1
|
Stokes MA, Kamel NA, Festa MS, Sandaradura I, Stocker SL. Scoping Review of Paediatric Population Pharmacokinetic Models of Morphine. Clin Pharmacokinet 2025:10.1007/s40262-025-01477-5. [PMID: 40310579 DOI: 10.1007/s40262-025-01477-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE AND PURPOSE This scoping review aimed to summarise all available population pharmacokinetic models of morphine and its metabolites (morphine-3-glucoronide [M3G], morphine-6-glucoronide [M6G]) in children and describe how morphine exposure varies across paediatric age groups and settings. Identifying the factors that contribute to pharmacokinetic variability may improve our understanding of a patient's pharmacodynamic response to morphine. METHODS We searched Embase and MEDLINE databases from inception to 8 March 2024 for paediatric population pharmacokinetic models of morphine and its metabolites. Two reviewers independently screened abstracts and full texts and extracted the data. The review was conducted according to Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. RESULTS In total, 21 paediatric population pharmacokinetic models of morphine were identified; 12 studies also included morphine metabolites (M3G and/or M6G). Neonates and young children (< 6 years) were the most studied age groups (18/21; 86%), whereas older children (> 6 years) and adolescents (> 10 years) were included in only 6 of the 21 (29%) models. Morphine pharmacokinetics were most commonly described with two-compartment (52%) and one-compartment (38%) structure with first-order elimination. Several model covariates were identified: bodyweight, post-natal age for neonates, body temperature, therapeutic cooling, duration of mechanical ventilation, and genetic variation in drug transporters that mediate the uptake of morphine (e.g. OCT1). CONCLUSION Several population pharmacokinetic models of morphine and its metabolites in paediatrics have been published across diverse patient groups. Bodyweight and age-related covariates emerged as the most common factors affecting clearance and distribution; other covariates, including mechanical ventilation, therapeutic cooling, and genetic variation, also impacted morphine pharmacokinetics. Further research should focus on validating the predictive accuracy of paediatric morphine models in different patient populations and the combined effect of covariates, such as those related to critical illness and genetic variation, on morphine pharmacokinetics.
Collapse
Affiliation(s)
- Michael A Stokes
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia
- Kids Critical Care Research, Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Noha A Kamel
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marino S Festa
- Kids Critical Care Research, Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Indy Sandaradura
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, Westmead Clinical School, The University of Sydney, Sydney, NSW, Australia
- Institute of Clinical Pathology and Medical Research, New South Wales Health Pathology, Westmead Hospital, Sydney, NSW, Australia
| | - Sophie L Stocker
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia.
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia.
- St. Vincent's Clinical Campus, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- Sydney Musculoskeletal Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
2
|
van Valkengoed DW, Krekels EHJ, Knibbe CAJ. All You Need to Know About Allometric Scaling: An Integrative Review on the Theoretical Basis, Empirical Evidence, and Application in Human Pharmacology. Clin Pharmacokinet 2025; 64:173-192. [PMID: 39644458 PMCID: PMC11782306 DOI: 10.1007/s40262-024-01444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 12/09/2024]
Abstract
Scaling approaches are used to describe or predict clearance for paediatric or obese populations from normal-weight adult values. Theoretical allometry assumes the existence of a universal bodyweight-based scaling relationship. Although theoretical allometry is highly disputed, it is commonly applied in pharmacological data analyses and clinical practice. The aim of the current review is to (1) increase pharmacologists' understanding of theoretical allometry to better understand the (implicit) assumptions and (dis)advantages and (2) highlight important methodological considerations with the application of this methodology. Theoretical allometry originated in an empirical, and later debated, observation by Kleiber of a scaling exponent of 0.75 between basal metabolic rate and body mass of mammals. The mathematical framework of West, Brown, and Enquist provides one possible explanation for this value. To date, multiple key assumptions of this framework have been disputed or disproven, and an increasing body of evidence is emerging against the existence of one universal allometric exponent. The promise of ease and universality of use that comes with theoretical approaches may be the reason they are so strongly sought after and defended. However, ecologists have suggested that the theory should move from a 'Newtonian approach', in which physical explanations are sought for a universal law and variability is of minor importance, to a 'Darwinian approach', in which variability is considered of primary importance for which evolutionary explanations can be found. No scientific support was found for the application of allometry for within-species scaling, so the application of basal metabolic rate-based scaling principles to clearance scaling remains unsubstantiated. Recent insights from physiologically based modelling approaches emphasise the interplay between drugs with different properties and physiological variables that underlie drug clearance, which drives the variability in the allometric scaling exponent in the field of pharmacology. To deal with this variability, drug-specific or patient-specific adaptations to theoretical allometric scaling are proposed, that introduce empiric elements and reduce the universality of the theory. The use of allometric scaling with an exponent of 0.75 may hold empirical merit for paediatric populations, except for the youngest individuals (aged ≤ 5 years). Nevertheless, biological interpretations and extrapolation potential attributed to models based on 0.75 allometric scaling are theoretically unfounded, and merits of the empirical application of this function should, as for all models, always be supported by appropriate model validation procedures. In this respect, it is not the value of the allometric exponent but the description and prediction of individual clearance values and drug concentrations that are of primary interest.
Collapse
Affiliation(s)
- Daan W van Valkengoed
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elke H J Krekels
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
- Certara Inc, Princeton, NJ, USA
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands.
- Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands.
| |
Collapse
|
3
|
Zeilmaker-Roest G, de Vries-Rink C, van Rosmalen J, van Dijk M, de Wildt SN, Knibbe CAJ, Koomen E, Jansen NJG, Kneyber MCJ, Maebe S, Van den Berghe G, Haghedooren R, Vlasselaers D, Bogers AJJC, Tibboel D, Wildschut ED. Intermittent intravenous paracetamol versus continuous morphine in infants undergoing cardiothoracic surgery: a multi-center randomized controlled trial. Crit Care 2024; 28:143. [PMID: 38689310 PMCID: PMC11061924 DOI: 10.1186/s13054-024-04905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/07/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND To determine whether intermittent intravenous (IV) paracetamol as primary analgesic would significantly reduce morphine consumption in children aged 0-3 years after cardiac surgery with cardiopulmonary bypass. METHODS Multi-center, randomized, double-blinded, controlled trial in four level-3 Pediatric Intensive Care Units (PICU) in the Netherlands and Belgium. Inclusion period; March 2016-July 2020. Children aged 0-3 years, undergoing cardiac surgery with cardiopulmonary bypass were eligible. Patients were randomized to continuous morphine or intermittent IV paracetamol as primary analgesic after a loading dose of 100 mcg/kg morphine was administered at the end of surgery. Rescue morphine was given if numeric rating scale (NRS) pain scores exceeded predetermined cutoff values. Primary outcome was median weight-adjusted cumulative morphine dose in mcg/kg in the first 48 h postoperative. For the comparison of the primary outcome between groups, the nonparametric Van Elteren test with stratification by center was used. For comparison of the proportion of patients with one or more NRS pain scores of 4 and higher between the two groups, a non-inferiority analysis was performed using a non-inferiority margin of 20%. RESULTS In total, 828 were screened and finally 208 patients were included; parents of 315 patients did not give consent and 305 were excluded for various reasons. Fourteen of the enrolled 208 children were withdrawn from the study before start of study medication leaving 194 patients for final analysis. One hundred and two patients received intermittent IV paracetamol, 106 received continuous morphine. The median weight-adjusted cumulative morphine consumption in the first 48 h postoperative in the IV paracetamol group was 5 times lower (79%) than that in the morphine group (median, 145.0 (IQR, 115.0-432.5) mcg/kg vs 692.6 (IQR, 532.7-856.1) mcg/kg; P < 0.001). The rescue morphine consumption was similar between the groups (p = 0.38). Non-inferiority of IV paracetamol administration in terms of NRS pain scores was proven; difference in proportion - 3.1% (95% CI - 16.6-10.3%). CONCLUSIONS In children aged 0-3 years undergoing cardiac surgery, use of intermittent IV paracetamol reduces the median weight-adjusted cumulative morphine consumption in the first 48 h after surgery by 79% with equal pain relief showing equipoise for IV paracetamol as primary analgesic. Trial Registration Clinicaltrials.gov, Identifier: NCT05853263; EudraCT Number: 2015-001835-20.
Collapse
MESH Headings
- Humans
- Morphine/therapeutic use
- Morphine/administration & dosage
- Acetaminophen/therapeutic use
- Acetaminophen/administration & dosage
- Male
- Female
- Infant
- Double-Blind Method
- Pain, Postoperative/drug therapy
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Belgium
- Netherlands
- Infant, Newborn
- Administration, Intravenous
- Cardiac Surgical Procedures/methods
- Child, Preschool
- Analgesics, Non-Narcotic/administration & dosage
- Analgesics, Non-Narcotic/therapeutic use
- Intensive Care Units, Pediatric/organization & administration
- Intensive Care Units, Pediatric/statistics & numerical data
- Pain Measurement/methods
Collapse
Affiliation(s)
- Gerdien Zeilmaker-Roest
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, The Netherlands.
| | - Christine de Vries-Rink
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Joost van Rosmalen
- Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Monique van Dijk
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Saskia N de Wildt
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital Nieuwegein/Utrecht, Utrecht, The Netherlands
| | - Erik Koomen
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nicolaas J G Jansen
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin C J Kneyber
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Sofie Maebe
- Department of Pediatrics, Maastricht University Medical Center+, MosaKids Children's Hospital, Maastricht, The Netherlands
| | | | | | - Dirk Vlasselaers
- Department of Intensive Care Medicine, UZ Leuven, Louvain, Belgium
| | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Dick Tibboel
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Enno D Wildschut
- Department of Neonatal and Pediatric Intensive Care, Division of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Dawes JM, Howard RF. Neonatal Pain: Significance, Assessment, and Management. NEONATAL ANESTHESIA 2023:505-527. [DOI: 10.1007/978-3-031-25358-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Hu TM. A General Biphasic Bodyweight Model for Scaling Basal Metabolic Rate, Glomerular Filtration Rate, and Drug Clearance from Birth to Adulthood. AAPS J 2022; 24:67. [PMID: 35538161 DOI: 10.1208/s12248-022-00716-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
The objective of this study is to propose a unified, continuous, and bodyweight-only equation to quantify the changes of human basal metabolic rate (BMR), glomerular filtration rate (GFR), and drug clearance (CL) from infancy to adulthood. The BMR datasets were retrieved from a comprehensive historical database of male and female subjects (0.02 to 64 years). The CL datasets for 17 drugs and the GFR dataset were generated from published maturation and growth models with reported parameter values. A statistical approach was used to simulate the model-generated CL and GFR data for a hypothetical population with 26 age groups (from 0 to 20 years). A biphasic equation with two power-law functions of bodyweight was proposed and evaluated as a general model using nonlinear regression and dimensionless analysis. All datasets universally reveal biphasic curves with two distinct linear segments on log-log plots. The biphasic equation consists of two reciprocal allometric terms that asymptotically determine the overall curvature. The fitting results show a superlinear scaling phase (asymptotic exponent >1; ca. 1.5-3.5) and a sublinear scaling phase (asymptotic exponent <1; ca. 0.5-0.7), which are separated at the phase transition bodyweight ranging from 5 to 20 kg with a mean value of 10 kg (corresponding to 1 year of age). The dimensionless analysis generalizes and offers quantitative realization of the maturation and growth process. In conclusion, the proposed mixed-allometry equation is a generic model that quantitatively describes the phase transition in the human maturation process of diverse human functions.
Collapse
Affiliation(s)
- Teh-Min Hu
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
6
|
Mørk ML, Andersen JT, Lausten-Thomsen U, Gade C. The Blind Spot of Pharmacology: A Scoping Review of Drug Metabolism in Prematurely Born Children. Front Pharmacol 2022; 13:828010. [PMID: 35242037 PMCID: PMC8886150 DOI: 10.3389/fphar.2022.828010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/25/2022] [Indexed: 12/30/2022] Open
Abstract
The limit for possible survival after extremely preterm birth has steadily improved and consequently, more premature neonates with increasingly lower gestational age at birth now require care. This specialized care often include intensive pharmacological treatment, yet there is currently insufficient knowledge of gestational age dependent differences in drug metabolism. This potentially puts the preterm neonates at risk of receiving sub-optimal drug doses with a subsequent increased risk of adverse or insufficient drug effects, and often pediatricians are forced to prescribe medication as off-label or even off-science. In this review, we present some of the particularities of drug disposition and metabolism in preterm neonates. We highlight the challenges in pharmacometrics studies on hepatic drug metabolism in preterm and particularly extremely (less than 28 weeks of gestation) preterm neonates by conducting a scoping review of published literature. We find that >40% of included studies failed to report a clear distinction between term and preterm children in the presentation of results making direct interpretation for preterm neonates difficult. We present summarized findings of pharmacokinetic studies done on the major CYP sub-systems, but formal meta analyses were not possible due the overall heterogeneous approaches to measuring the phase I and II pathways metabolism in preterm neonates, often with use of opportunistic sampling. We find this to be a testament to the practical and ethical challenges in measuring pharmacokinetic activity in preterm neonates. The future calls for optimized designs in pharmacometrics studies, including PK/PD modeling-methods and other sample reducing techniques. Future studies should also preferably be a collaboration between neonatologists and clinical pharmacologists.
Collapse
Affiliation(s)
- Mette Louise Mørk
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Jón Trærup Andersen
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christina Gade
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| |
Collapse
|
7
|
van Rongen A, Krekels EH, Calvier EA, de Wildt SN, Vermeulen A, Knibbe CA. An update on the use of allometric and other scaling methods to scale drug clearance in children: towards decision tables. Expert Opin Drug Metab Toxicol 2022; 18:99-113. [PMID: 35018879 DOI: 10.1080/17425255.2021.2027907] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION When pediatric data are not available for a drug, allometric and other methods are applied to scale drug clearance across the pediatric age-range from adult values. This is applied when designing first-in-child studies, but also for off-label drug prescription. AREAS COVERED This review provides an overview of the systematic accuracy of allometric and other pediatric clearance scaling methods compared to gold-standard PBPK predictions. The findings are summarized in decision tables to provide a priori guidance on the selection of appropriate pediatric clearance scaling methods for both novel drugs for which no pediatric data are available and existing drugs in clinical practice. EXPERT OPINION While allometric scaling principles are commonly used to scale pediatric clearance, there is no universal allometric exponent (i.e., 1, 0.75 or 0.67) that can accurately scale clearance for all drugs from adults to children of all ages. Therefore, pediatric scaling decision tables based on age, drug elimination route, binding plasma protein, fraction unbound, extraction ratio, and/or isoenzyme maturation are proposed to a priori select the appropriate (allometric) clearance scaling method, thereby reducing the need for full PBPK-based clearance predictions. Guidance on allometric scaling when estimating pediatric clearance values is provided as well.
Collapse
Affiliation(s)
- Anne van Rongen
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elke Hj Krekels
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elisa Am Calvier
- Sanofi Pharmacokinetics-Dynamics and Metabolism (PKDM), Translational Medicine and Early Development, Sanofi R&D, Montpellier, France
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands.,Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.,Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Catherijne Aj Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
8
|
Model-Informed Bayesian Estimation Improves the Prediction of Morphine Exposure in Neonates and Infants. Ther Drug Monit 2021; 42:778-786. [PMID: 32427759 DOI: 10.1097/ftd.0000000000000763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pain control in infants is an important clinical concern, with potential long-term adverse neurodevelopmental effects. Intravenous morphine is routinely administered for postoperative pain management; however, its dose-concentration-response relationship in neonates and infants has not been well characterized. Although the current literature provides dosing guidelines for the average infant, it fails to control for the large unexplained variability in morphine clearance and response in individual patients. Bayesian estimation can be used to control for some of this variability. The authors aimed to evaluate morphine pharmacokinetics (PKs) and exposure in critically ill neonates and infants receiving standard-of-care morphine therapy and compare a population-based approach to the model-informed Bayesian techniques. METHODS The PKs and exposure of morphine and its active metabolites were evaluated in a prospective opportunistic PK study using 221 discarded blood samples from 57 critically ill neonates and infants in the neonatal intensive care unit. Thereafter, a population-based PK model was compared with a Bayesian adaptive control strategy to predict an individual's PK profile and morphine exposure over time. RESULTS Among the critically ill neonates and infants, morphine clearance showed substantial variability with a 40-fold range (ie, 2.2 to 87.1, mean 23.7 L/h/70 kg). Compared with the observed morphine concentrations, the population-model based predictions had an R of 0.13, whereas the model-based Bayesian predictions had an R of 0.61. CONCLUSIONS Model-informed Bayesian estimation is a better predictor of morphine exposure than PK models alone in critically ill neonates and infants. A large variability was also identified in morphine clearance. A further study is warranted to elucidate the predictive covariates and precision dosing strategies that use morphine concentration and pain scores as feedbacks.
Collapse
|
9
|
Goulooze SC, de Kluis T, van Dijk M, Ceelie I, de Wildt SN, Tibboel D, Krekels EHJ, Knibbe CAJ. Quantifying the pharmacodynamics of morphine in the treatment of postoperative pain in preverbal children. J Clin Pharmacol 2021; 62:99-109. [PMID: 34383975 PMCID: PMC9293015 DOI: 10.1002/jcph.1952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/08/2021] [Indexed: 11/07/2022]
Abstract
While the pharmacokinetics of morphine in children have been studied extensively, little is known about the pharmacodynamics of morphine in this population. Here, we quantified the concentration‐effect relationship of morphine for postoperative pain in preverbal children between 0 and 3 years of age. For this, we applied item response theory modeling in the pharmacokinetic/pharmacodynamic analysis of COMFORT‐Behavior (COMFORT‐B) scale data from 2 previous clinical studies. In the model, we identified a sigmoid maximal efficacy model for the effect of morphine and found that in 26% of children, increasing morphine concentrations were not associated with lower pain scores (nonresponders to morphine up‐titration). In responders to morphine up‐titration, the COMFORT‐B score slowly decreases with increasing morphine concentrations at morphine concentrations >20 ng/mL. In nonresponding children, no decrease in COMFORT‐B score is expected. In general, lower baseline COMFORT‐B scores (2.1 points on average) in younger children (postnatal age <10.3 days) were found. Based on the model, we conclude that the percentage of children at a desirable COMFORT‐B score is maximized at a morphine concentration between 5 and 30 ng/mL for children aged <10 days, and between 5 and 40 ng/mL for children >10 days. These findings support a dosing regimen previously suggested by Krekels et al, which would put >95% of patients within this morphine target concentration range at steady state. Our modeling approach provides a promising platform for pharmacodynamic research of analgesics and sedatives in children.
Collapse
Affiliation(s)
- Sebastiaan C Goulooze
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,LAP&P Consultants BV, Leiden, The Netherlands
| | - Tirsa de Kluis
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Monique van Dijk
- Department of Pediatric Surgery, Erasmus University MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Section Nursing Science, Department of Internal Medicine, Erasmus University MC-, Rotterdam, The Netherlands
| | - Ilse Ceelie
- Department of Anesthesiology, University MC Utrecht-Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Saskia N de Wildt
- Department of Pediatric Surgery, Erasmus University MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pharmacology and Toxicology, Research Institute Health Sciences, Radboud University MC, Nijmegen, The Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Erasmus University MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Elke H J Krekels
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
10
|
Goulooze SC, Ista E, van Dijk M, Tibboel D, Krekels EHJ, Knibbe CAJ. Towards Evidence-Based Weaning: a Mechanism-Based Pharmacometric Model to Characterize Iatrogenic Withdrawal Syndrome in Critically Ill Children. AAPS JOURNAL 2021; 23:71. [PMID: 34002290 PMCID: PMC8128736 DOI: 10.1208/s12248-021-00586-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/22/2021] [Indexed: 11/30/2022]
Abstract
For the management of iatrogenic withdrawal syndrome (IWS) in children, a quantitative understanding of the dynamics of IWS of commonly used opioids and sedatives is lacking. Here, we introduce a new mechanism-based pharmacokinetic-pharmacodynamic (PKPD) modeling approach for studying IWS in pediatric clinical datasets. One thousand seven hundred eighty-two NRSwithdrawal scores of IWS severity were analyzed, which were collected from 81 children (age range: 1 month–18 years) that received opioids or sedatives by continuous infusion for 5 days or more. These data were successfully fitted with a PKPD model consisting of a plasma and a dependence compartment that well characterized the dynamics of IWS from morphine, fentanyl, and ketamine. The results suggest that (1) instead of decreasing the infusion rate by a set percentage at set intervals, it would be better to lengthen the weaning period when higher infusion rates are administered prior to weaning; (2) for fentanyl specifically, the risk of IWS might be lower when weaning with smaller dose reductions every 12 h instead of weaning with greater dose reductions every 48 h. The developed PKPD model can be used to evaluate the risk of IWS over time and the extent to which it is affected by different weaning strategies. The results yield hypotheses that could guide future clinical research on optimal weaning strategies. The mechanism-based PKPD modeling approach can be applied in other datasets to characterize the IWS dynamics of other drugs used in pediatric intensive care.
Collapse
Affiliation(s)
- Sebastiaan C Goulooze
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands.,LAP&P Consultants BV, Leiden, The Netherlands
| | - Erwin Ista
- Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Monique van Dijk
- Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Division of Nursing Science, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dick Tibboel
- Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Elke H J Krekels
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333, CC, Leiden, The Netherlands. .,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands.
| |
Collapse
|
11
|
Verscheijden LFM, Litjens CHC, Koenderink JB, Mathijssen RHJ, Verbeek MM, de Wildt SN, Russel FGM. Physiologically based pharmacokinetic/pharmacodynamic model for the prediction of morphine brain disposition and analgesia in adults and children. PLoS Comput Biol 2021; 17:e1008786. [PMID: 33661919 PMCID: PMC7963108 DOI: 10.1371/journal.pcbi.1008786] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/16/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Morphine is a widely used opioid analgesic, which shows large differences in clinical response in children, even when aiming for equivalent plasma drug concentrations. Age-dependent brain disposition of morphine could contribute to this variability, as developmental increase in blood-brain barrier (BBB) P-glycoprotein (Pgp) expression has been reported. In addition, age-related pharmacodynamics might also explain the variability in effect. To assess the influence of these processes on morphine effectiveness, a multi-compartment brain physiologically based pharmacokinetic/pharmacodynamic (PB-PK/PD) model was developed in R (Version 3.6.2). Active Pgp-mediated morphine transport was measured in MDCKII-Pgp cells grown on transwell filters and translated by an in vitro-in vivo extrapolation approach, which included developmental Pgp expression. Passive BBB permeability of morphine and its active metabolite morphine-6-glucuronide (M6G) and their pharmacodynamic parameters were derived from experiments reported in literature. Model simulations after single dose morphine were compared with measured and published concentrations of morphine and M6G in plasma, brain extracellular fluid (ECF) and cerebrospinal fluid (CSF), as well as published drug responses in children (1 day– 16 years) and adults. Visual predictive checks indicated acceptable overlays between simulated and measured morphine and M6G concentration-time profiles and prediction errors were between 1 and -1. Incorporation of active Pgp-mediated BBB transport into the PB-PK/PD model resulted in a 1.3-fold reduced brain exposure in adults, indicating only a modest contribution on brain disposition. Analgesic effect-time profiles could be described reasonably well for older children and adults, but were largely underpredicted for neonates. In summary, an age-appropriate morphine PB-PK/PD model was developed for the prediction of brain pharmacokinetics and analgesic effects. In the neonatal population, pharmacodynamic characteristics, but not brain drug disposition, appear to be altered compared to adults and older children, which may explain the reported differences in analgesic effect. Developmental processes in children can affect pharmacokinetics: “what the body does to the drug” as well as pharmacodynamics: “what the drug does to the body”. A typical example is morphine, of which the analgesic response is variable and particularly neonates suffer more often from respiratory depression, even when receiving doses corrected for differences in elimination. One way to mathematically incorporate developmental processes is by employing physiologically based pharmacokinetic/pharmacodynamic (PB-PK/PD) models, where physiological differences between individuals are incorporated. In this study, we developed a morphine PB-PK/PD model to predict brain drug disposition as well as analgesic response in adults and children, as both processes could potentially contribute to developmental variability in the effect of morphine. We found that age-related variation in BBB expression of the main morphine efflux transporter P-glycoprotein was not responsible for differences in brain exposure. In contrast, pharmacodynamic modelling suggested an increased sensitivity to morphine in neonates.
Collapse
Affiliation(s)
- Laurens F. M. Verscheijden
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carlijn H. C. Litjens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Jan B. Koenderink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marcel M. Verbeek
- Departments of Neurology and Laboratory Medicine, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Saskia N. de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Intensive Care and Department of Paediatric Surgery, Erasmus MC-Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Frans G. M. Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
12
|
van Hoogdalem MW, McPhail BT, Hahn D, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T. Pharmacotherapy of neonatal opioid withdrawal syndrome: a review of pharmacokinetics and pharmacodynamics. Expert Opin Drug Metab Toxicol 2020; 17:87-103. [PMID: 33049155 DOI: 10.1080/17425255.2021.1837112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Neonatal opioid withdrawal syndrome (NOWS) often arises in infants born to mothers who used opioids during pregnancy. Morphine, methadone, and buprenorphine are the most common first-line treatments, whereas clonidine and phenobarbital are generally reserved for adjunctive therapy. These drugs exhibit substantial pharmacokinetic (PK) and pharmacodynamic (PD) variability. Current pharmacological treatments for NOWS are based on institutional protocols and largely rely on empirical treatment of patient symptoms. AREAS COVERED This article reviews the PK/PD of NOWS pharmacotherapies with a focus on the implication of physiological development and maturation. Body size-standardized clearance is consistently low in neonates, except for methadone. This can be ascribed to underdeveloped metabolic and elimination pathways. The effects of pharmacogenetics have been clarified especially for morphine. The PK/PD relationship of medications used in the treatment of NOWS is generally understudied. EXPERT OPINION Providing an appropriate opioid dose in neonates is challenging. Advancements in quantitative pharmacology and PK/PD modeling approaches facilitate identification of key factors driving PK/PD variability and characterization of exposure-response relationships. PK/PD model-informed simulations have been widely employed to define age-appropriate pediatric dosing regimens. The model-informed approach holds promise to aid more rational use of medications in the treatment of NOWS.
Collapse
Affiliation(s)
- Matthijs W van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,James L. Winkle College of Pharmacy, University of Cincinnati , Cincinnati, OH, USA
| | - Brooks T McPhail
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,School of Medicine Greenville, University of South Carolina , Greenville, SC, USA
| | - David Hahn
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA
| | - Scott L Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Henry T Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| |
Collapse
|
13
|
Prediction of Clearance in Children from Adults Following Drug-Drug Interaction Studies: Application of Age-Dependent Exponent Model. Drugs R D 2020; 20:47-54. [PMID: 32056156 PMCID: PMC7067713 DOI: 10.1007/s40268-020-00295-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background and Objective Pharmacokinetic drug–drug interaction (DDI) studies are conducted in adult subjects during drug development but there are limited studies that have characterized pharmacokinetic DDI studies in children. The objective of this study was to evaluate if the DDI clearance values from adults can be allometrically extrapolated from adults to children. Methods Fifteen drugs were included in this study and the age of the children ranged from premature neonates to adolescents (30 observations across the age groups). The age-dependent exponent (ADE) model was used to predict the clearance of drugs in children from adults following DDI studies. Results The prediction error of drug clearances following DDIs in children ranged from 4 to 67%. Of 30 observations, 17 (57%) and 27 (90%) observations had a prediction error ≤ 30% and ≤ 50%, respectively. Conclusion This study indicates that it is possible to predict the clearance of drugs with reasonable accuracy in children from adults following DDI studies using an ADE model. The method is simple, robust, and reliable and can replace other complex empirical models.
Collapse
|
14
|
Extrapolation of Drug Clearance in Children ≤ 2 Years of Age from Empirical Models Using Data from Children (> 2 Years) and Adults. Drugs R D 2020; 20:1-10. [PMID: 31820365 PMCID: PMC7067721 DOI: 10.1007/s40268-019-00291-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The application of modeling and simulation approaches in clinical pharmacology studies has gained momentum over the last 20 years. OBJECTIVES The objective of this study was to develop six empirical models from clearance data obtained from children aged > 2 years and adults to evaluate the suitability of the models to predict drug clearance in children aged ≤ 2 years (preterm, term, and infants). METHODS Ten drugs were included in this study and administered intravenously: alfentanil, amikacin, busulfan, cefetamet, meperidine, oxycodone, propofol, sufentanil, theophylline, and tobramycin. These drugs were selected according to the availability of individual subjects' weight, age, and clearance data (concentration-time data for these drugs were not available to the author). The chosen drugs are eliminated by extensive metabolism by either the renal route or both the renal and hepatic routes. The six empirical models were (1) age and body weight-dependent sigmoidal maximum possible effect (Emax) maturation model, (2) body weight-dependent sigmoidal Emax model, (3) uridine 5'-diphospho [body weight-dependent allometric exponent model (BDE)], (4) age-dependent allometric exponent model (ADE), (5) a semi-physiological model, and (6) an allometric model developed from children aged > 2 years to adults. The model-predicted clearance values were compared with observed clearance values in an individual child. In this analysis, a prediction error of ≤ 50% for mean or individual clearance values was considered acceptable. RESULTS Across all age groups and the ten drugs, data for 282 children were compared between observed and model-predicted clearance values. The validation data consisted of 33 observations (sum of different age groups for ten drugs). Only three of the six models (body weight-dependent sigmoidal Emax model, ADE, and semi-physiological model) provided reasonably accurate predictions of clearance (> 80% observation with ≤ 50% prediction error) in children aged ≤ 2 years. In most instances, individual predicted clearance values were erratic (as indicated by % error) and were not in agreement with the observed clearance values. CONCLUSIONS The study indicated that simple empirical models can provide more accurate results than complex empirical models.
Collapse
|
15
|
Prediction of Clearance of Monoclonal and Polyclonal Antibodies and Non-Antibody Proteins in Children: Application of Allometric Scaling. Antibodies (Basel) 2020; 9:antib9030040. [PMID: 32764408 PMCID: PMC7551666 DOI: 10.3390/antib9030040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
Allometric scaling can be used for the extrapolation of pharmacokinetic parameters from adults to children. The objective of this study was to predict clearance of therapeutic proteins (monoclonal and polyclonal antibodies and non-antibody proteins) allometrically in preterm neonates to adolescents. There were 13 monoclonal antibodies, seven polyclonal antibodies, and nine therapeutic proteins (non-antibodies) in the study. The clearance of therapeutic proteins was predicted using the age dependent exponents (ADE) model and then compared with the observed clearance values. There were in total 29 therapeutic proteins in this study with 75 observations. The number of observations with ≤30%, ≤50%, and >50% prediction error was 60 (80%), 72 (96%), and 3 (4%), respectively. Overall, the predicted clearance values of therapeutic proteins in children was good. The allometric method proposed in this manuscript can be used to select first-in-pediatric dose of therapeutic proteins in pediatric clinical trials.
Collapse
|
16
|
Mizuno T, Dong M, Taylor ZL, Ramsey LB, Vinks AA. Clinical implementation of pharmacogenetics and model-informed precision dosing to improve patient care. Br J Clin Pharmacol 2020; 88:1418-1426. [PMID: 32529759 DOI: 10.1111/bcp.14426] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Providing maximal therapeutic efficacy without toxicity is a universal goal of rational drug therapy. However, substantial between-patient variability in drug response often impedes such successful treatments and brings the necessity of tailoring drug dose to individual needs for more precise therapy. In many cases plenty of patient characteristics, such as body size, genetic makeup and environmental factors, need to be taken into consideration to find the optimal dose in clinical practice. A pharmacokinetics and pharmacodynamics (PK/PD) model-informed approach offers integration of various patient information to provide an expectation of drug response and derive practical dose estimates to support clinicians' dosing decisions. Such an approach was pioneered in the late 1970s, but its broad clinical acceptance and implementation have been hampered by the lack of widespread computer technology, including user-friendly software tools. This has significantly changed in recent years. With the advent of electronic health records (EHRs) and the ubiquity of user-friendly software tools, we now experience a convergence of clinical information, pharmacogenetics, systems pharmacology and pharmacometrics, and technology. Advanced pharmacometrics research is now more appliable and implementable to improve health care. This article presents examples of successful development and implementation of pharmacogenetics-guided and PK/PD model-informed decision support to facilitate precision dosing, including the development of an EHR-embedded decision support tool. Through the integration of clinical decision support tools in EHRs, clinical pharmacometrics support can be brought directly to the clinical team and the bedside.
Collapse
Affiliation(s)
- Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zachary L Taylor
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laura B Ramsey
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Badée J, Fowler S, de Wildt SN, Collier AC, Schmidt S, Parrott N. The Ontogeny of UDP-glucuronosyltransferase Enzymes, Recommendations for Future Profiling Studies and Application Through Physiologically Based Pharmacokinetic Modelling. Clin Pharmacokinet 2020; 58:189-211. [PMID: 29862468 DOI: 10.1007/s40262-018-0681-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Limited understanding of drug pharmacokinetics in children is one of the major challenges in paediatric drug development. This is most critical in neonates and infants owing to rapid changes in physiological functions, especially in the activity of drug-metabolising enzymes. Paediatric physiologically based pharmacokinetic models that integrate ontogeny functions for cytochrome P450 enzymes have aided our understanding of drug exposure in children, including those under the age of 2 years. Paediatric physiologically based pharmacokinetic models have consequently been recognised by the European Medicines Agency and the US Food and Drug Administration as innovative tools in paediatric drug development and regulatory decision making. However, little is currently known about age-related changes in UDP-glucuronosyltransferase-mediated metabolism, which represents the most important conjugation reaction for xenobiotics. Therefore, the objective of the review was to conduct a thorough literature survey to summarise our current understanding of age-related changes in UDP-glucuronosyltransferases as well as associated clinical and experimental sources of variance. Our findings indicate that there are distinct differences in UDP-glucuronosyltransferase expression and activity between isoforms for different age groups. In addition, there is substantial variability between individuals and laboratories reported for human liver microsomes, which results in part from a lack of standardised experimental conditions. Therefore, we provide a number of best practice recommendations for experimental conditions, which ultimately may help improve the quality of data used for quantitative clinical pharmacology approaches, and thus for safe and effective pharmacotherapy in children.
Collapse
Affiliation(s)
- Justine Badée
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, FL, USA
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands.,Intensive Care and Department of Paediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Abby C Collier
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, FL, USA
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
18
|
Vinks AA, Punt NC, Menke F, Kirkendall E, Butler D, Duggan TJ, Cortezzo DE, Kiger S, Dietrich T, Spencer P, Keefer R, Setchell KD, Zhao J, Euteneuer JC, Mizuno T, Dufendach KR. Electronic Health Record-Embedded Decision Support Platform for Morphine Precision Dosing in Neonates. Clin Pharmacol Ther 2020; 107:186-194. [PMID: 31618453 PMCID: PMC7378965 DOI: 10.1002/cpt.1684] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
Morphine is the opioid most commonly used for neonatal pain management. In intravenous form, it is administered as continuous infusions and intermittent injections, mostly based on empirically established protocols. Inadequate pain control in neonates can cause long-term adverse consequences; however, providing appropriate individualized morphine dosing is particularly challenging due to the interplay of rapid natural physiological changes and multiple life-sustaining procedures in patients who cannot describe their symptoms. At most institutions, morphine dosing in neonates is largely carried out as an iterative process using a wide range of starting doses and then titrating to effect based on clinical response and side effects using pain scores and levels of sedation. Our background data show that neonates exhibit large variability in morphine clearance resulting in a wide range of exposures, which are poorly predicted by dose alone. Here, we describe the development and implementation of an electronic health record-integrated, model-informed decision support platform for the precision dosing of morphine in the management of neonatal pain. The platform supports pharmacokinetic model-informed dosing guidance and has functionality to incorporate real-time drug concentration information. The feedback is inserted directly into prescribers' workflows so that they can make data-informed decisions. The expected outcomes are better clinical efficacy and safety with fewer side effects in the neonatal population.
Collapse
Affiliation(s)
- Alexander A. Vinks
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Frank Menke
- Department Information Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Eric Kirkendall
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Wake Forest Center for Healthcare Innovation, Wake Forest School of Medicine, Winston-Salem, N.C
| | - Dawn Butler
- Division of Pharmacy, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Thomas J. Duggan
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - DonnaMaria E. Cortezzo
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pain and Palliative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sam Kiger
- Department Information Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Tom Dietrich
- Department of Interactive Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | | | - Rob Keefer
- Pomiet, Health IT Systems, Cincinnati, OH, USA
| | - Kenneth D.R. Setchell
- Division of Pathology and Laboratory Medicine, Clinical Mass Spectrometry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH. USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Junfang Zhao
- Division of Pathology and Laboratory Medicine, Clinical Mass Spectrometry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH. USA
| | | | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Children’s Hospital & Medical Center and Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin R. Dufendach
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
19
|
Cravero JP, Agarwal R, Berde C, Birmingham P, Coté CJ, Galinkin J, Isaac L, Kost‐Byerly S, Krodel D, Maxwell L, Voepel‐Lewis T, Sethna N, Wilder R. The Society for Pediatric Anesthesia recommendations for the use of opioids in children during the perioperative period. Paediatr Anaesth 2019; 29:547-571. [PMID: 30929307 PMCID: PMC6851566 DOI: 10.1111/pan.13639] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022]
Abstract
Opioids have long held a prominent role in the management of perioperative pain in adults and children. Published reports concerning the appropriate, and inappropriate, use of these medications in pediatric patients have appeared in various publications over the last 50 years. For this document, the Society for Pediatric Anesthesia appointed a taskforce to evaluate the available literature and formulate recommendations with respect to the most salient aspects of perioperative opioid administration in children. The recommendations are graded based on the strength of the available evidence, with consensus of the experts applied for those issues where evidence is not available. The goal of the recommendations was to address the most important issues concerning opioid administration to children after surgery, including appropriate assessment of pain, monitoring of patients on opioid therapy, opioid dosing considerations, side effects of opioid treatment, strategies for opioid delivery, and assessment of analgesic efficacy. Regular updates are planned with a re-release of guidelines every 2 years.
Collapse
Affiliation(s)
- Joseph P. Cravero
- Department of Anesthesiology, Critical Care, and Pain MedicineBoston Children's Hospital, Harvard Medical SchoolBostonMassachusetts
| | - Rita Agarwal
- Pediatric Anesthesiology DepartmentLucille Packard Children's Hospital, Stanford University Medical SchoolStanfordCalifornia
| | - Charles Berde
- Department of Anesthesiology, Critical Care, and Pain MedicineBoston Children's Hospital, Harvard Medical SchoolBostonMassachusetts
| | - Patrick Birmingham
- Department of AnesthesiologyAnn and Robert H. Lurie Children's Hospital Northwestern University Feinberg School of MedicineEvanstonIllinois
| | - Charles J. Coté
- Department of AnesthesiologyMass General Hospital for Children, Harvard UniversityBostonMassachusetts
| | - Jeffrey Galinkin
- Anesthesiology DepartmentChildren's Hospital of Colorado, University of ColoradoAuroraColorado
| | - Lisa Isaac
- Department of Anesthesia and Pain MedicineHospital for Sick Children, University of TorontoTorontoOntarioCanada
| | - Sabine Kost‐Byerly
- Pediatric Anesthesiology and Critical Care MedicineJohns Hopkins University HospitalBaltimoreMaryland
| | - David Krodel
- Department of AnesthesiologyAnn and Robert H. Lurie Children's Hospital Northwestern University Feinberg School of MedicineEvanstonIllinois
| | - Lynne Maxwell
- Department of Aneshtesiology and Critical Care MedicineChildren's Hospital of Philadelphia, Perelman School of Medicine at the University of PennsylvaniaPhiladelphia
| | - Terri Voepel‐Lewis
- Department of AneshteiologyC. S. Mott Children's Hospital, University of Michigan Medical SchoolAnn ArborMichigan
| | - Navil Sethna
- Department of Anesthesiology, Critical Care, and Pain MedicineBoston Children's Hospital, Harvard Medical SchoolBostonMassachusetts
| | - Robert Wilder
- Department of Anesthesiology and Perioperative MedicineMayo ClinicRochesterMinnesota
| |
Collapse
|
20
|
Age- and Bodyweight-dependent Allometric Exponent Model for Scaling Clearance and Maintenance Dose of Theophylline From Neonates to Adults. Ther Drug Monit 2019; 40:635-641. [PMID: 30086085 DOI: 10.1097/ftd.0000000000000543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND In population pharmacokinetic modeling, bodyweight is often incorporated as an important covariate using fixed (0.75) or single-exponent model. In recent years, several variations of allometric models have been suggested for the prediction of drug clearance across a wide age range. The objective of this study is to develop and evaluate single-exponent, bodyweight-dependent allometric exponent (BDE), age-dependent exponent (ADE), and segmented regression models for predicting clearance and maintenance dose of theophylline. METHODS The BDE model was described by the following equation: (Equation is included in full-text article.), where L × BW defines the BDE for clearance. The coefficient and the exponents L and M were estimated. The ADE model consisted of several empirical exponents based on age and ranged from 0.75 (children >5 years and adults) to 1.2 (premature neonates). Data for model development and validation were based on 52 subjects each. RESULTS All structural and statistical parameters were estimated with acceptable precision for single-exponent and BDE models (<30%); however, the BDE model was superior in describing theophylline clearance across a wide age range for the training data. The segmented regression model on log-transformed data also adequately described theophylline clearance. When models were evaluated with validation data, a single-exponent model overpredicted clearance and dosing rate in premature neonates and adults with a mean prediction error of ≥50%. For premature neonates and adults, mean clearance and dosing rate were predicted within a 30% prediction error using the BDE, ADE, and segmented models. CONCLUSIONS This study demonstrates that the BDE, ADE, and segmented models performed better than a single-exponent model for predicting clearance and dose of theophylline across a wide age range.
Collapse
|
21
|
Favié LMA, Groenendaal F, van den Broek MPH, Rademaker CMA, de Haan TR, van Straaten HLM, Dijk PH, van Heijst A, Dudink J, Dijkman KP, Rijken M, Zonnenberg IA, Cools F, Zecic A, van der Lee JH, Nuytemans DHGM, van Bel F, Egberts TCG, Huitema ADR, on behalf of the PharmaCool study group. Pharmacokinetics of morphine in encephalopathic neonates treated with therapeutic hypothermia. PLoS One 2019; 14:e0211910. [PMID: 30763356 PMCID: PMC6375702 DOI: 10.1371/journal.pone.0211910] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Morphine is a commonly used drug in encephalopathic neonates treated with therapeutic hypothermia after perinatal asphyxia. Pharmacokinetics and optimal dosing of morphine in this population are largely unknown. The objective of this study was to describe pharmacokinetics of morphine and its metabolites morphine-3-glucuronide and morphine-6-glucuronide in encephalopathic neonates treated with therapeutic hypothermia and to develop pharmacokinetics based dosing guidelines for this population. STUDY DESIGN Term and near-term encephalopathic neonates treated with therapeutic hypothermia and receiving morphine were included in two multicenter cohort studies between 2008-2010 (SHIVER) and 2010-2014 (PharmaCool). Data were collected during hypothermia and rewarming, including blood samples for quantification of morphine and its metabolites. Parental informed consent was obtained for all participants. RESULTS 244 patients (GA mean (sd) 39.8 (1.6) weeks, BW mean (sd) 3,428 (613) g, male 61.5%) were included. Morphine clearance was reduced under hypothermia (33.5°C) by 6.89%/°C (95% CI 5.37%/°C- 8.41%/°C, p<0.001) and metabolite clearance by 4.91%/°C (95% CI 3.53%/°C- 6.22%/°C, p<0.001) compared to normothermia (36.5°C). Simulations showed that a loading dose of 50 μg/kg followed by continuous infusion of 5 μg/kg/h resulted in morphine plasma concentrations in the desired range (between 10 and 40 μg/L) during hypothermia. CONCLUSIONS Clearance of morphine and its metabolites in neonates is affected by therapeutic hypothermia. The regimen suggested by the simulations will be sufficient in the majority of patients. However, due to the large interpatient variability a higher dose might be necessary in individual patients to achieve the desired effect. TRIAL REGISTRATION www.trialregister.nl NTR2529.
Collapse
Affiliation(s)
- Laurent M. A. Favié
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marcel P. H. van den Broek
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Carin M. A. Rademaker
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Timo R. de Haan
- Department of Neonatology, Emma Children’s Hospital, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Peter H. Dijk
- Department of Neonatology, Groningen University Medical Centre, Groningen, the Netherlands
| | - Arno van Heijst
- Department of Neonatology, Radboud university medical center-Amalia Children’s Hospital, Nijmegen, the Netherlands
| | - Jeroen Dudink
- Department of Pediatrics, Division of Neonatology, Erasmus Medical Centre-Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Koen P. Dijkman
- Department of Neonatology, Máxima Medical Center Veldhoven, Veldhoven, the Netherlands
| | - Monique Rijken
- Department of Neonatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge A. Zonnenberg
- Department of Neonatology, VU University Medical Center, Amsterdam, the Netherlands
| | - Filip Cools
- Department of Neonatology, UZ Brussel—Vrije Universiteit Brussel, Brussels, Belgium
| | - Alexandra Zecic
- Department of Neonatology, University Hospital Gent, Gent, Belgium
| | - Johanna H. van der Lee
- Paediatric Clinical Research Office, Emma Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Toine C. G. Egberts
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Alwin D. R. Huitema
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
22
|
Anderson BJ, Lerman J, Coté CJ. Pharmacokinetics and Pharmacology of Drugs Used in Children. A PRACTICE OF ANESTHESIA FOR INFANTS AND CHILDREN 2019:100-176.e45. [DOI: 10.1016/b978-0-323-42974-0.00007-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Mahmood I, Tegenge MA. A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling for the Prediction of Drug Clearance From Neonates to Adolescents. J Clin Pharmacol 2018; 59:189-197. [DOI: 10.1002/jcph.1310] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/09/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Iftekhar Mahmood
- Office of Tissue & Advanced Therapies; Center for Biologics Evaluation and Research; Food & Drug Administration; Silver Spring MD USA
| | - Million A. Tegenge
- Office of Biostatistics & Epidemiology; Center for Biologics Evaluation and Research; Food & Drug Administration; Silver Spring MD USA
| |
Collapse
|
24
|
Mahmood I. Misconceptions and issues regarding allometric scaling during the drug development process. Expert Opin Drug Metab Toxicol 2018; 14:843-854. [PMID: 29999428 DOI: 10.1080/17425255.2018.1499725] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Allometry is the study of size and its consequences. The simple hypothesis of allometric scaling is that all physiological parameters are proportional to body size or body mass. This review examines the development of theory-based allometry or fixed exponents (0.75 and 1.0 for basal metabolic rate and volume, respectively) and the evidence for or against the theory. The main focus of this report is to show the readers that there is enough evidence from experimental data that negate the concept of theory-based allometry in biology, physiology, and pharmacokinetics. Areas covered: In this review, the history of the development of theoretical allometry and the strong evidence against theory-based allometry demonstrated by experimental data is provided. During drug development, allometry is applied to both inter-species (from animals to humans) and intra-species (adults to children) scaling. These two forms of allometric scaling in the context of theory-based allometry are discussed and provide insight on scientific progress that refute theory-based allometry. Expert opinion: Theory-based allometry is a mere theory and experimental data and real-life observations strongly negate the existence of such a theory. Pharmacostatistical and physiological models based on theory-based allometry can be misleading and incorrect because the theory-based allometric exponent 0.75 is not universal. The exponents of allometry are data dependent and are not fixed in the universe.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- a Office of Tissue & Advance Therapies (OTAT) , Center for Biologics Evaluation and Research, Food & Drug Administration , Silver Spring , MD , USA
| |
Collapse
|
25
|
Zeilmaker-Roest GA, van Rosmalen J, van Dijk M, Koomen E, Jansen NJG, Kneyber MCJ, Maebe S, van den Berghe G, Vlasselaers D, Bogers AJJC, Tibboel D, Wildschut ED. Intravenous morphine versus intravenous paracetamol after cardiac surgery in neonates and infants: a study protocol for a randomized controlled trial. Trials 2018; 19:318. [PMID: 29895289 PMCID: PMC5998570 DOI: 10.1186/s13063-018-2705-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/24/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Morphine is worldwide the analgesic of first choice after cardiac surgery in children. Morphine has unwanted hemodynamic and respiratory side effects. Therefore, post-cardiac surgery patients may potentially benefit from a non-opioid drug for pain relief. A previous study has shown that intravenous (IV) paracetamol is effective and opioid-sparing in children after major non-cardiac surgery. The aim of the study is to test the hypothesis that intermittent IV paracetamol administration in children after cardiac surgery will result in a reduction of at least 30% of the cumulative morphine requirement. METHODS This is a prospective, multi-center, randomized controlled trial at four level-3 pediatric intensive care units (ICUs) in the Netherlands and Belgium. Children who are 0-36 months old will be randomly assigned to receive either intermittent IV paracetamol or continuous IV morphine up to 48 h post-operatively. Morphine will be available as rescue medication for both groups. Validated pain and sedation assessment tools will be used to monitor patients. The sample size (n = 208, 104 per arm) was calculated in order to detect a 30% reduction in morphine dose; two-sided significance level was 5% and power was 95%. DISCUSSION This study will focus on the reduction, or replacement, of morphine by IV paracetamol in children (0-36 months old) after cardiac surgery. The results of this study will form the basis of a new pain management algorithm and will be implemented at the participating ICUs, resulting in an evidence-based guideline on post-operative pain after cardiac surgery in infants who are 0-36 months old. TRIAL REGISTRATION Dutch Trial Registry ( www.trialregister.nl ): NTR5448 on September 1, 2015. Institutional review board approval (MEC2015-646), current protocol version: July 3, 2017.
Collapse
Affiliation(s)
- Gerdien A Zeilmaker-Roest
- Department of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands. .,Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, The Netherlands.
| | | | - Monique van Dijk
- Department of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Erik Koomen
- Department of Pediatric Intensive Care, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nicolaas J G Jansen
- Department of Pediatric Intensive Care, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martin C J Kneyber
- Department of Pediatrics, division of Pediatric Critical Care Medicine, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Sofie Maebe
- Department of Intensive Care Medicine, UZ Leuven, Leuven, Belgium
| | | | - Dirk Vlasselaers
- Department of Intensive Care Medicine, UZ Leuven, Leuven, Belgium
| | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Dick Tibboel
- Department of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Enno D Wildschut
- Department of Pediatric Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Mahmood I. Prediction of Clearance, Volume of distribution, and Half-life of Drugs in Extremely Low to Low Birth Weight Neonates: An Allometric Approach. Eur J Drug Metab Pharmacokinet 2018; 42:601-610. [PMID: 27562171 DOI: 10.1007/s13318-016-0372-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES More than 20 million infants worldwide (15.5 % of all births) are born with low birth weight. Low birth weight is associated with poor growth in childhood and a higher incidence of adult diseases, such as type 2 diabetes, hypertension and cardiovascular disease. The objective of this study was to evaluate the predictive performance of allometric models to predict clearance, volume of distribution, and half-life in extremely low to low birth weight neonates (<1 to 2.5 kg body weight). METHODS Several allometric models were used to predict clearance (4 models), volume of distribution (2 models), and half-life (2 models) in extremely low to low birth weight neonates. From the literature, clearance, volume of distribution, and half-life values for 16 drugs for these neonates were obtained. The predictive performance of these allometric models was evaluated by comparing the predicted values of the aforementioned pharmacokinetic parameters with the observed pharmacokinetic parameters in an individual neonate. For the evaluation of the predictive performance of these allometric models, there were 16 drugs with 36 (n = 279), 34 (n = 261), and 31 (n = 197) weight groups for clearance, volume of distribution, and half-life, respectively. RESULTS The prediction error of ≤50 % for mean clearance, volume of distribution, and half-life values were 69, 79, and 58 %, respectively, by proposed allometric models. The prediction error of ≤50 % for mean clearance, volume of distribution, and half-life values by theoretical allometric exponents were 0 % (exponent = 0.75), 71 % (exponent = 1.0), and 0 % (exponent = 0.25), respectively. In this analysis, out of 16 drugs, there were three drugs (ibuprofen, zidovudine, and buprenorphine) which are metabolized by glucuronidation and one drug (furosemide) is renally secreted. The predicted clearances of these four drugs were substantially higher by the proposed allometric methods. It seems that drugs of these physiological characteristics may require different method(s) to improve the prediction of clearance in the neonates. CONCLUSIONS Overall, the proposed allometric methods can predict mean pharmacokinetic parameters of drugs in extremely low to low birth weight neonates with reasonable accuracy and are of practical value during neonatal drug development.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology Clinical Review, Office of Blood Review and Research (OBRR), Center for Biologic Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
27
|
Zeilmaker GA, Pokorna P, Mian P, Wildschut ED, Knibbe CAJ, Krekels EHJ, Allegaert K, Tibboel D. Pharmacokinetic considerations for pediatric patients receiving analgesia in the intensive care unit; targeting postoperative, ECMO and hypothermia patients. Expert Opin Drug Metab Toxicol 2018; 14:417-428. [PMID: 29623729 DOI: 10.1080/17425255.2018.1461836] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Adequate postoperative analgesia in pediatric patients in the intensive care unit (ICU) matters, since untreated pain is associated with negative outcomes. Compared to routine postoperative patients, children undergoing hypothermia (HT) or extracorporeal membrane oxygenation (ECMO), or recovering after cardiac surgery likely display non-maturational differences in pharmacokinetics (PK) and pharmacodynamics (PD). These differences warrant additional dosing recommendations to optimize pain treatment. Areas covered: Specific populations within the ICU will be discussed with respect to expected variations in PK and PD for various analgesics. We hereby move beyond maturational changes and focus on why PK/PD may be different in children undergoing HT, ECMO or cardiac surgery. We provide a stepwise manner to develop PK-based dosing regimens using population PK approaches in these populations. Expert opinion: A one-dose to size-fits-all for analgesia is suboptimal, but for several commonly used analgesics the impact of HT, ECMO or cardiac surgery on average PK parameters in children is not yet sufficiently known. Parameters considering both maturational and non-maturational covariates are important to develop population PK-based dosing advices as part of a strategy to optimize pain treatment.
Collapse
Affiliation(s)
- Gerdien A Zeilmaker
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Paula Pokorna
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,b Department of Pediatrics, General Faculty Hospital Prague, First Faculty of Medicine , Charles University and General University Hospital in Prague , Prague , Czech Republic.,c Institute of Pharmacology, First Faculty of Medicine , Charles University and General University Hospital in Prague , Prague , Czech Republic
| | - Paola Mian
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Enno D Wildschut
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Catherijne A J Knibbe
- d Division of Pharmacology , LACDR, Leiden University , Leiden , The Netherlands.,e Department of Clinical Pharmacy , St. Antonius Hospital , Nieuwegein , The Netherlands
| | - Elke H J Krekels
- d Division of Pharmacology , LACDR, Leiden University , Leiden , The Netherlands
| | - Karel Allegaert
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,f Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Dick Tibboel
- a Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| |
Collapse
|
28
|
Tegenge MA, Mahmood I, Jiang Z, Forshee R. Multistep Unified Models Using Prior Knowledge for the Prediction of Drug Clearance in Neonates and Infants. J Clin Pharmacol 2018; 58:877-884. [PMID: 29489016 DOI: 10.1002/jcph.1089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022]
Abstract
Allometric approaches are widely used for interspecies scaling for the prediction of pharmacokinetic (PK) parameters during drug development. The concept of allometry can also be extended to predict PK parameters from adults to children. Three methods for extrapolating pediatric clearance were developed and evaluated using the clearance values of 4 drugs. The first method was established using a simple allometric (SA) model with estimated coefficient and exponent based on data ranging from children older than 2 years to adult. Then we developed a unified multistep single-exponent (MSE) and multistep body-weight-dependent exponent (MBDE) models. The major steps in these 2 new methods include generating pseudopredicted clearance for unobserved new populations such as preterm neonates, term neonates, and infants. Subsequent steps involve incorporating the pseudopredicted clearance with the actual PK data from older children and adults. All 3 models were then used to predict drug clearance in children ≤2 years old (N = 278). Drug clearance was predicted with mean absolute error of 29.6, 14.2, and 12.9 using SA, MSE, MBDE, respectively. The root mean square error was 65.9, 29.8, 24.7 for SA, MSE, MBDE, respectively. Approximately 41%, 72%, and 74% of the children's clearance data were within 0.5 to 1.5-fold of the observed values when drug clearance was extrapolated using SA, MSE, and MBDE models, respectively. The present multistep unified extrapolation approaches improved the prediction of clearance from preterm neonates to 2 years of age and may have practical use for first-in-pediatric dose selection.
Collapse
Affiliation(s)
- Million A Tegenge
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, Food & Drug Administration, Silver Spring, MD, USA
| | - Iftekhar Mahmood
- Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissue and Advanced Therapies (OTAT), Center for Biologics Evaluation and Research, Food & Drug Administration, Silver Spring, MD, USA
| | - Zhen Jiang
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, Food & Drug Administration, Silver Spring, MD, USA
| | - Richard Forshee
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, Food & Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
29
|
Van Donge T, Mian P, Tibboel D, Van Den Anker J, Allegaert K. Drug metabolism in early infancy: opioids as an illustration. Expert Opin Drug Metab Toxicol 2018; 14:287-301. [PMID: 29363349 DOI: 10.1080/17425255.2018.1432595] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Drug dosing in infants frequently depends on body weight as a crude indicator for maturation. Fentanyl (metabolized by Cytochrome P450 3A4) and morphine (glucuronidated by UDP-glucuronosyltransferase-2B7) served as model drugs to provide insight in maturation patterns of these enzymes and provide understanding of the impact of non-maturational factors to optimize dosing in infants. Areas covered: Systematic searches on metabolism and population pharmacokinetic (Pop-PK) models for fentanyl and morphine were performed. Pre- and post-model selection criteria were applied to assess and evaluate the validity of these models. It was observed that maturational changes have been rather well investigated, be it with variability in the maturational function estimates. The same holds true for Pop-PK models, where non-maturational covariates have also been reported (pharmacogenetics, disease state or external influences), although less incorporated in the PK models and with limited knowledge on mechanisms involved. Expert opinion: PK models for fentanyl and morphine are currently available. Consequently, we suggest that researchers should not continue to develop new models, but should investigate whether collected data fit in already existing models and provide additional value concerning the impact of (non)-maturational factors like drug-drug interactions or pharmacogenetics.
Collapse
Affiliation(s)
- Tamara Van Donge
- a Intensive Care and Department of Paediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,b Systems Biomedicine and Pharmacology , LACDR, Leiden University , Leiden , The Netherlands
| | - Paola Mian
- a Intensive Care and Department of Paediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Dick Tibboel
- a Intensive Care and Department of Paediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - John Van Den Anker
- a Intensive Care and Department of Paediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,c Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland.,d Division of Clinical Pharmacology , Children's National Health System , Washington , DC , USA
| | - Karel Allegaert
- a Intensive Care and Department of Paediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,e Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| |
Collapse
|
30
|
Mahmood I, Tegenge MA. Population Pharmacokinetics: Some Observations in Pediatric Modeling for Drug Clearance. Clin Pharmacokinet 2017; 56:1567-1576. [PMID: 28405936 DOI: 10.1007/s40262-017-0542-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The objective of this study is to evaluate the predictive performance of several models to predict drug clearance in preterm and term neonates. Five models using different types of allometric and linear models were developed. Two sets of data were used to develop these models (data from preterm neonates to adults and data from preterm and term neonates). Models were also developed with (normalized to 70 kg) or without body weight normalization (body weight 1 kg). From the literature, clearance values for four drugs from neonates to adults were obtained. External data were used to evaluate the predictive performance of these models in preterm and term neonates. The results of the study indicated that (1) normalization to a standard body weight had no impact on the predictive performance of the models, (2) the model developed from preterm neonates to adults using fixed exponent 0.75 provided inaccurate estimate (overestimation) of drug clearance in neonates, (3) a far superior prediction of clearance was observed with the model when the exponents of allometry were estimated than the model using exponent 0.75, (4) linear models with the exception of the model with intercept provided comparable results to the estimated exponent model and were superior in their predictive performance to the model using exponent 0.75, and (5) when the models were developed from neonate data, the predictive performance of all models were similar. Overall, the study indicated that body weight normalization had no impact on the performance of model prediction, the exponents of allometry in pharmacostatistical models should be estimated rather than fixed, and more studies are needed to evaluate the suitability of linear models for the prediction of drug clearance in neonates.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Office of Tissue and Advance Therapeutics, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993-0002, USA.
| | - Million A Tegenge
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
31
|
Calvier EAM, Krekels EHJ, Välitalo PAJ, Rostami-Hodjegan A, Tibboel D, Danhof M, Knibbe CAJ. Allometric Scaling of Clearance in Paediatric Patients: When Does the Magic of 0.75 Fade? Clin Pharmacokinet 2017; 56:273-285. [PMID: 27510367 PMCID: PMC5315734 DOI: 10.1007/s40262-016-0436-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Allometric scaling on the basis of bodyweight raised to the power of 0.75 (AS0.75) is frequently used to scale size-related changes in plasma clearance (CLp) from adults to children. A systematic assessment of its applicability is undertaken for scenarios considering size-related changes with and without maturation processes. A physiologically-based pharmacokinetic (PBPK) simulation workflow was developed in R for 12,620 hypothetical drugs. In scenario one, only size-related changes in liver weight, hepatic blood flow, and glomerular filtration were included in simulations of 'true' paediatric CLp. In a second scenario, maturation in unbound microsomal intrinsic clearance (CLint,mic), plasma protein concentration, and haematocrit were also included in these simulated 'true' paediatric CLp values. For both scenarios, the prediction error (PE) of AS0.75-based paediatric CLp predictions was assessed, while, for the first scenario, an allometric exponent was also estimated based on 'true' CLp. In the first scenario, the PE of AS0.75-based paediatric CLp predictions reached up to 278 % in neonates, and the allometric exponent was estimated to range from 0.50 to 1.20 depending on age and drug properties. In the second scenario, the PE sensitivity to drug properties and maturation was higher in the youngest children, with AS0.75 resulting in accurate CLp predictions above 5 years of age. Using PBPK principles, there is no evidence for one unique allometric exponent in paediatric patients, even in scenarios that only consider size-related changes. As PE is most sensitive to the allometric exponent, drug properties and maturation in younger children, AS0.75 leads to increasingly worse predictions with decreasing age.
Collapse
Affiliation(s)
- Elisa A M Calvier
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Laboratories, Einsteinweg, 552333 CC, Leiden, The Netherlands
| | - Elke H J Krekels
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Laboratories, Einsteinweg, 552333 CC, Leiden, The Netherlands
| | - Pyry A J Välitalo
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Laboratories, Einsteinweg, 552333 CC, Leiden, The Netherlands
| | | | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Laboratories, Einsteinweg, 552333 CC, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Laboratories, Einsteinweg, 552333 CC, Leiden, The Netherlands.
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands.
| |
Collapse
|
32
|
Maturation of Oxycodone Pharmacokinetics in Neonates and Infants: a Population Pharmacokinetic Model of Three Clinical Trials. Pharm Res 2017; 34:1125-1133. [DOI: 10.1007/s11095-017-2122-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
|
33
|
Zeilmaker-Roest GA, Wildschut ED, van Dijk M, Anderson BJ, Breatnach C, Bogers AJJC, Tibboel D. An international survey of management of pain and sedation after paediatric cardiac surgery. BMJ Paediatr Open 2017; 1:e000046. [PMID: 29637103 PMCID: PMC5862168 DOI: 10.1136/bmjpo-2017-000046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE The mainstay of pain treatment after paediatric cardiac surgery is the use of opioids. Current guidelines for its optimal use are based on small, non-randomised clinical trials, and data on the pharmacokinetics (PK) and pharmacodynamics (PD) of opioids are lacking. This study aims at providing an overview of international hospital practices on the treatment of pain and sedation after paediatric cardiac surgery. DESIGN A multicentre survey study assessed the management of pain and sedation in children aged 0-18 years after cardiac surgery. SETTING Pediatric intensive care units (PICU)of 19 tertiary children's hospitals worldwide were invited to participate. The focus of the survey was on type and dose of analgesic and sedative drugs and the tools used for their pharmacodynamic assessment. RESULTS Fifteen hospitals (response rate 79%) filled out the survey. Morphine was the primary analgesic in most hospitals, and its doses for continuous infusion ranged from 10 to 60 mcg kg-1 h-1 in children aged 0-36 months. Benzodiazepines were the first choice for sedation, with midazolam used in all study hospitals. Eight hospitals (53%) reported routine use of sedatives with pain treatment. Overall, type and dosing of analgesic and sedative drugs differed substantially between hospitals. All participating hospitals used validated pain and sedation assessment tools. CONCLUSION There was a large variation in the type and dosing of drugs employed in the treatment of pain and sedation after paediatric cardiac surgery. As a consequence, there is a need to rationalise pain and sedation management for this vulnerable patient group.
Collapse
Affiliation(s)
- Gerdien A Zeilmaker-Roest
- Intensive Care, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands.,Cardio-Thoracic Surgery, Erasmus MC, Rotterdam, South Holland, The Netherlands
| | - Enno D Wildschut
- Intensive Care, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Monique van Dijk
- Intensive Care, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Brian J Anderson
- Intensive Care, Starship Children's Hospital, Auckland, New Zealand
| | - Cormac Breatnach
- Intensive Care, Our Lady's Children's Hospital, Crumlin, Ireland
| | - Ad J J C Bogers
- Cardio-Thoracic Surgery, Erasmus MC, Rotterdam, South Holland, The Netherlands
| | - Dick Tibboel
- Intensive Care, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | | |
Collapse
|
34
|
Germovsek E, Barker CIS, Sharland M, Standing JF. Scaling clearance in paediatric pharmacokinetics: All models are wrong, which are useful? Br J Clin Pharmacol 2016; 83:777-790. [PMID: 27767204 PMCID: PMC5346879 DOI: 10.1111/bcp.13160] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/06/2016] [Accepted: 10/15/2016] [Indexed: 12/11/2022] Open
Abstract
Linked Articles This article is commented on in the editorial by Holford NHG and Anderson BJ. Why standards are useful for predicting doses. Br J Clin Pharmacol 2017; 83: 685–7. doi: 10.1111/bcp.13230 Aim When different models for weight and age are used in paediatric pharmacokinetic studies it is difficult to compare parameters between studies or perform model‐based meta‐analyses. This study aimed to compare published models with the proposed standard model (allometric weight0.75 and sigmoidal maturation function). Methods A systematic literature search was undertaken to identify published clearance (CL) reports for gentamicin and midazolam and all published models for scaling clearance in children. Each model was fitted to the CL values for gentamicin and midazolam, and the results compared with the standard model (allometric weight exponent of 0.75, along with a sigmoidal maturation function estimating the time in weeks of postmenstrual age to reach half the mature value and a shape parameter). For comparison, we also looked at allometric size models with no age effect, the influence of estimating the allometric exponent in the standard model and, for gentamicin, using a fixed allometric exponent of 0.632 as per a study on glomerular filtration rate maturation. Akaike information criteria (AIC) and visual predictive checks were used for evaluation. Results No model gave an improved AIC in all age groups, but one model for gentamicin and three models for midazolam gave slightly improved global AIC fits albeit using more parameters: AIC drop (number of parameters), –4.1 (5), –9.2 (4), –10.8 (5) and –10.1 (5), respectively. The 95% confidence interval of estimated CL for all top performing models overlapped. Conclusion No evidence to reject the standard model was found; given the benefits of standardised parameterisation, its use should therefore be recommended.
Collapse
Affiliation(s)
- Eva Germovsek
- Inflammation, Infection and Rheumatology Section, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Charlotte I S Barker
- Inflammation, Infection and Rheumatology Section, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.,St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, UK
| | - Mike Sharland
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.,St George's University Hospitals NHS Foundation Trust, Blackshaw Road, London, UK
| | - Joseph F Standing
- Inflammation, Infection and Rheumatology Section, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| |
Collapse
|
35
|
Mahmood I, Tegenge MA. A bodyweight-dependent allometric exponent model for scaling clearance of clotting factor VIII and IX from infants to adults. Haemophilia 2016; 22:e570-e573. [DOI: 10.1111/hae.13116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2016] [Indexed: 12/26/2022]
Affiliation(s)
- I. Mahmood
- Division of Hematology Clinical Review Branch; Office of Blood Review & Research (OBRR); Center for Biologics Evaluation and Research Food & Drug Administration; Silver Spring MD USA
| | - M. A. Tegenge
- Office of Biostatistics & Epidemiology; Center for Biologics Evaluation and Research Food & Drug Administration; Silver Spring MD USA
| |
Collapse
|
36
|
Baarslag MA, Allegaert K, Van Den Anker JN, Knibbe CAJ, Van Dijk M, Simons SHP, Tibboel D. Paracetamol and morphine for infant and neonatal pain; still a long way to go? Expert Rev Clin Pharmacol 2016; 10:111-126. [PMID: 27785937 DOI: 10.1080/17512433.2017.1254040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pharmacologic pain management in newborns and infants is often based on limited scientific data. To close the knowledge gap, drug-related research in this population is increasingly supported by the authorities, but remains very challenging. This review summarizes the challenges of analgesic studies in newborns and infants on morphine and paracetamol (acetaminophen). Areas covered: Aspects such as the definition and multimodal character of pain are reflected to newborn infants. Specific problems addressed include defining pharmacodynamic endpoints, performing clinical trials in this population and assessing developmental changes in both pharmacokinetics and pharmacodynamics. Expert commentary: Neonatal and infant pain management research faces two major challenges: lack of clear biomarkers and very heterogeneous pharmacokinetics and pharmacodynamics of analgesics. There is a clear call for integral research addressing the multimodality of pain in this population and further developing population pharmacokinetic models towards physiology-based models.
Collapse
Affiliation(s)
- Manuel A Baarslag
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Karel Allegaert
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,b Department of development and regeneration , KU Leuven , Leuven , Belgium
| | - John N Van Den Anker
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,c Division of Clinical Pharmacology , Children's National Health System , Washington , DC , USA.,d Division of Pediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - Catherijne A J Knibbe
- e Department of Clinical Pharmacy , St. Antonius Hospital , Nieuwegein , The Netherlands.,f Division of Pharmacology, Leiden Academic Center for Drug Research , Leiden University , Leiden , the Netherlands
| | - Monique Van Dijk
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,g Department of Pediatrics, division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Sinno H P Simons
- g Department of Pediatrics, division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Dick Tibboel
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| |
Collapse
|
37
|
Mahmood I, Ahmad T, Mansoor N, Sharib SM. Prediction of Clearance in Neonates and Infants (≤ 3 Months of Age) for Drugs That Are Glucuronidated: A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 2016; 57:476-483. [DOI: 10.1002/jcph.837] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/01/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology, Clinical Review Branch; Office of Blood Review & Research (OBRR); Center for Biologic Evaluation and Research; Food & Drug Administration; Silver Spring MD USA
| | - Tasneem Ahmad
- Pharma Professional Services; A-93 Ettawah Society Gadap Town Karachi Pakistan
| | - Najia Mansoor
- Department of Pharmacology; Faculty of Pharmacy & Pharmaceutical Sciences; University of Karachi; Karachi Pakistan
| | - S. M. Sharib
- Pharma Professional Services; A-93 Ettawah Society Gadap Town Karachi Pakistan
| |
Collapse
|
38
|
Knøsgaard KR, Foster DJR, Kreilgaard M, Sverrisdóttir E, Upton RN, van den Anker JN. Pharmacokinetic models of morphine and its metabolites in neonates:: Systematic comparisons of models from the literature, and development of a new meta-model. Eur J Pharm Sci 2016; 92:117-30. [PMID: 27373670 DOI: 10.1016/j.ejps.2016.06.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 11/19/2022]
Abstract
Morphine is commonly used for pain management in preterm neonates. The aims of this study were to compare published models of neonatal pharmacokinetics of morphine and its metabolites with a new dataset, and to combine the characteristics of the best predictive models to design a meta-model for morphine and its metabolites in preterm neonates. Moreover, the concentration-analgesia relationship for morphine in this clinical setting was also investigated. A population of 30 preterm neonates (gestational age: 23-32weeks) received a loading dose of morphine (50-100μg/kg), followed by a continuous infusion (5-10μg/kg/h) until analgesia was no longer required. Pain was assessed using the Premature Infant Pain Profile. Five published population models were compared using numerical and graphical tests of goodness-of-fit and predictive performance. Population modelling was conducted using NONMEM® and the $PRIOR subroutine to describe the time-course of plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide, and the concentration-analgesia relationship for morphine. No published model adequately described morphine concentrations in this new dataset. Previously published population pharmacokinetic models of morphine, morphine-3-glucuronide, and morphine-6-glucuronide were combined into a meta-model. The meta-model provided an adequate description of the time-course of morphine and the concentrations of its metabolites in preterm neonates. Allometric weight scaling was applied to all clearance and volume terms. Maturation of morphine clearance was described as a function of postmenstrual age, while maturation of metabolite elimination was described as a function of postnatal age. A clear relationship between morphine concentrations and pain score was not established.
Collapse
Affiliation(s)
- Katrine Rørbæk Knøsgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - David John Richard Foster
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia
| | - Mads Kreilgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Eva Sverrisdóttir
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Richard Neil Upton
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia.
| | - Johannes N van den Anker
- Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA; Division of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Switzerland; Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Mooij MG, van de Steeg E, van Rosmalen J, Windster JD, de Koning BAE, Vaes WHJ, van Groen BD, Tibboel D, Wortelboer HM, de Wildt SN. Proteomic Analysis of the Developmental Trajectory of Human Hepatic Membrane Transporter Proteins in the First Three Months of Life. Drug Metab Dispos 2016; 44:1005-13. [PMID: 27103634 DOI: 10.1124/dmd.115.068577] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/20/2016] [Indexed: 01/02/2023] Open
Abstract
Human hepatic membrane-embedded transporter proteins are involved in trafficking endogenous and exogenous substrates. Even though impact of transporters on pharmacokinetics is recognized, little is known on maturation of transporter protein expression levels, especially during early life. We aimed to study the protein expression of 10 transporters in liver tissue from fetuses, infants, and adults. Transporter protein expression levels [ATP-binding cassette transporter (ABC)B1, ABCG2, ABCC2, ABCC3, bile salt efflux pump, glucose transporter 1, monocarboxylate transporter 1, organic anion transporter polypeptide (OATP)1B1, OATP2B1, and organic cation/carnitine transporter 2) were quantified using ultraperformance liquid chromatography tandem mass spectrometry in snap-frozen postmortem fetal, infant, and adult liver samples. Protein expression was quantified in isolated crude membrane fractions. The possible association between postnatal and postmenstrual age versus protein expression was studied. We studied 25 liver samples, as follows: 10 fetal [median gestational age 23.2 wk (range 16.4-37.9)], 12 infantile [gestational age at birth 35.1 wk (27.1-41.0), postnatal age 1 wk (0-11.4)], and 3 adult. The relationship of protein expression with age was explored by comparing age groups. Correlating age within the fetal/infant age group suggested four specific protein expression patterns, as follows: stable, low to high, high to low, and low-high-low. The impact of growth and development on human membrane transporter protein expression is transporter-dependent. The suggested age-related differences in transporter protein expression may aid our understanding of normal growth and development, and also may impact the disposition of substrate drugs in neonates and young infants.
Collapse
Affiliation(s)
- Miriam G Mooij
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Evita van de Steeg
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Joost van Rosmalen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Jonathan D Windster
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Barbara A E de Koning
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Wouter H J Vaes
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Bianca D van Groen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Heleen M Wortelboer
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| | - Saskia N de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (M.G.M., J.D.W., B.D.G., D.T., S.N.W.); Department of Biostatistics, Erasmus MC, Rotterdam, The Netherlands (J.R.); Division of Pediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.A.E.K.); TNO, Zeist, The Netherlands (E.v.d.S., H.M.W., W.H.J.V.); and Department of Pharmacology and Toxicology, Radboud University, Nijmegen, The Netherlands (S.N.d.W.)
| |
Collapse
|
40
|
Mahmood I, Staschen CM. Prediction of Human Glomerular Filtration Rate from Preterm Neonates to Adults: Evaluation of Predictive Performance of Several Empirical Models. AAPS J 2016; 18:445-54. [PMID: 26801317 PMCID: PMC4779094 DOI: 10.1208/s12248-016-9868-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 01/05/2016] [Indexed: 12/16/2022] Open
Abstract
The objective of this study was to evaluate the predictive performance of several allometric empirical models (body weight dependent, age dependent, fixed exponent 0.75, a data-dependent single exponent, and maturation models) to predict glomerular filtration rate (GFR) in preterm and term neonates, infants, children, and adults without any renal disease. In this analysis, the models were developed from GFR data obtained from inulin clearance (preterm neonates to adults; n = 93) and the predictive performance of these models were evaluated in 335 subjects (preterm neonates to adults). The primary end point was the prediction of GFR from the empirical allometric models and the comparison of the predicted GFR with measured GFR. A prediction error within ±30% was considered acceptable. Overall, the predictive performance of the four models (BDE, ADE, and two maturation models) for the prediction of mean GFR was good across all age groups but the prediction of GFR in individual healthy subjects especially in neonates and infants was erratic and may be clinically unacceptable.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology Clinical Review Branch, Office of Blood Review & Research (OBRR), Center for Biologic Evaluation and Research, Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993-0002, USA.
| | - Carl-Michael Staschen
- Division of Hematology Clinical Review Branch, Office of Blood Review & Research (OBRR), Center for Biologic Evaluation and Research, Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993-0002, USA
| |
Collapse
|
41
|
Liu T, Lewis T, Gauda E, Gobburu J, Ivaturi V. Mechanistic Population Pharmacokinetics of Morphine in Neonates With Abstinence Syndrome After Oral Administration of Diluted Tincture of Opium. J Clin Pharmacol 2016; 56:1009-18. [PMID: 26712409 DOI: 10.1002/jcph.696] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/16/2015] [Indexed: 11/09/2022]
Abstract
Conducting and analyzing clinical trials in vulnerable neonates are extremely challenging. The aim of this analysis is to develop a morphine population pharmacokinetics (PK) model using data collected during a randomized control trial in neonates with abstinence syndrome (NAS). A 3-compartment morphine structural PK model after intravenous (IV) administration from previously published work was utilized as prior, whereas an allometric scaling method with physiological consideration was used to extrapolate a PK profile from adults to pediatrics. The absorption rate constant and bioavailability were estimated in NAS after oral administration of diluted tincture of opium (DTO). Goodness-of-fit plots along with normalized prediction distribution error and bootstrap method were performed for model evaluation. We successfully extrapolated the PK profile from adults to pediatrics after IV administration. The estimated first-order absorption rate constant and bioavailability were 0.751 hour(-1) and 48.5%, respectively. Model evaluations showed that the model can accurately and precisely describe the observed data. The population pharmacokinetic model we derived for morphine after oral administration of DTO is reasonable and acceptable; therefore, it can be used to describe the PK and guide future studies. The integration of the previous population PK knowledge as prior information successfully overcomes the logistic and practical issue in vulnerable neonate population.
Collapse
Affiliation(s)
- Tao Liu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Tamorah Lewis
- Department of Pediatrics, Division of Neonatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Estelle Gauda
- Department of Pediatrics, Division of Neonatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Jogarao Gobburu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Vijay Ivaturi
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
42
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
43
|
Mahmood I. Prediction of Drug Clearance in Premature and Mature Neonates, Infants, and Children ≤2 Years of Age: A Comparison of the Predictive Performance of 4 Allometric Models. J Clin Pharmacol 2015; 56:733-9. [DOI: 10.1002/jcph.652] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology Clinical Review, Office of Blood Review & Research (OBRR); Center for Biologic Evaluation and Research, US Food and Drug Administration; Silver Spring MD USA
| |
Collapse
|
44
|
Mahmood I. Mechanistic versus allometric models for the prediction of drug clearance in neonates (<3 months of age). J Clin Pharmacol 2015; 55:718-20. [PMID: 25943826 DOI: 10.1002/jcph.487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/19/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology Clinical Review, Office of Blood Review & Research (OBRR), Center for Biologic Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
45
|
Response to letter to the editor from Dr. Mahmood. J Clin Pharmacol 2015; 55:721. [DOI: 10.1002/jcph.490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 11/07/2022]
|
46
|
A Compartmental Analysis for Morphine and Its Metabolites in Young Children After a Single Oral Dose. Clin Pharmacokinet 2015; 54:1083-90. [DOI: 10.1007/s40262-015-0256-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
47
|
Mahmood I. Prediction of drug clearance in children: a review of different methodologies. Expert Opin Drug Metab Toxicol 2015; 11:573-87. [DOI: 10.1517/17425255.2015.1019463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Mahmood I, Staschen CM, Goteti K. Prediction of drug clearance in children: an evaluation of the predictive performance of several models. AAPS J 2014; 16:1334-43. [PMID: 25274608 PMCID: PMC4389735 DOI: 10.1208/s12248-014-9667-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/04/2014] [Indexed: 01/19/2023] Open
Abstract
The objective of this study is to evaluate the predictive performance of several models to predict drug clearance in children ≤5 years of age. Six models (allometric model (data-dependent exponent), fixed exponent of 0.75 model, maturation model, body weight-dependent model, segmented allometric model, and age-dependent exponent model) were evaluated in this study. From the literature, the clearance values for six drugs from neonates to adults were obtained. External data were used to evaluate the predictive performance of these models in children ≤5 years of age. With the exception of a fixed exponent of 0.75, the mean predicted clearance in most of the age groups was within ≤50% prediction error. Individual clearance prediction was erratic by all models and cannot be used reliably to predict individual clearance. Maturation, body weight-dependent, and segmented allometric models to predict clearances of drugs in children ≤5 years of age are of limited practical value during drug development due to the lack of availability of data. Age-dependent exponent model can be used for the selection of first-in-children dose during drug development.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Division of Hematology, Office of Blood Review & Research (OBRR), Center for Biologic Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993-0002, USA,
| | | | | |
Collapse
|
49
|
Staschen CM, Mahmood I. A population pharmacokinetic model of remifentanil in pediatric patients using body-weight-dependent allometric exponents. ACTA ACUST UNITED AC 2014; 28:231-7. [PMID: 24114900 DOI: 10.1515/dmdi-2013-0038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/09/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Allometric exponents in population pharmacokinetic analysis are regularly used but the issue of fixing or estimating an allometric exponent remains controversial. The objective of the current analysis is to evaluate the performance of a body-weight-dependent allometric exponent (BDE) model of remifentanil. METHODS The study was conducted in 34 patients (neonates to 17 years and 2.5 to 97 kg body weight) following a single intravenous (IV) infusion of remifentanil (5 μg/kg). A population pharmacokinetic approach was taken to describe drug clearance by the following BDE equation: CL=CLpop(BW/14.6 kg)L×BW(-M). Three allometric models were used to explore the impact of allometric exponents on the total clearance of remifentanil. RESULTS All model-fitted structural, covariate, and statistical parameters were estimated with good to excellent precision (%RSE). However, on the basis of calculated Akaike weights (0.000 for model 1, 0.004 for model 2, and 0.996 for model 3), model 3 is the most robust model to describe individual clearance estimates. CONCLUSIONS The BDE model performed best for the estimation of remifentanil clearance and is realistic and of practical value. Further investigation should be conducted for such models.
Collapse
|
50
|
Prediction of glucuronidated drug clearance in pediatrics (≤5 years): An allometric approach. Eur J Drug Metab Pharmacokinet 2014; 40:53-9. [DOI: 10.1007/s13318-014-0178-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
|