1
|
Goelen J, Farrell G, McGeehan J, Titman CM, J W Rattray N, Johnson TN, Horniblow RD, Batchelor HK. Quantification of drug metabolising enzymes and transporter proteins in the paediatric duodenum via LC-MS/MS proteomics using a QconCAT technique. Eur J Pharm Biopharm 2023; 191:68-77. [PMID: 37625656 DOI: 10.1016/j.ejpb.2023.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Characterising the small intestine absorptive membrane is essential to enable prediction of the systemic exposure of oral formulations. In particular, the ontogeny of key intestinal Drug Metabolising Enzymes and Transporter (DMET) proteins involved in drug disposition needs to be elucidated to allow for accurate prediction of the PK profile of drugs in the paediatric cohort. Using pinch biopsies from the paediatric duodenum (n = 36; aged 11 months to 15 years), the abundance of 21 DMET proteins and two enterocyte markers were quantified via LC-MS/MS. An established LCMS nanoflow method was translated to enable analysis on a microflow LC system, and a new stable-isotope-labelled QconCAT standard developed to enable quantification of these proteins. Villin-1 was used to standardise abundancy values. The observed abundancies and ontogeny profiles, agreed with adult LC-MS/MS-based data, and historic paediatric data obtained via western blotting. A linear trend with age was observed for duodenal CYP3A4 and CES2 only. As this work quantified peptides on a pinch biopsy coupled with a microflow method, future studies using a wider population range are very feasible. Furthermore, this DMET ontogeny data can be used to inform paediatric PBPK modelling and to enhance the understanding of oral drug absorption and gut bioavailability in paediatric populations.
Collapse
Affiliation(s)
- Jan Goelen
- School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Gillian Farrell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | | | | | - Nicholas J W Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | | | - Richard D Horniblow
- School of Biomedical Science, University of Birmingham, Birmingham B15 2TT, UK
| | - Hannah K Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
2
|
Ridley A, Vial-Cholley E, Robert G, Jounis-Jahan F, Lervat C, Betremieux P, Viallard ML, Frache S, Cojean N. Nationwide Study of Continuous Deep Sedation Practices Among Pediatric Palliative Care Teams. J Pain Symptom Manage 2023; 65:308-317. [PMID: 36528187 DOI: 10.1016/j.jpainsymman.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/16/2022]
Abstract
CONTEXT Palliative sedation practices evolved in France when the Claeys-Leonetti law passed in 2016 authorized patient-requested continuous deep sedation (CDS) until death. Its implementation in the pediatric setting is less frequently encountered and can pose several clinical and ethical challenges for health care teams and families. OBJECTIVES Our study aimed to describe CDS requests and practices of patients receiving specialized pediatric palliative care in France since its legalization in 2016. METHODS We conducted a nationwide multicentric, descriptive, retrospective study using a self-report questionnaire completed by all Pediatric Palliative Care (PPC) Teams that were involved in a CDS case between January 2017 and December 2019. RESULTS Six PPC teams had cared for six patients that had requested CDS, predominantly male adolescents/young adults diagnosed with a solid tumour. The refractory symptoms were diverse (pain, bleeding, and sensory loss) and always coupled with psycho-existential suffering. Each request was analyzed in multidisciplinary collegial meetings. Parental consent was always obtained regardless of age. Sedation typically required the use of multiple drugs including Midazolam (n = 5 cases), Chlorpromazine (n = 3), Ketamine (n = 2), and Propofol (n = 2). Despite close monitoring, achieving a satisfactory level of deep sedation was challenging and most patients unexpectedly awoke during CDS. Death occurred between 27 and 96 hours after induction. CONCLUSION Managing patient-requested CDS in pediatrics is challenging due to its rarity, multi-factorial refractory symptoms and drug tolerance despite polytherapy. Few recommendations exist to guide CDS practice for pediatricians. Further studies investigating pediatric CDS practices across various cultural and legal settings, refractory symptom management and specific pharmacology are warranted.
Collapse
Affiliation(s)
- Ashley Ridley
- Necker Enfants Malades Children's Hospital, Pediatric Palliative Care Team (A.R., M.L.V.), Paris, France.
| | | | - Guillaume Robert
- Pediatric Palliative Care Team Bretagne (G.R.), Rennes, France; University of Rennes 1 (G.R.), Rennes, France
| | | | - Cyril Lervat
- Pediatric Palliative Care Team Hauts de France (C.L.), Lille, France; Pediatric, Adolescent and Young Adult Oncology Unit (C.L.), Oscar Lambret Centre, Lille, France
| | - Pierre Betremieux
- French Society of Pediatric Palliative Care, Société Française de Soins Palliatifs Pédiatriques (P.B., S.F., N.C.)
| | - Marcel-Louis Viallard
- Necker Enfants Malades Children's Hospital, Pediatric Palliative Care Team (A.R., M.L.V.), Paris, France
| | - Sandra Frache
- Pediatric Palliative Care Team Franche-Comté (S.F.), Besançon, France; French Society of Pediatric Palliative Care, Société Française de Soins Palliatifs Pédiatriques (P.B., S.F., N.C.)
| | - Nadine Cojean
- Pediatric Palliative Care Team Alsace (N.C.), Strasbourg, France; French Society of Pediatric Palliative Care, Société Française de Soins Palliatifs Pédiatriques (P.B., S.F., N.C.)
| |
Collapse
|
3
|
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
|
4
|
Jovanović M, Vučićević K. Pediatric pharmacokinetic considerations and implications for drug dosing. ARHIV ZA FARMACIJU 2022; 72:340-352. [DOI: 10.5937/arhfarm72-37605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Optimizing the dosing of medicines for pediatric patients in routine clinical practice and determining the dose for clinical trials is still a challenging task. Children differ from adults in their response to drugs due to inherent differences in pharmacokinetics and/or pharmacodynamics, and responses may also vary among pediatric patients of different ages. However, the greatest disparities compared to adult pharmacokinetic profiles are observed in children below 2 years of age. The maturation of the liver and the kidneys, as well as the variation in body composition, are considered to be the main sources of pharmacokinetic variability. Hence, besides specific pharmacodynamic features, understanding age-related changes in drug absorption, distribution, and elimination is fundamental for optimizing drug efficacy and avoiding toxicity. This paper summarizes the pharmacokinetic changes throughout the childhood, along with the effect of developmental changes on drug dosage calculation. In clinical practice, age and body weight-based dosing regimens are usually used. In spite of dosing recommendations based on age and/or body weight, variabilities in pharmacokinetics and pharmacodynamic response remain, implying a need to monitor patients and optimize the dosing regimen according to physiological characteristics, disease characteristics and therapy.
Collapse
|
5
|
Preterm Physiologically Based Pharmacokinetic Model. Part II: Applications of the Model to Predict Drug Pharmacokinetics in the Preterm Population. Clin Pharmacokinet 2021; 59:501-518. [PMID: 31587145 DOI: 10.1007/s40262-019-00827-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Preterm neonates are usually not part of a traditional drug development programme, however they are frequently administered medicines. Developing modelling and simulation tools, such as physiologically based pharmacokinetic (PBPK) models that incorporate developmental physiology and maturation of drug metabolism, can be used to predict drug exposure in this group of patients, and may help to optimize drug dose adjustment. OBJECTIVE The aim of this study was to assess and verify the predictability of a preterm PBPK model using compounds that undergo diverse renal and/or hepatic clearance based on the knowledge of their disposition in adults. METHODS A PBPK model was developed in the Simcyp Simulator V17 to predict the pharmacokinetics (PK) of drugs in preterm neonates. Drug parameters for alfentanil, midazolam, caffeine, ibuprofen, gentamicin and vancomycin were collated from the literature. Predicted PK parameters and profiles were compared against the observed data. RESULTS The preterm PBPK model predicted the PK changes of the six compounds using ontogeny functions for cytochrome P450 (CYP) 1A2, CYP2C9 and CYP3A4 after oral and intravenous administrations. For gentamicin and vancomycin, the maturation of renal function was able to predict the exposure of these two compounds after intravenous administration. All PK parameter predictions were within a twofold error criteria. CONCLUSION While the developed preterm model for the prediction of PK behaviour in preterm patients is not intended to replace clinical studies, it can potentially help with deciding on first-time dosing in this population and study design in the absence of clinical data.
Collapse
|
6
|
Zimmerman KO, Spears TG, Cobbaert M, Boakye-Agyeman F, Wu H, Cohen-Wolkowiez M, Watt KM, Benjamin DK, Becker ML, Traube C, Smith PB. Use of Electronic Health Records to Identify Exposure-Response Relationships in Critically Ill Children: An Example of Midazolam and Delirium. J Pediatr Intensive Care 2021; 11:300-307. [DOI: 10.1055/s-0041-1725148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/21/2021] [Indexed: 10/21/2022] Open
Abstract
AbstractAdverse drug events are common in critically ill children and often result from systemic or target organ drug exposure. Methods of drug dosing and titration that consider pharmacokinetic alterations may improve our ability to optimally dose critically ill patients and reduce the risk for drug-related adverse events. To demonstrate this possibility, we explored the exposure-response relationship between midazolam and delirium in critically ill children. We retrospectively examined electronic health records (EHRs) of critically ill children <18 years of age hospitalized in the pediatric intensive care unit at Duke University; these children were administered midazolam during mechanical ventilation and had ≥1 Cornell Assessment of Pediatric Delirium (CAPD) score. We used individual-level data extracted from the EHR and a previously published population pharmacokinetic (PK) model developed in critically ill children to simulate plasma concentrations at the time of CAPD scores in 1,000 representative datasets. We used multilevel repeated measures models, with clustering at patient and simulation levels, to evaluate the associations between measures of drug exposure (e.g., concentration and area under concentration time curve) and delirium scores. We included 61 children, median age 1.5 years (range = 0.1–16.3), with 181 CAPD assessments. We identified similarities between simulated Empirical Bayesian parameter estimates from the EHR cohort and those from the PK model population. We identified a stronger association between drug concentration at the time of score and CAPD scores (coefficient 1.78; 95% confidence interval: 1.66–1.90) compared with cumulative dose per kilogram and CAPD scores (coefficient −0.01; 95% confidence interval: −0.01 to −0.01). EHR and PK models can be leveraged to investigate exposure-response relationships in critically ill children.
Collapse
Affiliation(s)
- Kanecia O. Zimmerman
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| | - Tracy G. Spears
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Marjan Cobbaert
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Felix Boakye-Agyeman
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Huali Wu
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Michael Cohen-Wolkowiez
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| | - Kevin M. Watt
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| | - Daniel K. Benjamin
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| | - Mara L. Becker
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Chani Traube
- Division of Critical Care Medicine, Weill Cornell Medical College, New York City, New York, United States
| | - P. Brian Smith
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
7
|
Siivola E, Weber S, Vehtari A. Qualifying drug dosing regimens in pediatrics using Gaussian processes. Stat Med 2021; 40:2355-2372. [PMID: 33586203 DOI: 10.1002/sim.8907] [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: 12/12/2019] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 11/10/2022]
Abstract
Drug development commonly studies an adult population first and then the pediatric population. The knowledge from the adult population is taken advantage of for the design of the pediatric trials. Adjusted drug doses for these are often derived from adult pharmacokinetic (PK) models which are extrapolated to patients with smaller body size. This extrapolation is based on scaling physiologic model parameters with a body size measure accounting for organ size differences. The inherent assumption is that children are merely small adults. However, children can be subject to additional effects such as organ maturation. These effects are not present in the adult population and are possibly overlooked at the design stage of the pediatric trials. It is thus crucial to qualify the extrapolation assumptions once the pediatric trial data are available. In this work, we propose a model based on a non-parametric regression method called Gaussian process (GP) to detect deviations from the made extrapolation assumptions. We introduce the theoretical background of this model and compare its performance to a parametric expansion of the adult model. The comparison includes simulations and a clinical study data example. The results show that the GP approach can reliably detect maturation trends from sparse pediatric data.
Collapse
Affiliation(s)
- Eero Siivola
- Advanced Exploratory Analytics, Novartis Pharma AG, Basel, Switzerland.,Department of Computer Science, Aalto University, Espoo, Finland
| | - Sebastian Weber
- Advanced Exploratory Analytics, Novartis Pharma AG, Basel, Switzerland
| | - Aki Vehtari
- Department of Computer Science, Aalto University, Espoo, Finland
| |
Collapse
|
8
|
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
|
9
|
Admiraal R, Jol-van der Zijde CM, Furtado Silva JM, Knibbe CAJ, Lankester AC, Boelens JJ, Hale G, Etuk A, Wilson M, Adams S, Veys P, van Kesteren C, Bredius RGM. Population Pharmacokinetics of Alemtuzumab (Campath) in Pediatric Hematopoietic Cell Transplantation: Towards Individualized Dosing to Improve Outcome. Clin Pharmacokinet 2019; 58:1609-1620. [PMID: 31131436 PMCID: PMC6885503 DOI: 10.1007/s40262-019-00782-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Alemtuzumab (Campath®) is used to prevent graft-versus-host disease and graft failure following pediatric allogeneic hematopoietic cell transplantation. The main toxicity includes delayed immune reconstitution, subsequent viral reactivations, and leukemia relapse. Exposure to alemtuzumab is highly variable upon empirical milligram/kilogram dosing. METHODS A population pharmacokinetic (PK) model for alemtuzumab was developed based on a total of 1146 concentration samples from 206 patients, aged 0.2-19 years, receiving a cumulative intravenous dose of 0.2-1.5 mg/kg, and treated between 2003 and 2015 in two centers. RESULTS Alemtuzumab PK were best described using a two-compartment model with a parallel saturable and linear elimination pathway. The linear clearance pathway, central volume of distribution, and intercompartmental distribution increased with body weight. Blood lymphocyte counts, a potential substrate for alemtuzumab, did not impact clearance. CONCLUSION The current practice with uniform milligram/kilogram doses leads to highly variable exposures in children due to the non-linear relationship between body weight and alemtuzumab PK. This model may be used for individualized dosing of alemtuzumab.
Collapse
Affiliation(s)
- Rick Admiraal
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Cornelia M Jol-van der Zijde
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Arjan C Lankester
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap Jan Boelens
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
- Stem Cell Transplant and Cellular Therapies, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | | | - Aniekan Etuk
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Melanie Wilson
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Stuart Adams
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Paul Veys
- Bone Marrow Transplantation Department, Great Ormond Street Hospital, London, UK
| | - Charlotte van Kesteren
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Robbert G M Bredius
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
10
|
Gonzalez D, Laughon MM, Smith PB, Ge S, Ambalavanan N, Atz A, Sokol GM, Hornik CD, Stewart D, Mundakel G, Poindexter BB, Gaedigk R, Mills M, Cohen‐Wolkowiez M, Martz K, Hornik CP, on behalf of the Best Pharmaceuticals for Children Act – Pediatric Trials Network Steering Committee. Population pharmacokinetics of sildenafil in extremely premature infants. Br J Clin Pharmacol 2019; 85:2824-2837. [PMID: 31475367 PMCID: PMC6955411 DOI: 10.1111/bcp.14111] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 11/30/2022] Open
Abstract
AIMS To characterize the population pharmacokinetics (PK) of sildenafil and its active metabolite, N-desmethyl sildenafil (DMS), in premature infants. METHODS We performed a multicentre, open-label trial to characterize the PK of sildenafil in infants ≤28 weeks gestation and < 365 postnatal days (cohort 1) or < 32 weeks gestation and 3-42 postnatal days (cohort 2). In cohort 1, we obtained PK samples from infants receiving sildenafil as ordered per the local standard of care (intravenous [IV] or enteral). In cohort 2, we administered a single IV dose of sildenafil and performed PK sampling. We performed a population PK analysis and dose-exposure simulations using the software NONMEM®. RESULTS We enrolled 34 infants (cohort 1 n = 25; cohort 2 n = 9) and collected 109 plasma PK samples. Sildenafil was given enterally (0.42-2.09 mg/kg) in 24 infants in cohort 1 and via IV (0.125 or 0.25 mg/kg) in all infants in cohort 2. A 2-compartment PK model for sildenafil and 1-compartment model for DMS, with presystemic conversion of sildenafil to DMS, characterized the data well. Coadministration of fluconazole (n = 4), a CYP3A inhibitor, resulted in an estimated 59% decrease in sildenafil clearance. IV doses of 0.125, 0.5 and 1 mg/kg every 8 hours (in the absence of fluconazole) resulted in steady-state maximum sildenafil concentrations that were generally within the range of those reported to inhibit phosphodiesterase type 5 activity in vitro. CONCLUSIONS We successfully characterized the PK of sildenafil and DMS in premature infants and applied the model to inform dosing for a follow-up, phase II study.
Collapse
MESH Headings
- Administration, Oral
- Cohort Studies
- Cytochrome P-450 CYP3A/blood
- Cytochrome P-450 CYP3A/genetics
- Fluconazole/administration & dosage
- Fluconazole/pharmacokinetics
- Gestational Age
- Humans
- Hypertension, Pulmonary/blood
- Hypertension, Pulmonary/drug therapy
- Infant
- Infant, Newborn
- Infant, Premature/blood
- Infant, Premature, Diseases/blood
- Infant, Premature, Diseases/drug therapy
- Injections, Intravenous
- Models, Biological
- Phosphodiesterase 5 Inhibitors/administration & dosage
- Phosphodiesterase 5 Inhibitors/blood
- Phosphodiesterase 5 Inhibitors/pharmacokinetics
- Phosphodiesterase 5 Inhibitors/therapeutic use
- Sildenafil Citrate/administration & dosage
- Sildenafil Citrate/blood
- Sildenafil Citrate/pharmacokinetics
- Sildenafil Citrate/therapeutic use
Collapse
Affiliation(s)
- Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - Matthew M. Laughon
- Department of Pediatrics, School of MedicineThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - P. Brian Smith
- Department of PediatricsDuke University School of MedicineDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | - Shufan Ge
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNCUSA
| | - Namasivayam Ambalavanan
- Division of Neonatology, School of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Andrew Atz
- Department of PediatricsMedical University of South Carolina Children's HospitalCharlestonSCUSA
| | - Gregory M. Sokol
- Section of Neonatal‐Perinatal MedicineIndiana University School of MedicineIndianapolisINUSA
| | - Chi D. Hornik
- Department of PediatricsDuke University School of MedicineDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
- Department of PharmacyDuke University Medical CenterDurhamNCUSA
| | - Dan Stewart
- University of Louisville Norton Children's HospitalLouisvilleKYUSA
| | - Gratias Mundakel
- Kings County Hospital Center/SUNY Downstate Medical CenterBrooklynNYUSA
| | | | - Roger Gaedigk
- Department of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy‐Kansas CityUniversity of Missouri‐Kansas City School of MedicineKansas CityMOUSA
| | - Mary Mills
- Duke Clinical Research InstituteDurhamNCUSA
| | - Michael Cohen‐Wolkowiez
- Department of PediatricsDuke University School of MedicineDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | | | - Christoph P. Hornik
- Department of PediatricsDuke University School of MedicineDurhamNCUSA
- Duke Clinical Research InstituteDurhamNCUSA
| | | |
Collapse
|
11
|
Pharmacokinetic modeling of intravenous sildenafil in newborns with congenital diaphragmatic hernia. Eur J Clin Pharmacol 2019; 76:219-227. [PMID: 31740991 DOI: 10.1007/s00228-019-02767-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/15/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE We developed a pharmacokinetic model of intravenous sildenafil in newborns with congenital diaphragmatic hernia (CDH) to achieve a target plasma concentration of over 50 μg/l. METHODS Twenty-three CDH newborns with pulmonary hypertension (64 blood samples) received intravenous sildenafil. Patients received a loading dose of 0.35 mg/kg (IQR 0.16 mg/kg) for 3 h, followed by a continuous infusion of 1.5 mg/kg/day (IQR 0.1 mg/kg/day). For model development, non-linear mixed modeling was used. Inter-individual variability (IIV) and inter-occasion variability were tested. Demographic and laboratory parameters were evaluated as covariates. Normalized prediction distribution errors (NPDE) and visual predictive check (VPC) were used for model validation. RESULTS A two-compartment disposition model of sildenafil and a one-compartment disposition model of desmethyl sildenafil (DMS) was observed with IIV in sildenafil and DMS clearance and volume of distribution of sildenafil. NPDE and VPC revealed adequate predictability. Only postnatal age increased sildenafil clearance. This was partly compensated by a higher DMS concentration, which also has a therapeutic effect. In this small group of patients, sildenafil was tolerated well. CONCLUSIONS This model for sildenafil in CDH patients shows that concentration-targeted sildenafil dosing of 0.4 mg/kg in 3 h, followed by 1.6 mg/kg/day continuous infusion achieves appropriate sildenafil plasma levels.
Collapse
|
12
|
Kos MK, Miksić M, Jovanović M, Roškar R, Grosek Š, Grabnar I. Maturation of midazolam clearance in critically ill children with severe bronchiolitis: A population pharmacokinetic analysis. Eur J Pharm Sci 2019; 141:105095. [PMID: 31626965 DOI: 10.1016/j.ejps.2019.105095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/24/2019] [Accepted: 09/29/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of the present study was to develop a population pharmacokinetic model of midazolam, and to evaluate the influence of maturation process and other variability factors in critically ill children with severe acute bronchiolitis, who received a long-term intravenous infusion of midazolam. METHODS In the study were included 49 critically ill children of both genders (from 0 to 130 weeks of age) with severe acute bronchiolitis hospitalised in intensive care units. Nonlinear mixed effects modelling approach was applied for data analyses and simulations. RESULTS The final model is a two-compartment model that includes the effects of body weight using allometric scaling with fixed exponents and maturation of clearance. For a typical subject, scaled to the adult body weight of 70 kg, population pharmacokinetic values were estimated at 8.52 L/h for clearance (when maturation function was 1), 25.5 L/h for intercompartmental clearance, and 5.71 L and 39.8 L for the volume of the central and peripheral compartment, respectively. Based on the final model, maturation reaches 50% of the adult clearance in 45.9 weeks of postmenstrual age. The influence of gender, ABCB1 genotype and biochemical parameters on midazolam clearance was not detected. Results of simulations indicate the need for reduced dosing in certain groups of patients in order to maintain plasma concentrations of midazolam within recommended values. CONCLUSIONS The developed population pharmacokinetic model can contribute to the dosing optimisation of midazolam, especially in critically ill children as it includes the influence of size and maturation of clearance, which are important parameters for achieving the desired plasma concentrations of midazolam.
Collapse
Affiliation(s)
- Mojca Kerec Kos
- Faculty of Pharmacy, University of Ljubljana, Askerceva cesta 7, Ljubljana 1000, Slovenia.
| | - Mirjana Miksić
- Division of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, Maribor 2000, Slovenia; Division of Gynaecology and Perinatology, Department of Perinatology, University Medical Centre Maribor, Ljubljanska ulica 5, Maribor 2000, Slovenia
| | - Marija Jovanović
- Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade 11221, Serbia
| | - Robert Roškar
- Faculty of Pharmacy, University of Ljubljana, Askerceva cesta 7, Ljubljana 1000, Slovenia
| | - Štefan Grosek
- Division of Surgery, Department of Paediatric Surgery and Intensive Therapy, University Medical Centre Ljubljana, Bohoriceva ulica 20, Ljubljana 1525, Slovenia; Department of Perinatology, Divison of Gynecology and Obstetrics, University Medical Centre Ljubljana, Šlajmerjeva 3, Ljubljana 1000, Slovenia; Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana 1000, Slovenia
| | - Iztok Grabnar
- Faculty of Pharmacy, University of Ljubljana, Askerceva cesta 7, Ljubljana 1000, Slovenia
| |
Collapse
|
13
|
Whalen ME, Kajubi R, Chamankhah N, Huang L, Orukan F, Wallender E, Kamya MR, Dorsey G, Jagannathan P, Rosenthal PJ, Mwebaza N, Aweeka FT. Reduced Exposure to Piperaquine, Compared to Adults, in Young Children Receiving Dihydroartemisinin-Piperaquine as Malaria Chemoprevention. Clin Pharmacol Ther 2019; 106:1310-1318. [PMID: 31173649 DOI: 10.1002/cpt.1534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 11/10/2022]
Abstract
Dihydroartemisinin (DHA)-piperaquine is being evaluated as intermittent preventive therapy for malaria, but dosing has not been optimized for children. We assessed exposure to DHA and piperaquine in Ugandan children at two ages during infancy. Intensive sampling was performed in 32 children at 32 weeks of age, 31 children at 104 weeks, and 30 female adult controls. Compared with adults, DHA area under the concentration-time curve (AUC0-8 hr ) was 52% higher at 32 weeks and comparable at 104 weeks. Compared with adults, piperaquine AUC0-21 d was 35% lower at 32 weeks and 53% lower at 104 weeks. Terminal piperaquine concentrations on days 7, 14, and 21 were lower in children compared with adults and lower at 104 compared with 32 weeks. Piperaquine exposure was lower in young children compared with adults, and lower at 104 compared with 32 weeks of age, suggesting a need for age-based DHA-piperaquine dose optimization for chemoprevention.
Collapse
Affiliation(s)
- Meghan E Whalen
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Richard Kajubi
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Nona Chamankhah
- Department of Pharmacy, Rady Children's Hospital, San Diego, California, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Francis Orukan
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Erika Wallender
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Moses R Kamya
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | | | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Francesca T Aweeka
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
14
|
Prediction of Clearance and Dose of Midazolam in Preterm and Term Neonates: A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling. Am J Ther 2019; 26:e32-e37. [DOI: 10.1097/mjt.0000000000000506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
van den Anker J, Reed MD, Allegaert K, Kearns GL. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J Clin Pharmacol 2018; 58 Suppl 10:S10-S25. [DOI: 10.1002/jcph.1284] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022]
Affiliation(s)
- John van den Anker
- Division of Clinical Pharmacology; Children's National Health System; Washington DC USA
- Division of Paediatric Pharmacology and Pharmacometrics; University of Basel Children's Hospital; Basel Switzerland
- Intensive Care and Department of Pediatric Surgery; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
| | - Michael D. Reed
- Emeritus Professor of Pediatrics; School of Medicine; Case Western Reserve University; Cleveland OH USA
| | - Karel Allegaert
- Intensive Care and Department of Pediatric Surgery; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
- Department of Pediatrics; Division of Neonatology; Erasmus Medical Center-Sophia Children's Hospital; Rotterdam the Netherlands
- Department of Development and Regeneration; KU Leuven; Leuven Belgium
| | | |
Collapse
|
16
|
Leroux S, Elie V, Zhao W, Magreault S, Jacqz-Aigrain E. Principles and applications of pharmacometrics in drug evaluation in children. Therapie 2018; 73:165-170. [DOI: 10.1016/j.therap.2017.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
|
17
|
Neyro V, Elie V, Médard Y, Jacqz-Aigrain E. mRNA expression of drug metabolism enzymes and transporter genes at birth using human umbilical cord blood. Fundam Clin Pharmacol 2018; 32:422-435. [DOI: 10.1111/fcp.12357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/18/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Virginia Neyro
- Department of Pediatric Clinical Pharmacology and Pharmacogenetics; Assistance Publique - Hôpitaux de Paris; Hôpital Robert Debré; Paris France
- Ecole Doctorale MTCI - Paris Descartes University; Paris France
| | - Valéry Elie
- Department of Pediatric Clinical Pharmacology and Pharmacogenetics; Assistance Publique - Hôpitaux de Paris; Hôpital Robert Debré; Paris France
| | - Yves Médard
- Department of Pediatric Clinical Pharmacology and Pharmacogenetics; Assistance Publique - Hôpitaux de Paris; Hôpital Robert Debré; Paris France
| | - Evelyne Jacqz-Aigrain
- Department of Pediatric Clinical Pharmacology and Pharmacogenetics; Assistance Publique - Hôpitaux de Paris; Hôpital Robert Debré; Paris France
- APHP INSERM Clinical Investigation Center CIC1426; Hôpital Robert Debré; Paris France
- Paris Diderot University; Sorbonne Paris-Cité; Paris France
| |
Collapse
|
18
|
Population pharmacokinetic evaluation of ADV6209, an innovative oral solution of midazolam containing cyclodextrin. Eur J Pharm Sci 2018; 114:46-54. [DOI: 10.1016/j.ejps.2017.11.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/01/2017] [Accepted: 11/30/2017] [Indexed: 11/23/2022]
|
19
|
Moorthy GS, Jogiraju H, Vedar C, Zuppa AF. Development and validation of a sensitive assay for analysis of midazolam, free and conjugated 1-hydroxymidazolam and 4-hydroxymidazolam in pediatric plasma: Application to Pediatric Pharmacokinetic Study. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1067:1-9. [PMID: 28978489 DOI: 10.1016/j.jchromb.2017.09.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/07/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022]
Abstract
Pharmacokinetic, pharmacodynamic and pharmacogenomic studies of midazolam are currently being performed in critically ill children to find suitable dose regimens. Sensitive assays using small volumes of plasma are necessary to determine the concentrations of midazolam and its respective metabolites in pediatric studies. Midazolam is metabolized to hydroxylated midazolam isomers, which are present as free as well as the corresponding glucuronide conjugates. A high-performance liquid chromatographic method with tandem mass spectrometry has been developed and validated for the quantification of midazolam, and free and total 1-hydroxymidazolam and 4-hydroxymidazolam metabolites in small volumes of plasma. Cleanup consisted of 96-well μ-elution solid phase extraction (SPE). The analytes were separated by gradient elution using a C18 analytical column with a total run time of 5min. Multiple reaction monitoring was employed using precursor to product ion transitions of m/z 326.2→291.3 for midazolam, m/z 342.1→203.0 for 1-hydroxymidazolam, m/z 342.1→325.1 for 4-hydroxymidazolam and m/z 330.2→295.3 for 2H4-midazolam (internal standard). Since authentic hydroxymidazolamglucuronide standards are not available, samples were hydrolyzed with β-glucuronidase under optimized conditions. Assay conditions were modified and optimized to provide appropriate recovery and stability because 4-hydroxymidazolam was very acid sensitive. Standard curves were linear from 0.5 to 1000ng/mL for all three analytes. Intra- and inter day accuracy and precision for quality control samples (2, 20, 200 and 800ng/mL) were within 85-115% and 15% (coefficient of variation), respectively. Stability in plasma and extracts were sufficient under assay conditions. Plasma samples were processed and analyzed for midazolam, and free 1-hydroxymidazolam and 4-hydroxymidazolam metabolites. Plasma samples that were hydrolyzed with β-glucuronidase were processed and analyzed for midazolam, and total 1-hydroxymidazolam and 4-hydroxymidazolam metabolites under the same assay conditions. The difference in concentration between the total and free hydroxymidazolam metabolites provided an estimate of conjugated hydroxymidazolam metabolites. The combination of 96-well μ-elution SPE and LC-MS/MS allows reliable quantification of midazolam and its metabolites in small volumes of plasma for pediatric patients. This assay is currently being successfully utilized for analysis of samples from ongoing clinical trials.
Collapse
Affiliation(s)
- Ganesh S Moorthy
- Center for Clinical Pharmacology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Harini Jogiraju
- Center for Clinical Pharmacology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Christina Vedar
- Center for Clinical Pharmacology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Athena F Zuppa
- Center for Clinical Pharmacology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
20
|
Semi-Mechanistic Model for Predicting the Dosing Rate in Children and Neonates for Drugs Mainly Eliminated by Cytochrome Metabolism. Clin Pharmacokinet 2017; 57:831-841. [DOI: 10.1007/s40262-017-0596-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Borsuk A, Wołoszczuk-Gębicka B, Bartkowska-Śniatkowska A, Rosada-Kurasińska J, Bienert A, Wiczling P. Flip-Flop Phenomenon in Epidural Sufentanil Pharmacokinetics: A Population Study in Children and Infants. J Clin Pharmacol 2017; 57:1194-1206. [PMID: 28510304 DOI: 10.1002/jcph.912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/10/2017] [Indexed: 11/07/2022]
Abstract
The aims of this study were to develop a population pharmacokinetic model of sufentanil coadministered with 0.2% ropivacaine as an epidural infusion in infants and describe the sufentanil absorption profile from epidural space. Data from 2 previously published studies were merged for analysis-20 infants aged 3-36 months receiving sufentanil as an epidural infusion and 41 children 0-17 years old receiving sufentanil as a long-term intravenous infusion. A population nonlinear mixed-effects model was built in NONMEM. Sufentanil pharmacokinetics were described by a 2-compartment model with first-order absorption. The effect of body size on all volume and clearance parameters was included in the model according to allometric scaling with theoretical exponents. The maturation process of metabolic clearance was described by the Hill model. During the model-building process the population was divided into 2 fractions with different typical values of metabolic clearance (CL1 and CL2). The typical values of systemic clearance scaled to a 70-kg patient for the 2 subpopulations were CL1 = 52.6 L/h and CL2 = 158 L/h. The parameters of the Hill function were 54.9 weeks for the postmenstrual age of 50% clearance maturation and 0.802 for the Hill coefficient. The typical values of distribution clearance and volumes of the central and peripheral compartments for a patient with a weight of 70 kg were Q = 40.5 L/h, VC = 7.63 L, and VT = 473 L, respectively. The value of the absorption rate constant from the epidural space was 0.0459/h, which suggests flip-flop pharmacokinetics of sufentanil after epidural administration.
Collapse
Affiliation(s)
- Agnieszka Borsuk
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Bogumiła Wołoszczuk-Gębicka
- Department of Intensive Therapy and Toxicology, Chair of Rescue Medicine, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | | | - Jowita Rosada-Kurasińska
- Department of Pediatric Anesthesiology and Intensive Therapy, Poznań University of Medical Sciences, Poznań, Poland
| | - Agnieszka Bienert
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland
| | - Paweł Wiczling
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
22
|
Knowing Risk Factors for Iatrogenic Withdrawal Syndrome in Children May Still Leave Us Empty-Handed*. Crit Care Med 2017; 45:141-142. [DOI: 10.1097/ccm.0000000000002160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
23
|
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
|
24
|
Standing JF. Understanding and applying pharmacometric modelling and simulation in clinical practice and research. Br J Clin Pharmacol 2016; 83:247-254. [PMID: 27567102 PMCID: PMC5237699 DOI: 10.1111/bcp.13119] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 12/13/2022] Open
Abstract
Understanding the dose–concentration–effect relationship is a fundamental component of clinical pharmacology. Interpreting data arising from observations of this relationship requires the use of mathematical models; i.e. pharmacokinetic (PK) models to describe the relationship between dose and concentration and pharmacodynamic (PD) models describing the relationship between concentration and effect. Drug development requires several iterations of pharmacometric model‐informed learning and confirming. This includes modelling to understand the dose–response in preclinical studies, deriving a safe dose for first‐in‐man, and the overall analysis of Phase I/II data to optimise the dose for safety and efficacy in Phase III pivotal trials. However, drug development is not the boundary at which PKPD understanding and application stops. PKPD concepts will be useful to anyone involved in the prescribing and administration of medicines for purposes such as determining off‐label dosing in special populations, individualising dosing based on a measured biomarker (personalised medicine) and in determining whether lack of efficacy or unexpected toxicity maybe solved by adjusting the dose rather than the drug. In clinical investigator‐led study design, PKPD can be used to ensure the optimal dose is used, and crucially to define the expected effect size, thereby ensuring power calculations are based on sound prior information. In the clinical setting the most likely people to hold sufficient expertise to advise on PKPD matters will be the pharmacists and clinical pharmacologists. This paper reviews fundamental PKPD principles and provides some real‐world examples of PKPD use in clinical practice and applied clinical research.
Collapse
Affiliation(s)
- Joseph F Standing
- Infection, Immunity, Inflammation Section, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH.,Department of Pharmacy, Great Ormond Street Hospital for Children, London, WC1N 3JH.,Paediatric Infectious Diseases Research Group, St George's, University of London, Cranmer Terrace, London, SW17 0RE
| |
Collapse
|
25
|
Foissac F, Bouazza N, Valade E, De Sousa Mendes M, Fauchet F, Benaboud S, Hirt D, Tréluyer JM, Urien S. Prediction of drug clearance in children. J Clin Pharmacol 2015; 55:739-47. [DOI: 10.1002/jcph.488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/24/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Frantz Foissac
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Naïm Bouazza
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Elodie Valade
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Mailys De Sousa Mendes
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Floris Fauchet
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| | - Sihem Benaboud
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Déborah Hirt
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Jean-Marc Tréluyer
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
- Laboratoire de Pharmacologie; Hôpital Cochin; APHP; Paris France
| | - Saïk Urien
- EA 08; Université Paris Descartes; Sorbonne Paris Cité France
- Unité de Recherche Clinique; Assistance Publique Hôpitaux de Paris (APHP); Hôpital Tarnier; Paris France
- CIC-1419 Inserm; Cochin-Necker; Paris France
| |
Collapse
|
26
|
Abstract
Indinavir is a protease inhibitor antiretroviral (ARV) drug, which forms part of the highly active antiretroviral therapy during the treatment of HIV/AIDS. Indinavir undergoes first-pass metabolism through the cytochrome P450 (CYP) enzymes in the human liver, of which CYP3A4 is the most influential isoenzyme. Multidrug combination therapy and, as such, therapeutic drug monitoring (TDM) during HIV/AIDS treatment are therefore critical, to prevent adverse interactions. The conventional sensitive and specific assays available for quantifying ARV drugs, however, suffer from distinct disadvantages. In this regard, biosensors can be used to provide real time information on the metabolic profile of the drug. In this study, a biosensor with cobalt(III) sepulchrate trichloride{CoSep3+}as diffusional mediator was constructed. The biosensor platform consisted of CYP3A4 immobilized onto a gold nanoparticle (GNP) overoxidized polypyrrole (OvOxPpy) carrier matrix. The biosensor exhibited reversible electrochemistry, with formal potential determined as −624 ± 5 mV, from voltammetric analysis, with overall electron transfer being diffusion controlled. The biosensor showed typical electrocatalytic response to dioxygen (O2), exemplified by the distinct increase in the cathodic peak current (Ip,c). A concentration-dependent increase inIp,cwas observed in response to consecutive additions of Indinavir.
Collapse
|
27
|
Abstract
OBJECTIVES To determine the extent of inter-individual variation in clearance of midazolam in children and establish which factors are responsible for this variation. METHODS A systematic literature review was performed to identify papers describing the clearance of midazolam in children. The following databases were searched: Medline, Embase, International Pharmaceutical Abstracts, CINAHL and Cochrane Library. From the papers, the range in plasma clearance and the coefficient of variation (CV) in plasma clearance were determined. RESULTS 25 articles were identified. Only 13 studies gave the full range of clearance values for individual patients. The CV was greater in critically ill patients (18%-170%) than non-critically ill patients (13%-54%). Inter-individual variation was a major problem in all age groups of critically ill patients. The CV was 72%-106% in preterm neonates, 18%-73% in term neonates, 31%-130% in infants, 21%-170% in children and 47%-150% in adolescents. The mean clearance was higher in children (1.1-16.7 mL/min/kg) than in neonates (0.78-2.5 mL/min/kg). CONCLUSIONS Large inter-individual variation was seen in midazolam clearance values in critically ill neonates, infants, children and adolescents.
Collapse
Affiliation(s)
- Mohammed I Altamimi
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, UK
| | - Helen Sammons
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, UK
| | - Imti Choonara
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, UK
| |
Collapse
|
28
|
Abstract
As a standard of care for preterm/term newborns effective pain management may improve their clinical and neurodevelopmental outcomes. Neonatal pain is assessed using context-specific, validated, and objective pain methods, despite the limitations of currently available tools. Therapeutic approaches reducing invasive procedures and using pharmacologic, behavioral, or environmental measures are used to manage neonatal pain. Nonpharmacologic approaches like kangaroo care, facilitated tucking, non-nutritive sucking, sucrose, and others can be used for procedural pain or adjunctive therapy. Local/topical anesthetics, opioids, NSAIDs/acetaminophen and other sedative/anesthetic agents can be incorporated into NICU protocols for managing moderate/severe pain or distress in all newborns.
Collapse
|
29
|
De Cock RFW, Allegaert K, Brussee JM, Sherwin CMT, Mulla H, de Hoog M, van den Anker JN, Danhof M, Knibbe CAJ. Simultaneous pharmacokinetic modeling of gentamicin, tobramycin and vancomycin clearance from neonates to adults: towards a semi-physiological function for maturation in glomerular filtration. Pharm Res 2014; 31:2643-54. [PMID: 24789450 DOI: 10.1007/s11095-014-1361-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 03/15/2014] [Indexed: 01/07/2023]
Abstract
PURPOSE Since glomerular filtration rate (GFR) is responsible for the elimination of a large number of water-soluble drugs, the aim of this study was to develop a semi-physiological function for GFR maturation from neonates to adults. METHODS In the pharmacokinetic analysis (NONMEM VI) based on data of gentamicin, tobramycin and vancomycin collected in 1,760 patients (age 1 day-18 years, bodyweight 415 g-85 kg), a distinction was made between drug-specific and system-specific information. Since the maturational model for clearance is considered to contain system-specific information on the developmental changes in GFR, one GFR maturational function was derived for all three drugs. RESULTS Simultaneous analysis of these three drugs showed that maturation of GFR mediated clearance from preterm neonates to adults was best described by a bodyweight-dependent exponent (BDE) function with an exponent varying from 1.4 in neonates to 1.0 in adults (ClGFR = Cldrug*(BW/4 kg)(BDE) with BDE = 2.23*BW(-0.065)). Population clearance values (Cldrug) for gentamicin, tobramycin and vancomycin were 0.21, 0.28 and 0.39 L/h for a full term neonate of 4 kg, respectively. DISCUSSION Based on an integrated analysis of gentamicin, tobramycin and vancomycin, a semi-physiological function for GFR mediated clearance was derived that can potentially be used to establish evidence based dosing regimens of renally excreted drugs in children.
Collapse
|
30
|
Salem F, Johnson TN, Abduljalil K, Tucker GT, Rostami-Hodjegan A. A Re-evaluation and Validation of Ontogeny Functions for Cytochrome P450 1A2 and 3A4 Based on In Vivo Data. Clin Pharmacokinet 2014; 53:625-36. [DOI: 10.1007/s40262-014-0140-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
31
|
Clinical pharmacology of midazolam in neonates and children: effect of disease-a review. Int J Pediatr 2014; 2014:309342. [PMID: 24696691 PMCID: PMC3948203 DOI: 10.1155/2014/309342] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/26/2013] [Indexed: 12/04/2022] Open
Abstract
Midazolam is a benzodiazepine with rapid onset of action and short duration of effect. In healthy neonates the half-life (t1/2) and the clearance (Cl) are 3.3-fold longer and 3.7-fold smaller, respectively, than in adults. The volume of distribution (Vd) is 1.1 L/kg both in neonates and adults. Midazolam is hydroxylated by CYP3A4 and CYP3A5; the activities of these enzymes surge in the liver in the first weeks of life and thus the metabolic rate of midazolam is lower in neonates than in adults. Midazolam acts as a sedative, as an antiepileptic, for those infants who are refractory to standard antiepileptic therapy, and as an anaesthetic. Information of midazolam as an anaesthetic in infants are very little. Midazolam is usually administered intravenously; when minimal sedation is required, intranasal administration of midazolam is employed. Disease affects the pharmacokinetics of midazolam in neonates; multiple organ failure reduces the Cl of midazolam and mechanical ventilation prolongs the t1/2 of this drug. ECMO therapy increases t1/2, Cl, and Vd of midazolam several times. The adverse effects of midazolam in neonates are scarce: pain, tenderness, and thrombophlebitis may occur. Respiratory depression and hypotension appear in a limited percentage of infants following intravenous infusion of midazolam. In conclusion, midazolam is a safe and effective drug which is employed as a sedative, as antiepileptic agent, for infants who are refractory to standard antiepileptic therapy, and as an anaesthetic.
Collapse
|
32
|
Wang C, Sadhavisvam S, Krekels EHJ, Dahan A, Tibboel D, Danhof M, Vinks AA, Knibbe CAJ. Developmental changes in morphine clearance across the entire paediatric age range are best described by a bodyweight-dependent exponent model. Clin Drug Investig 2014; 33:523-34. [PMID: 23754691 DOI: 10.1007/s40261-013-0097-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Morphine clearance has been successfully scaled from preterm neonates to 3-year-old children on the basis of a bodyweight-based exponential (BDE) function and age younger or older than 10 days. The aim of the current study was to characterize the developmental changes in morphine clearance across the entire paediatric age range. METHODS Morphine and morphine-3-glucuronide (M3G) concentration data from 358 (pre)term neonates, infants, children and adults, and morphine concentration data from 117 adolescents were analysed using NONMEM 7.2. Based on available data, two models were developed: I. using morphine data; II. using morphine and M3G data. RESULTS In model I, morphine clearance across the paediatric age range was very well described by a BDE function in which the allometric exponent decreased in a sigmoidal manner with bodyweight (BDE model) from 1.47 to 0.88, with half the decrease in exponent reached at 4.01 kg. In model II, the exponent for the formation and elimination clearance of M3G was found to decrease from 1.56 to 0.89 and from 1.06 to 0.61, with half the decrease reached at 3.89 and 4.87 kg, respectively. Using the BDE model, there was no need to use additional measures for size or age. CONCLUSION The BDE model was able to scale both total morphine clearance and glucuronidation clearance through the M3G pathway across all age ranges between (pre)term neonates and adults by allowing the allometric exponent to decrease across the paediatric age range from values higher than 1 for neonates to values lower than 1 for infants and children.
Collapse
Affiliation(s)
- Chenguang Wang
- LACDR, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Wang C, Allegaert K, Tibboel D, Danhof M, van der Marel CD, Mathot RAA, Knibbe CAJ. Population pharmacokinetics of paracetamol across the human age-range from (pre)term neonates, infants, children to adults. J Clin Pharmacol 2014; 54:619-29. [PMID: 24375166 DOI: 10.1002/jcph.259] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 12/26/2013] [Indexed: 11/11/2022]
Abstract
In order to characterize the variation in pharmacokinetics of paracetamol across the human age span, we performed a population pharmacokinetic analysis from preterm neonates to adults with specific focus on clearance. Concentration-time data obtained in 220 neonates (post-natal age 1-76 days, gestational age 27-42 weeks), infants (0.11-1.33 yrs), children (2-7 yrs) and adults (19-34 yrs) were analyzed using NONMEM 7.2. In the covariate analysis, linear functions, power functions, and a power function with a bodyweight-dependent exponent were tested. Between preterm neonates and adults, linear bodyweight functions were identified for Q2, Q3, V1, V2, and V3, while for CL a power function with a bodyweight-dependent exponent k was identified (CLi = CLp × (BW/70)(k) ). The exponent k was found to decrease in a sigmoidal manner with bodyweight from 1.2 to 0.75, with half the decrease in exponent reached at 12.2 kg. No other covariates such as age were identified. A pharmacokinetic model for paracetamol characterizing changes in pharmacokinetic parameters across the pediatric age-range was developed. Clearance was found to change in a nonlinear manner with bodyweight. Based on the final model, dosing guidelines are proposed from preterm neonates to adolescents resulting in similar exposure across all age ranges.
Collapse
Affiliation(s)
- Chenguang Wang
- Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands; Erasmus MC Sophia Children's Hospital, Intensive Care and Department of Paediatric Intensive Care, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|