose to neonates may facilitate early therapeutic target attainment.
Collapse
Affiliation(s)
- Aaqilah Fataar
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Veshni Pillay-Fuentes Lorente
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; and
| | - Eric H Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; and
| | - Andrew van Eck
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Kessendri Reddy
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University and National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| | - Angela Dramowski
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Adrie Bekker
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
8
|
Aithal N, Kandasamy Y. The Babyccino: The Role of Caffeine in the Prevention of Acute Kidney Injury in Neonates-A Literature Review. Healthcare (Basel) 2024; 12:529. [PMID: 38470639 PMCID: PMC10931184 DOI: 10.3390/healthcare12050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Acute kidney injury (AKI) in neonates is associated with increased morbidity and mortality. Theophylline (a methylxanthine) has been shown to prevent neonatal AKI but is seldom used due to its unfavorable profile. Caffeine, another methylxanthine, is utilized ubiquitously to treat apnea of prematurity, but there are no randomized trials evaluating its efficacy in preventing neonatal AKI. This literature review aims to summarize the existing research pertaining to the relationship between caffeine and neonatal AKI. The review was conducted using Pubmed, Embase, Google Scholar, and Cochrane. Inclusion criteria incorporated empirical studies, being published in English, and being available electronically. All eight studies identified were included. Seven studies found caffeine-exposed premature neonates had lower rates of AKI than caffeine-unexposed neonates. Four found reduced AKI severity with caffeine exposure. One study included term neonates and did not find a difference in the AKI rate between caffeine-exposed and non-exposed babies. Limitations include exclusively observational studies, short study periods, heterogenous definitions of prematurity, and a lack of assessment of dose-effect relationships. In conclusion, premature neonates exposed to caffeine appear to have lower rates and potentially less severe AKI. Further research is needed before caffeine can be considered for use in the primary prevention of neonatal AKI.
Collapse
Affiliation(s)
- Nimisha Aithal
- Department of Pediatrics, Townsville University Hospital, Townsville, QLD 4811, Australia
| | - Yogavijayan Kandasamy
- Department of Neonatology, Townsville University Hospital, Townsville, QLD 4811, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4814, Australia
| |
Collapse
|
9
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [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: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
10
|
Kandasamy Y, Baker S. An Exploratory Review on the Potential of Artificial Intelligence for Early Detection of Acute Kidney Injury in Preterm Neonates. Diagnostics (Basel) 2023; 13:2865. [PMID: 37761232 PMCID: PMC10529317 DOI: 10.3390/diagnostics13182865] [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: 06/06/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
A preterm birth is a live birth that occurs before 37 completed weeks of pregnancy. Approximately 15 million babies are born preterm annually worldwide, indicating a global preterm birth rate of about 11%. Up to 50% of premature neonates in the gestational age (GA) group of <29 weeks' gestation will develop acute kidney injury (AKI) in the neonatal period; this is associated with high mortality and morbidity. There are currently no proven treatments for established AKI, and no effective predictive tool exists. We propose that the development of advanced artificial intelligence algorithms with neural networks can assist clinicians in accurately predicting AKI. Clinicians can use pathology investigations in combination with the non-invasive monitoring of renal tissue oxygenation (rSO2) and renal fractional tissue oxygenation extraction (rFTOE) using near-infrared spectroscopy (NIRS) and the renal resistive index (RRI) to develop an effective prediction algorithm. This algorithm would potentially create a therapeutic window during which the treating clinicians can identify modifiable risk factors and implement the necessary steps to prevent the onset and reduce the duration of AKI.
Collapse
Affiliation(s)
- Yogavijayan Kandasamy
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW 2308, Australia
- Department of Neonatology, Townsville University Hospital, Townsville, QLD 4814, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4810, Australia
| | - Stephanie Baker
- College of Science and Engineering, James Cook University, Cairns, QLD 4878, Australia;
| |
Collapse
|
11
|
Ketharanathan N, Lili A, de Vries JMP, Wildschut ED, de Hoog M, Koch BCP, de Winter BCM. A Population Pharmacokinetic Model of Pentobarbital for Children with Status Epilepticus and Severe Traumatic Brain Injury. Clin Pharmacokinet 2023; 62:1011-1022. [PMID: 37247187 PMCID: PMC10338388 DOI: 10.1007/s40262-023-01249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Pentobarbital pharmacokinetics (PK) remain elusive and the therapeutic windows narrow. Administration is frequent in critically ill children with refractory status epilepticus (SE) and severe traumatic brain injury (sTBI). OBJECTIVES To investigate pentobarbital PK in SE and sTBI patients admitted to the paediatric intensive care unit (PICU) with population-based PK (PopPK) modelling and dosing simulations. METHODS Develop a PopPK model with non-linear mixed-effects modelling (NONMEM®) with retrospective data (n = 36; median age 1.3 years; median weight 10 kg; 178 blood samples) treated with continuous intravenous pentobarbital. An independent dataset was used for external validation (n = 9). Dosing simulations with the validated model evaluated dosing regimens. RESULTS A one-compartment PK model with allometrically scaled weight on clearance (CL; 0.75) and volume of distribution (Vd; 1) captured data well. Typical CL and Vd values were 3.59 L/70 kg/h and 142 L/70 kg, respectively. Elevated creatinine and C-reactive protein (CRP) levels significantly correlated to decreased CL, explaining 84% of inter-patient variability, and were incorporated in the final model. External validation using stratified visual predictive checks showed good results. Simulations demonstrated patients with elevated serum creatinine and CRP failed to achieve steady state yet progressed to toxic levels with current dosing regimens. CONCLUSIONS The one-compartment PK model of intravenous pentobarbital described data well whereby serum creatinine and CRP significantly correlated with pentobarbital CL. Dosing simulations formulated adjusted dosing advice in patients with elevated creatinine and/or CRP. Prospective PK studies with pharmacodynamic endpoints, are imperative to optimise pentobarbital dosing in terms of safety and clinical efficacy in critically ill children.
Collapse
Affiliation(s)
- Naomi Ketharanathan
- Department of Neonatal and Paediatric Intensive Care, Division of Paediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Room Sp-3435, Wytemaweg 80, 3015GD, Rotterdam, The Netherlands.
| | - Anastasia Lili
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
| | | | - Enno D Wildschut
- Department of Neonatal and Paediatric Intensive Care, Division of Paediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Room Sp-3435, Wytemaweg 80, 3015GD, Rotterdam, The Netherlands
| | - Matthijs de Hoog
- Department of Neonatal and Paediatric Intensive Care, Division of Paediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Room Sp-3435, Wytemaweg 80, 3015GD, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
| | - Brenda C M de Winter
- Rotterdam Clinical Pharmacometrics Group, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023; 11:biomedicines11030940. [PMID: 36979919 PMCID: PMC10046592 DOI: 10.3390/biomedicines11030940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/22/2023] Open
Abstract
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.
Collapse
|
13
|
Staub E. Current and potential methods to assess kidney structure and morphology in term and preterm neonates. Anat Rec (Hoboken) 2023. [PMID: 36883787 DOI: 10.1002/ar.25195] [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: 11/20/2022] [Revised: 01/25/2023] [Accepted: 02/21/2023] [Indexed: 03/09/2023]
Abstract
After birth, the kidney structure in neonates adapt to the functional demands of extrauterine life. Nephrogenesis is complete in the third trimester, but glomeruli, tubuli, and vasculature mature with the rapidly increasing renal blood flow and glomerular filtration. In preterm infants, nephrogenesis remains incomplete and maturation is slower and may be aberrant. This structural and functional deficit has life-long consequences: preterm born individuals are at higher risk for chronic kidney disease and arterial hypertension later in life. This review assembles the literature on existing and potential methods to visualize neonatal kidney structure and morphology and explore their potential to longitudinally document the developmental deviation after preterm birth. X-rays with and without contrast, fluoroscopy and computed tomography (CT) involve relevant ionizing radiation exposure and, apart from CT, do not provide sufficient structural details. Ultrasound has evolved into a safe and noninvasive high-resolution imaging method which is excellent for longitudinal observations. Doppler ultrasound modes can characterize and quantify blood flow to and through the kidneys. Microvascular flow imaging has opened new possibilities of visualizing previously unseen vascular structures. Recent advances in magnetic resonance imaging display renal structure and function in unprecedented detail, but are offset by the logistical challenges of the imaging procedure and limited experience with the new techniques in neonates. Kidney biopsies visualize structure histologically, but are too invasive and remain anecdotal in newborns. All the explored methods have predominantly been examined in term newborns and require further research on longitudinal structural observation in the kidneys of preterm infants.
Collapse
Affiliation(s)
- Eveline Staub
- Department of Neonatology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- University of Sydney Northern Clinical School, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
14
|
Lee-Sarwar KA, Chen YC, Lasky-Su J, Kelly RS, Zeiger RS, O’Connor GT, Bacharier LB, Jia X, Beigelman A, Gold DR, Laranjo N, Bunyavanich S, Weiss ST, Litonjua AA, Brennan PJ. Early-life fecal metabolomics of food allergy. Allergy 2023; 78:512-521. [PMID: 36448508 PMCID: PMC10590492 DOI: 10.1111/all.15602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Intestinal microenvironmental perturbations may increase food allergy risk. We hypothesize that children with clinical food allergy, those with food sensitization, and healthy children can be differentiated by intestinal metabolites in the first years of life. METHODS In this ancillary analysis of the Vitamin D Antenatal Asthma Reduction Trial (VDAART), we performed untargeted metabolomic profiling in 824 stool samples collected at ages 3-6 months, 1 year and 3 years. Subjects included 23 with clinical food allergy at age 3 and/or 6 years, 151 with food sensitization but no clinical food allergy, and 220 controls. We identified modules of correlated, functionally related metabolites and sought associations of metabolite modules and individual metabolites with food allergy/sensitization using regression models. RESULTS Several modules of functionally related intestinal metabolites were reduced among subjects with food allergy, including bile acids at ages 3-6 months and 1 year, amino acids at age 3-6 months, steroid hormones at 1 year, and sphingolipids at age 3 years. One module primarily containing diacylglycerols was increased in those with food allergy at age 3-6 months. Fecal caffeine metabolites at age 3-6 months, likely derived from breast milk, were increased in those with food allergy and/or sensitization (beta = 5.9, 95% CI 1.0-10.8, p = .02) and were inversely correlated with fecal bile acids and bilirubin metabolites, though maternal plasma caffeine levels were not associated with food allergy and/or sensitization. CONCLUSIONS Several classes of bioactive fecal metabolites are associated with food allergy and/or sensitization including bile acids, steroid hormones, sphingolipids, and caffeine metabolites.
Collapse
Affiliation(s)
- Kathleen A. Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Yih-Chieh Chen
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Robert S. Zeiger
- Department of Clinical Science Kaiser Permanente Bernard J. Tyson School of Medicine; Pasadena, CA, USA
| | - George T. O’Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine; Boston, MA, USA
| | - Leonard B. Bacharier
- Department of Pediatric Allergy, Immunology, and Pulmonary, Vanderbilt Children’s Hospital, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Xiaojiong Jia
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Avraham Beigelman
- Schneider Children’s Medical Center of Israel, Tel Aviv University; Tel Aviv, Israel; Division of Pediatric Allergy, Immunology & Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine; St. Louis, MO, USA
| | - Diane R. Gold
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Supinda Bunyavanich
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai; New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Scott T. Weiss
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical Center; Rochester, NY, USA
| | - Patrick J. Brennan
- Division of Allergy & Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA, USA
| |
Collapse
|
15
|
Acute Kidney Injury in Very Low Birth Weight Infants: A Major Morbidity and Mortality Risk Factor. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020242. [PMID: 36832371 PMCID: PMC9955621 DOI: 10.3390/children10020242] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVES Very low birth weight (VLBW) infants are at high risk of developing acute kidney injury (AKI), presumably secondary to low kidney reserves, stressful postnatal events, and drug exposures. Our study aimed to identify the prevalence, risk factors, and outcomes associated with AKI in VLBW infants. STUDY DESIGN Records of all VLBW infants admitted to two medical campuses between January 2019 and June 2020 were retrospectively reviewed. AKI was classified using the modified KDIGO definition to include only serum creatinine. Risk factors and composite outcomes were compared between infants with and without AKI. We evaluated the main predictors of AKI and death with forward stepwise regression analysis. RESULTS 152 VLBW infants were enrolled. 21% of them developed AKI. Based on the multivariable analysis, the most significant predictors of AKI were the use of vasopressors, patent ductus arteriosus, and bloodstream infection. AKI had a strong and independent association with neonatal mortality. CONCLUSIONS AKI is common in VLBW infants and is a significant risk factor for mortality. Efforts to prevent AKI are necessary to prevent its harmful effects.
Collapse
|
16
|
Obiagwu PN, Morrow B, McCulloch M, Argent A. Burden and severity of deranged electrolytes and kidney function in children seen in a tertiary hospital in Kano, northern Nigeria. PLoS One 2023; 18:e0283220. [PMID: 36930619 PMCID: PMC10022757 DOI: 10.1371/journal.pone.0283220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Derangement in serum electrolytes and kidney function is often overlooked, especially in resource-constrained settings, and associated with increased risk of morbidity and mortality. This study aimed to describe the burden of derangements in serum electrolytes and kidney function in children presenting to a tertiary hospital in Nigeria. METHODS The laboratory records of all children who had serum electrolytes urea and creatinine ordered on their first presentation to hospital between January 1 and June 30, 2017 were retrospectively reviewed. Basic demographic data including admission status (inpatient or outpatient) were recordedandserum levels of sodium, potassium, chloride and bicarbonate were assessed for derangements usingnormal values from established reference ranges. Results of repeat samples were excluded. Kidney function was classified based on the serum creatinine relative to normal values for age and sex. RESULTS During the study period, 1909 children (60.3% male); median (IQR) age 42 (11.9) months had serum chemistry and 1248 (65.4%) were admitted. Results of their first samples were analyzed. Electrolyte derangements were present in 78.6% of the samples most commonly hyponatraemia (41.1%), low bicarbonate(37.2%), hypochloraemia (33.5%) and hypokalemia(18.9%). Azotaemia was found in 20.1% of the results. Elevated serum creatinine levels were found in 399 children (24.7%), 24.1% of those being in the severe category. Children aged 5 years and younger accounted for 76.4% of those with derangement in kidney function. One hundred and eight outpatients (17.8%) had deranged kidney function. CONCLUSION Deranged serum electrolytes and kidney function were common in this cohort.
Collapse
Affiliation(s)
- Patience N. Obiagwu
- Department of Paediatrics, Aminu Kano Teaching Hospital and Bayero University, Kano, Nigeria
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- * E-mail:
| | - Brenda Morrow
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mignon McCulloch
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Andrew Argent
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
17
|
Filler G, Gipson DS, Iyamuremye D, Díaz González de Ferris ME. Artificial Intelligence in Pediatric Nephrology-A Call for Action. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:17-24. [PMID: 36723276 DOI: 10.1053/j.akdh.2022.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
Artificial intelligence is playing an increasingly important role in many fields of clinical care to assist health care providers in patient management. In adult-focused nephrology, artificial intelligence is beginning to be used to improve clinical care, hemodialysis prescriptions, and follow-up of transplant recipients. This article provides an overview of medical artificial intelligence applications relevant to pediatric nephrology. We describe the core concepts of artificial intelligence and machine learning and cover the basics of neural networks and deep learning. We also discuss some examples for clinical applications of artificial intelligence in pediatric nephrology, including neonatal kidney function, early recognition of acute kidney injury, renally cleared drug dosing, intrapatient variability, urinary tract infection workup in infancy, and longitudinal disease progression. Furthermore, we consider the future of artificial intelligence in clinical pediatric nephrology and its potential impact on medical practice and address the ethical issues artificial intelligence raises in terms of clinical decision-making, health care provider-patient relationship, patient privacy, and data collection. This article also represents a call for action involving those of us striving to provide optimal services for children, adolescents, and young adults with chronic conditions.
Collapse
Affiliation(s)
- Guido Filler
- Division of Pediatric Nephrology, Departments of Paediatrics, Western University, London, Ontario, Canada; Departments of Medicine, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.
| | - Debbie S Gipson
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | | |
Collapse
|
18
|
Li Y, Wang B, Wang L, Shi K, Zhao W, Gao S, Chen J, Ding C, Du J, Gao W. Postoperative day 1 serum cystatin C level predicts postoperative delayed graft function after kidney transplantation. Front Med (Lausanne) 2022; 9:863962. [PMID: 36035383 PMCID: PMC9411520 DOI: 10.3389/fmed.2022.863962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background Delayed graft function (DGF) commonly occurs after kidney transplantation, but no clinical predictors for guiding post-transplant management are available. Materials and methods Data including demographics, surgery, anesthesia, postoperative day 1 serum cystatin C (S-CysC) level, kidney functions, and postoperative complications in 603 kidney transplant recipients who met the enrollment criteria from January 2017 to December 2018 were collected and analyzed to form the Intention-To-Treat (ITT) set. All perioperative data were screened using the least absolute shrinkage and selection operator. The discrimination, calibration, and clinical effectiveness of the predictor were verified with area under curve (AUC), calibration plot, clinical decision curve, and impact curve. The predictor was trained in Per-Protocol set, validated in the ITT set, and its stability was further tested in the bootstrap resample data. Result Patients with DGF had significantly higher postoperative day 1 S-CysC level (4.2 ± 1.2 vs. 2.8 ± 0.9 mg/L; P < 0.001), serum creatinine level (821.1 ± 301.7 vs. 554.3 ± 223.2 μmol/L; P < 0.001) and dialysis postoperative (74 [82.2%] vs. 25 [5.9%]; P < 0.001) compared with patients without DGF. Among 41 potential predictors, S-CysC was the most effective in the parsimonious model, and its diagnostic cut-off value was 3.80 mg/L with the risk score (OR, 13.45; 95% CI, 8.02–22.57; P < 0.001). Its specificity and sensitivity indicated by AUC was 0.832 (95% CI, 0.779–0.884; P < 0.001) with well fit calibration. S-CysC yielded up to 50% of clinical benefit rate with 1:4 of cost/benefit ratio. Conclusion The postoperative day 1 S-CysC level predicts DGF and may be used as a predictor of DGF but warrants further study.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Anesthesiology, 521 Hospital of Norinco Group, Xi’an, China
| | - Bo Wang
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Le Wang
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kewei Shi
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wangcheng Zhao
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Sai Gao
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiayu Chen
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chenguang Ding
- Department of Renal Transplantation, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Junkai Du
- Department of Emergency, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wei Gao,
| | - Wei Gao
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Junkai Du,
| |
Collapse
|
19
|
Kowalczyk A, Diaz-Gonzalez de Ferris ME, Filler G. Still trouble with serum creatinine measurements. Pediatr Nephrol 2022; 37:469-471. [PMID: 35048164 DOI: 10.1007/s00467-021-05387-z] [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: 11/26/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Alexandra Kowalczyk
- Departments of Paediatrics and Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5W9, Canada
| | | | - Guido Filler
- Departments of Paediatrics and Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5W9, Canada. .,The Lilibeth Caberto Kidney Clinical Research Unit, Western University, London, ON, N6A 5W9, Canada. .,Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N5A 5A5, Canada. .,Paediatrics, Medicine, and Pathology & Laboratory Medicine, Department of Paediatrics, Children's Hospital of Western Ontario, 800 Commissioners Road East, London, Ontario, N6A 5W9, Canada.
| |
Collapse
|
20
|
Filler G, Salerno F, McIntyre CW, de Ferris MEDG. Animal, Human, and 23Na MRI Imaging Evidence for the Negative Impact of High Dietary Salt in Children. CURRENT PEDIATRICS REPORTS 2021; 9:110-117. [PMID: 34567839 PMCID: PMC8449209 DOI: 10.1007/s40124-021-00249-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF THE REVIEW Conditions typically prevalent in adults such as hypertension, kidney stones, osteoporosis, and chronic kidney disease are increasing among adolescents and young adults (AYA). The purpose of this review is to describe the association of these conditions to a high salt diet among pediatric patients. RECENT FINDINGS We present animal, human, and 23Na MRI evidence associated with the negative impact of high dietary salt in children. Special focus is placed on novel 23Na MRI imaging which reveals the important concept of a third compartment for sodium storage in soft tissue. Finally, we make recommendations on who should not be on a low salt diet. SUMMARY A high salt intake predisposes children and AYA to considerable morbidity. We exhort the reader to engage in advocacy efforts to curve the incidence and prevalence of high salt-related life-limiting conditions.
Collapse
Affiliation(s)
- Guido Filler
- Departments of Pediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, 800 Commissioners Road East, London, ON E3-206N6A 5W9 Canada
- Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- Pathology & Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- Children’s Health Research Institute, University of Western Ontario, London, Canada
- Lilibeth Caberto Kidney Clinical Research Unit, London, ON Canada
| | - Fabio Salerno
- Lilibeth Caberto Kidney Clinical Research Unit, London, ON Canada
- Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Christopher William McIntyre
- Departments of Pediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, 800 Commissioners Road East, London, ON E3-206N6A 5W9 Canada
- Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- Children’s Health Research Institute, University of Western Ontario, London, Canada
- Lilibeth Caberto Kidney Clinical Research Unit, London, ON Canada
- Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | | |
Collapse
|
21
|
Filler G, Ferris MEDGD. Discrepant changes of urinary cystatin C and other urinary biomarkers in preterm neonates. J Pediatr (Rio J) 2021; 97:473-475. [PMID: 33639089 PMCID: PMC9432192 DOI: 10.1016/j.jped.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Guido Filler
- Western University, Schulich School of Medicine & Dentistry, Departments of Paediatrics and Medicine, London, Canada; Western University, The Lilibeth Caberto Kidney Clinical Research Unit, London, Canada; Western University, Schulich School of Medicine & Dentistry, Department of Pathology and Laboratory Medicine, London, Ontario, Canada.
| | | |
Collapse
|
22
|
Borloo N, Smits A, Thewissen L, Annaert P, Allegaert K. Creatinine Trends and Patterns in Neonates Undergoing Whole Body Hypothermia: A Systematic Review. CHILDREN-BASEL 2021; 8:children8060475. [PMID: 34200017 PMCID: PMC8228260 DOI: 10.3390/children8060475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022]
Abstract
Many neonates undergoing whole body hypothermia (WBH) following moderate to severe perinatal asphyxia may also suffer from renal impairment. While recent data suggest WBH-related reno-protection, differences in serum creatinine (Scr) patterns to reference patterns were not yet reported. We therefore aimed to document Scr trends and patterns in asphyxiated neonates undergoing WBH and compared these to centiles from a reference Scr data set of non-asphyxiated (near)term neonates. Using a systematic review strategy, reports on Scr trends (mean ± SD, median or interquartile range) were collected (day 1-7) in WBH cohorts and compared to centiles of an earlier reported reference cohort of non-asphyxia cases. Based on 13 papers on asphyxia + WBH cases, a pattern of postnatal Scr trends in asphyxia + WBH cases was constructed. Compared to the reference 50th centile Scr values, mean or median Scr values at birth and up to 48 h were higher in asphyxia + WBH cases with a subsequent uncertain declining trend towards, at best, high or high-normal creatinine values afterwards. Such patterns are valuable for anticipating average changes in renal drug clearance but do not yet cover the relevant inter-patient variability observed in WBH cases, as this needs pooling of individual Screa profiles, preferably beyond the first week of life.
Collapse
Affiliation(s)
- Noor Borloo
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; (N.B.); (A.S.)
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; (N.B.); (A.S.)
- Neonatal Intensive Care Unit, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Liesbeth Thewissen
- Neonatal Intensive Care Unit, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; (N.B.); (A.S.)
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Department of Clinical Pharmacy, Erasmus MC, Postbus 2040, 3000 GA Rotterdam, The Netherlands
- Correspondence: ; Tel.: +32-(16)-342020
| |
Collapse
|
23
|
|