1
|
Wolthuis DFGJ, Freriksen JJM, Ter Avest M, Kartha RV, de Wildt SN, Wijnsma K, van de Kar NCAJ, Ter Heine R. Model-informed repurposing of eliglustat for treatment and prophylaxis of Shiga toxin-producing Escherichia coli hemolytic-uremic syndrome (STEC-HUS) in children. Pediatr Nephrol 2025; 40:2009-2019. [PMID: 39900743 PMCID: PMC12031897 DOI: 10.1007/s00467-025-06688-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Shiga toxin-producing Escherichia coli hemolytic-uremic syndrome (STEC-HUS) is a severe illness predominantly affecting young children, with limited treatment options beyond supportive care. Eliglustat, approved for Gaucher disease, shows potential in reducing Shiga toxin binding to target glomerular endothelial cells in vitro, prompting interest as a treatment for STEC-HUS. However, it remains unknown what dose is likely to be effective and safe for treatment of STEC-HUS in the pediatric population. We hypothesize that effective and safe levels of eliglustat can be reached in children. METHODS We identified pharmacokinetic targets of efficacy for treatment and prophylaxis of STEC-HUS based on a preclinical model and human cardiac safety data. Then, we developed oral and intravenous dosing regimens using population pharmacokinetic (popPK) simulations based on an existing model enriched to allow extrapolation to a simulated virtual pediatric population. These dosing regimens were then confirmed using a verified physiologically based pharmacokinetic (PBPK) model. RESULTS We simulated, using popPK data, oral and intravenous dosing regimens resulting in adequate target exposure in > 90% of all patients, with minimal expected risk for cardiotoxicity. Confirmation of these dosing regimens with PBPK modeling resulted in very similar exposure, with lower interindividual variability and minimal toxicity potential. CONCLUSIONS Based on pharmacokinetic modeling, we developed oral and intravenous eliglustat dosing regimens that are likely safe and effective for treatment of STEC-HUS and prophylaxis in case of outbreaks of STEC infections. Clinical evaluation of these dosing regimens in children suspected of or diagnosed with STEC-HUS is required and should include assessment of pharmacokinetics, efficacy, and safety (e.g., ECG monitoring).
Collapse
Affiliation(s)
- David F G J Wolthuis
- Department of Internal Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands.
| | | | - Mendy Ter Avest
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, USA
| | - Saskia N de Wildt
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
- Department of Intensive Care, Radboudumc, Nijmegen, The Netherlands
- Department of Neonatal and Pediatric Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Kioa Wijnsma
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Nicole C A J van de Kar
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Ivashchenko DV, Buromskaya NI, Shimanov PV, Shevchenko YS, Sychev DA. Exploring Risk Factors for Adverse Reactions in Children with an Acute Psychotic Episode Using the Global Trigger Tool: Does Age Matter? J Child Adolesc Psychopharmacol 2024; 34:319-326. [PMID: 38716826 DOI: 10.1089/cap.2024.0012] [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] [Indexed: 09/19/2024]
Abstract
Aim: To establish significant risk factors for the development of adverse drug effects (ADEs) in children and adolescents with an acute psychotic episode taking antipsychotics. Materials and Methods: The research team randomly selected 15 patient records each month for 3 years (2016-2018). Overall, 450 patient records were included (223 boys and 227 girls, mean age was 14.52 ± 2.21 years). Adverse effects were identified using the standard algorithm of the Global Trigger Tool method. A "trigger" is an indication that an adverse reaction is likely to occur, e.g., an antihistamine prescription on a prescribing list. When a trigger was detected, the case history was studied in further detail to confirm the occurrence of ADEs. We divided patients into two groups: the "children" group (under 12 years old) and the "adolescents" group (13 years and older). Data were analyzed using the statistical package IBM SPSS Statistics 23.0. Results: Of the 450 patient records, 402 (89.3%) had at least one trigger detected. In total, 126 case histories contained evidence of ADE (28%). The total number of ADEs per 1000 patient days was 5.39 and the number of ADEs per 100 admissions was 32.0. Among adolescents, two or more triggers per patient were significantly more frequently identified (61.3% vs. 44.6%; p = 0.001). ADEs were rare in "Children" compared with "Adolescents" (13.8% vs. 30.4%; p = 0.006). The logistic regression analysis confirmed high predictive role of "Adolescence" (odds ratio [OR] = 2.58; 95% confidence interval [CI] 1.22-5.4; p = 0.013), "Polypharmacy" (OR = 1.96; 95% CI 1.23-3.1; p = 0.004), and "First-life hospitalization" (OR = 2.17; 95% CI 1.34-3.48; p = 0.001) for ADE fact in patient records. Conclusion: We found that significant risk factors for ADEs to antipsychotics in patients with acute psychotic episode were adolescence (13 years and older), polypharmacy, and first-life hospitalization. The fact that children (i.e., younger than 13 years of age) are less likely to experience ADEs was not associated with high-risk drugs or higher doses in our study.
Collapse
Affiliation(s)
- Dmitriy V Ivashchenko
- Child Psychiatry and Psychotherapy Department, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Nina I Buromskaya
- Scientific-Practical Children's and Adolescents Mental Health Center n.a. G.E. Sukhareva, Moscow, Russia
| | - Pavel V Shimanov
- Scientific-Practical Children's and Adolescents Mental Health Center n.a. G.E. Sukhareva, Moscow, Russia
| | - Yuriy S Shevchenko
- Child Psychiatry and Psychotherapy Department, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Dmitriy A Sychev
- Department of Clinical Pharmacology and Therapeutics, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
3
|
Fairman K, Choi MK, Gonnabathula P, Lumen A, Worth A, Paini A, Li M. An Overview of Physiologically-Based Pharmacokinetic Models for Forensic Science. TOXICS 2023; 11:126. [PMID: 36851001 PMCID: PMC9964742 DOI: 10.3390/toxics11020126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
A physiologically-based pharmacokinetic (PBPK) model represents the structural components of the body with physiologically relevant compartments connected via blood flow rates described by mathematical equations to determine drug disposition. PBPK models are used in the pharmaceutical sector for drug development, precision medicine, and the chemical industry to predict safe levels of exposure during the registration of chemical substances. However, one area of application where PBPK models have been scarcely used is forensic science. In this review, we give an overview of PBPK models successfully developed for several illicit drugs and environmental chemicals that could be applied for forensic interpretation, highlighting the gaps, uncertainties, and limitations.
Collapse
Affiliation(s)
- Kiara Fairman
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Me-Kyoung Choi
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Pavani Gonnabathula
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | | | - Miao Li
- Division of Biochemical Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
4
|
Prediction of Drug-Drug-Gene Interaction Scenarios of ( E)-Clomiphene and Its Metabolites Using Physiologically Based Pharmacokinetic Modeling. Pharmaceutics 2022; 14:pharmaceutics14122604. [PMID: 36559098 PMCID: PMC9781104 DOI: 10.3390/pharmaceutics14122604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Clomiphene, a selective estrogen receptor modulator (SERM), has been used for the treatment of anovulation for more than 50 years. However, since (E)-clomiphene ((E)-Clom) and its metabolites are eliminated primarily via Cytochrome P450 (CYP) 2D6 and CYP3A4, exposure can be affected by CYP2D6 polymorphisms and concomitant use with CYP inhibitors. Thus, clomiphene therapy may be susceptible to drug-gene interactions (DGIs), drug-drug interactions (DDIs) and drug-drug-gene interactions (DDGIs). Physiologically based pharmacokinetic (PBPK) modeling is a tool to quantify such DGI and DD(G)I scenarios. This study aimed to develop a whole-body PBPK model of (E)-Clom including three important metabolites to describe and predict DGI and DD(G)I effects. Model performance was evaluated both graphically and by calculating quantitative measures. Here, 90% of predicted Cmax and 80% of AUClast values were within two-fold of the corresponding observed value for DGIs and DD(G)Is with clarithromycin and paroxetine. The model also revealed quantitative contributions of different CYP enzymes to the involved metabolic pathways of (E)-Clom and its metabolites. The developed PBPK model can be employed to assess the exposure of (E)-Clom and its active metabolites in as-yet unexplored DD(G)I scenarios in future studies.
Collapse
|
5
|
Saleem MT, Shoaib MH, Yousuf RI, Ahmed FR, Ahmed K, Siddiqui F, Mahmood ZA, Sikandar M, Imtiaz MS. SeDeM tool-driven full factorial design for osmotic drug delivery of tramadol HCl: Formulation development, physicochemical evaluation, and in-silico PBPK modeling for predictive pharmacokinetic evaluation using GastroPlus™. Front Pharmacol 2022; 13:974715. [PMID: 36278217 PMCID: PMC9585207 DOI: 10.3389/fphar.2022.974715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The study is based on using SeDeM expert system in developing controlled-release tramadol HCl osmotic tablets and its in-silico physiologically based pharmacokinetic (PBPK) modeling for in-vivo pharmacokinetic evaluation. A Quality by Design (QbD) based approach in developing SeDEM-driven full factorial osmotic drug delivery was applied. A 24 Full-factorial design was used to make the trial formulations of tramadol HCl osmotic tablets using NaCl as osmogen, Methocel K4M as rate controlling polymer, and avicel pH 101 as diluent. The preformulation characteristics of formulations (F1-F16) were determined by applying SeDeM Expert Tool. The formulation was optimized followed by in-vivo predictive pharmacokinetic assessment using PBPK “ACAT” model of GastroPlus™. The FTIR results showed no interaction among the ingredients. The index of good compressibility (ICG) values of all trial formulation blends were ≥5, suggesting direct compression is the best-suited method. Formulation F3 and F4 were optimized based on drug release at 2, 10, and 16 h with a zero-order kinetic release (r2 = 0.992 and 0.994). The SEM images confirmed micropores formation on the surface of the osmotic tablet after complete drug release. F3 and F4 were also stable (shelf life 29.41 and 23.46 months). The in vivo simulation of the pharmacokinetics of the PBPK in-silico model revealed excellent relative bioavailability of F3 and F4 with reference to tramadol HCl 50 mg IR formulations. The SeDeM expert tool was best utilized to evaluate the compression characteristics of selected formulation excipients and their blends for direct compression method in designing once-daily osmotically controlled-release tramadol HCl tablets. The in-silico GastroPlus™ PBPK modeling provided a thorough pharmacokinetic assessment of the optimized formulation as an alternative to tramadol HCl in vivo studies.
Collapse
|
6
|
Simeoli R, Cairoli S, Decembrino N, Campi F, Dionisi Vici C, Corona A, Goffredo BM. Use of Antibiotics in Preterm Newborns. Antibiotics (Basel) 2022; 11:antibiotics11091142. [PMID: 36139921 PMCID: PMC9495226 DOI: 10.3390/antibiotics11091142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Due to complex maturational and physiological changes that characterize neonates and affect their response to pharmacological treatments, neonatal pharmacology is different from children and adults and deserves particular attention. Although preterms are usually considered part of the neonatal population, they have physiological and pharmacological hallmarks different from full-terms and, therefore, need specific considerations. Antibiotics are widely used among preterms. In fact, during their stay in neonatal intensive care units (NICUs), invasive procedures, including central catheters for parental nutrition and ventilators for respiratory support, are often sources of microbes and require antimicrobial treatments. Unfortunately, the majority of drugs administered to neonates are off-label due to the lack of clinical studies conducted on this special population. In fact, physiological and ethical concerns represent a huge limit in performing pharmacokinetic (PK) studies on these subjects, since they limit the number and volume of blood sampling. Therapeutic drug monitoring (TDM) is a useful tool that allows dose adjustments aiming to fit plasma concentrations within the therapeutic range and to reach specific drug target attainment. In this review of the last ten years’ literature, we performed Pubmed research aiming to summarize the PK aspects for the most used antibiotics in preterms.
Collapse
Affiliation(s)
- Raffaele Simeoli
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Sara Cairoli
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Nunzia Decembrino
- Neonatal Intensive Care Unit, University Hospital “Policlinico-San Marco” Catania, Integrated Department for Maternal and Child’s Health Protection, 95100 Catania, Italy
| | - Francesca Campi
- Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Carlo Dionisi Vici
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Alberto Corona
- ICU and Accident & Emergency Department, ASST Valcamonica, 25043 Breno, Italy
| | - Bianca Maria Goffredo
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: ; Tel.: +39-0668592174; Fax: + 39-0668593009
| |
Collapse
|
7
|
Chen SY, Ourshalimian S, Kim E, Russell CJ, Kelley-Quon LI. Tramadol Use in Pediatric Surgery: Trends After the Food and Drug Administration Black-Box Warning. J Surg Res 2022; 280:10-18. [PMID: 35944445 DOI: 10.1016/j.jss.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 10/15/2022]
Abstract
INTRODUCTION The U.S. Food and Drug Administration (FDA) issued a black-box warning in 2017 contraindicating tramadol in children <12 y. Longitudinal trends and factors associated with perioperative tramadol use in children remain unclear. METHODS A retrospective, multi-institutional cohort study utilizing the Pediatric Health Information System database was performed for children 2-18 y who underwent one of ten common surgeries from 1/2009-2/2020. Temporal trends correlated with the FDA tramadol contraindication were evaluated. Hierarchical multivariable logistic regression analysis identified factors associated with tramadol use. RESULTS Of 477,153 children undergoing surgery, 5857(1.2%) received tramadol during hospitalization. Tramadol use occurred in 942 (16.1%) children after the black-box warning, 390 of whom were <12 y. For children <12 y, annual tramadol use peaked at 1.87% (2016) and decreased to 0.66% (2019). Female sex (odds ratio OR 1.32; 95% confidence interval CI:1.24,1.40), age ≥12 y (OR 2.79; 95%CI: 2.62,2.97), and Midwest location (OR 4.07; 95% CI:1.64,10.11) increased likelihood of receiving tramadol. Tramadol use was more likely after cholecystectomy (OR 1.17; 95% CI:1.04,1.32) and in children with gastrointestinal (OR 2.39; 95% CI: 2.19,2.60), metabolic (OR 1.39; 95% CI:1.26,1.53) or transplant-related (OR 1.82; 95% CI: 1.57,2.10) comorbidities. Children of Hispanic/Latino ethnicity and those with public insurance had decreased likelihood of receiving tramadol. Adjusting for patient and hospital characteristics, children <12 y were less likely to receive tramadol following the black-box warning (OR 0.65; 95% CI: 0.59,0.70). CONCLUSIONS Despite the FDA contraindication, tramadol prescribing continues among children <12 y undergoing surgery, with use varying by patient and institutional factors. Interventions are required to reduce perioperative tramadol use in children.
Collapse
Affiliation(s)
- Stephanie Y Chen
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Shadassa Ourshalimian
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Eugene Kim
- Division of Pain Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Christopher J Russell
- Division of Hospital Medicine, Children's Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Lorraine I Kelley-Quon
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California; Department of Population and Public Health Sciences, University of Southern California, Los Angeles, California.
| |
Collapse
|
8
|
Wollmer E, Ungell AL, Nicolas JM, Klein S. Review of paediatric gastrointestinal physiology relevant to the absorption of orally administered medicines. Adv Drug Deliv Rev 2022; 181:114084. [PMID: 34929252 DOI: 10.1016/j.addr.2021.114084] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/13/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Despite much progress in regulations to improve paediatric drug development, there remains a significant need to develop better medications for children. For the design of oral dosage forms, a detailed understanding of the specific gastrointestinal (GI) conditions in children of different age categories and how they differ from GI conditions in adults is essential. Several review articles have been published addressing the ontogeny of GI characteristics, including luminal conditions in the GI tract of children. However, the data reported in most of these reviews are of limited quality because (1) information was cited from very old publications and sometimes low quality sources, (2) data gaps in the original data were filled with textbook knowledge, (3) data obtained on healthy and sick children were mixed, (4) average data obtained on groups of patients were mixed with data obtained on individual patients, and (5) results obtained using investigative techniques that may have altered the outcome of the respective studies were considered. Consequently, many of these reviews draw conclusions that may be incorrect. The aim of the present review was to provide a comprehensive and updated overview of the available original data on the ontogeny of GI luminal conditions relevant to oral drug absorption in the paediatric population. To this end, the PubMed and Web of Science metadatabases were searched for appropriate studies that examined age-related conditions in the oral cavity, esophagus, stomach, small intestine, and colon. Maturation was observed for several GI parameters, and corresponding data sets were identified for each paediatric age group. However, it also became clear that the ontogeny of several GI traits in the paediatric population is not yet known. The review article provides a robust and valuable data set for the development of paediatric in vitro and in silico biopharmaceutical tools to support the development of age-appropriate dosage forms. In addition, it provides important information on existing data gaps and should provide impetus for further systematic and well-designed in vivo studies on GI physiology in children of specific age groups in order to close existing knowledge gaps and to sustainably improve oral drug therapy in children.
Collapse
|
9
|
Long T, Cristofoletti R, Cicali B, Michaud V, Dow P, Turgeon J, Schmidt S. Physiologically-based Pharmacokinetic Modeling to Assess the Impact of CYP2D6-Mediated Drug-Drug Interactions on Tramadol and O-Desmethyltramadol Exposures via Allosteric and Competitive Inhibition. J Clin Pharmacol 2021; 62:76-86. [PMID: 34383318 PMCID: PMC9293201 DOI: 10.1002/jcph.1951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/06/2021] [Indexed: 11/11/2022]
Abstract
Tramadol is an opioid medication used to treat moderately severe pain. Cytochrome P450 (CYP) 2D6 inhibition could be important for tramadol, as it decreases the formation of its pharmacologically active metabolite, O‐desmethyltramadol, potentially resulting in increased opioid use and misuse. The objective of this study was to evaluate the impact of allosteric and competitive CYP2D6 inhibition on tramadol and O‐desmethyltramadol pharmacokinetics using quinidine and metoprolol as prototypical perpetrator drugs. A physiologically based pharmacokinetic model for tramadol and O‐desmethyltramadol was developed and verified in PK‐Sim version 8 and linked to respective models of quinidine and metoprolol to evaluate the impact of allosteric and competitive CYP2D6 inhibition on tramadol and O‐desmethyltramadol exposure. Our results show that there is a differentiated impact of CYP2D6 inhibitors on tramadol and O‐desmethyltramadol based on their mechanisms of inhibition. Following allosteric inhibition by a single dose of quinidine, the exposure of both tramadol (51% increase) and O‐desmethyltramadol (52% decrease) was predicted to be significantly altered after concomitant administration of a single dose of tramadol. Following multiple‐dose administration of tramadol and a single‐dose or multiple‐dose administration of quinidine, the inhibitory effect of quinidine was predicted to be long (≈42 hours) and to alter exposure of tramadol and O‐desmethyltramadol by up to 60%, suggesting that coadministration of quinidine and tramadol should be avoided clinically. In comparison, there is no predicted significant impact of metoprolol on tramadol and O‐desmethyltramadol exposure. In fact, tramadol is predicted to act as a CYP2D6 perpetrator and increase metoprolol exposure, which may necessitate the need for dose separation.
Collapse
Affiliation(s)
- Tao Long
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Brian Cicali
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Veronique Michaud
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL, USA.,Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada
| | - Pamela Dow
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL, USA
| | - Jacques Turgeon
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL, USA.,Faculty of Pharmacy, Université de Montréal, Montréal, Quebec, Canada
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
10
|
Chapron BD, Chapron A, Leeder JS. Recent advances in the ontogeny of drug disposition. Br J Clin Pharmacol 2021; 88:4267-4284. [PMID: 33733546 DOI: 10.1111/bcp.14821] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Developmental changes that occur throughout childhood have long been known to impact drug disposition. However, pharmacokinetic studies in the paediatric population have historically been limited due to ethical concerns arising from incorporating children into clinical trials. As such, much of the early work in the field of developmental pharmacology was reliant on difficult-to-interpret in vitro and in vivo animal studies. Over the last 2 decades, our understanding of the mechanistic processes underlying age-related changes in drug disposition has advanced considerably. Progress has largely been driven by technological advances in mass spectrometry-based methods for quantifying proteins implicated in drug disposition, and in silico tools that leverage these data to predict age-related changes in pharmacokinetics. This review summarizes our current understanding of the impact of childhood development on drug disposition, particularly focusing on research of the past 20 years, but also highlighting select examples of earlier foundational research. Equally important to the studies reviewed herein are the areas that we cannot currently describe due to the lack of research evidence; these gaps provide a map of drug disposition pathways for which developmental trends still need to be characterized.
Collapse
Affiliation(s)
- Brian D Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Alenka Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA.,Schools of Medicine and Pharmacy, University of Missouri-Kansas City, MO, USA
| |
Collapse
|
11
|
Lang J, Vincent L, Chenel M, Ogungbenro K, Galetin A. Impact of Hepatic CYP3A4 Ontogeny Functions on Drug–Drug Interaction Risk in Pediatric Physiologically‐Based Pharmacokinetic/Pharmacodynamic Modeling: Critical Literature Review and Ivabradine Case Study. Clin Pharmacol Ther 2020; 109:1618-1630. [DOI: 10.1002/cpt.2134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Jennifer Lang
- Centre for Applied Pharmacokinetic Research Division of Pharmacy and Optometry, School of Health Sciences Faculty of Biology, Medicine and Health Manchester Academic Health Science Centre University of Manchester Manchester UK
| | - Ludwig Vincent
- Centre de Pharmacocinétique et Métabolisme Technologie Servier Orléans France
| | - Marylore Chenel
- Clinical Pharmacokinetics and Pharmacometrics Institut de Recherches Internationales Servier Suresnes France
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research Division of Pharmacy and Optometry, School of Health Sciences Faculty of Biology, Medicine and Health Manchester Academic Health Science Centre University of Manchester Manchester UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research Division of Pharmacy and Optometry, School of Health Sciences Faculty of Biology, Medicine and Health Manchester Academic Health Science Centre University of Manchester Manchester UK
| |
Collapse
|
12
|
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
|
13
|
Toward precision medicine in pediatric population using cytochrome P450 phenotyping approaches and physiologically based pharmacokinetic modeling. Pediatr Res 2020; 87:441-449. [PMID: 31600772 DOI: 10.1038/s41390-019-0609-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 01/18/2023]
Abstract
The activity of drug-metabolizing enzymes (DME) shows high inter- and intra-individual variability. Genetic polymorphisms, exposure to drugs, and environmental toxins are known to significantly alter DME expression. In addition, the activity of these enzymes is highly age-dependent due to maturation processes that occur during development. Currently, there is a vast choice of phenotyping methods in adults using exogenous probes to characterize the activity of these enzymes. However, this can hardly be applied to children since it requires the intake of non-therapeutic xenobiotics. In addition, sampling may be challenging in the pediatric population for a variety of reasons: limited volume (e.g., blood), inappropriate sampling methods for age (e.g., urine), and metric requiring invasive or multiple blood samples. This review covers the main existing methods that can be used in the pediatric population to determine DME activity, with a particular focus on cytochrome P450 enzymes. Less invasive tools are described, including phenotyping using endogenous probes. Finally, the potential of pediatric model-informed precision dosing using physiologically based pharmacokinetic modeling is discussed.
Collapse
|
14
|
Evaluation of the Effect of CYP2D6 Genotypes on Tramadol and O-Desmethyltramadol Pharmacokinetic Profiles in a Korean Population Using Physiologically-Based Pharmacokinetic Modeling. Pharmaceutics 2019; 11:pharmaceutics11110618. [PMID: 31744222 PMCID: PMC6920759 DOI: 10.3390/pharmaceutics11110618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 01/04/2023] Open
Abstract
Tramadol is a μ-opioid receptor agonist and a monoamine reuptake inhibitor. O-desmethyltramadol (M1), the major active metabolite of tramadol, is produced by CYP2D6. A physiologically-based pharmacokinetic model was developed to predict changes in time-concentration profiles for tramadol and M1 according to dosage and CYP2D6 genotypes in the Korean population. Parallel artificial membrane permeation assay was performed to determine tramadol permeability, and the metabolic clearance of M1 was determined using human liver microsomes. Clinical study data were used to develop the model. Other physicochemical and pharmacokinetic parameters were obtained from the literature. Simulations for plasma concentrations of tramadol and M1 (after 100 mg tramadol was administered five times at 12-h intervals) were based on a total of 1000 virtual healthy Koreans using SimCYP® simulator. Geometric mean ratios (90% confidence intervals) (predicted/observed) for maximum plasma concentration at steady-state (Cmax,ss) and area under the curve at steady-state (AUClast,ss) were 0.79 (0.69-0.91) and 1.04 (0.85-1.28) for tramadol, and 0.63 (0.51-0.79) and 0.67 (0.54-0.84) for M1, respectively. The predicted time-concentration profiles of tramadol fitted well to observed profiles and those of M1 showed under-prediction. The developed model could be applied to predict concentration-dependent toxicities according to CYP2D6 genotypes and also, CYP2D6-related drug interactions.
Collapse
|
15
|
Quantitative mass spectrometry-based proteomics in the era of model-informed drug development: Applications in translational pharmacology and recommendations for best practice. Pharmacol Ther 2019; 203:107397. [DOI: 10.1016/j.pharmthera.2019.107397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/29/2019] [Indexed: 02/08/2023]
|
16
|
Yun YE, Edginton AN. Model qualification of the PK-Sim® pediatric module for pediatric exposure assessment of CYP450 metabolized compounds. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:789-814. [PMID: 31405354 DOI: 10.1080/15287394.2019.1652215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pediatric physiologically based pharmacokinetic (PBPK) models facilitate the estimation of pharmacokinetic (PK) parameters in children under specific exposure conditions. In human health risk assessment, PBPK modeling has been used to determine a chemical-specific human kinetic adjustment factor (HKAF). Due to increased demands in regulatory assessment, model evaluation and qualification have gained growing attention. The aim of this study was to undertake model qualification of pediatric PBPK models for compounds that are primarily metabolized by cytochrome P450 (CYP) enzymes. The objectives were to determine the appropriateness of the virtual individual creating algorithm in PK-Sim® in predicting PK parameters and their variability in children and identify critical system-specific inputs. PBPK models in adults were constructed for several pharmaceuticals (grouped by major clearance process such as CYP3A4). Several age groups of virtual individuals were created to represent children in pediatric clinical studies. The mean and variance of clearance (CL) from virtual populations were compared to observed values. Sensitivity analysis on area under the curve (AUC) was performed. System-specific parameters of virtual children that contribute to inter-individual PK properties were assessed. Eighty-one percent of the comparisons between simulated and observed clearance values were within twofold error. The mean fold errors were 1.1, 1, 0.7 and 1.8 in adolescents, children, infants and neonates, respectively. CL variability was reasonably predicted for 70% of the comparisons with comparable coefficients of variation between observed and predicted. The sensitivity analysis revealed that fraction unbound in plasma, parameters related to CYP enzyme-mediated metabolism and liver volumewere most important in the estimation of pediatric exposure. A comparison of variabilities in weight, height and liver volume in virtual children showed reliable agreement with observed data. The presented results of predictive performance and properties of virtual populations provide confidence in the use of PK-Sim for pediatric PBPK modeling in toxicological applications including PBPK-based-HKAF derivation.
Collapse
Affiliation(s)
- Yejin Esther Yun
- School of Pharmacy, University of Waterloo , Waterloo , Ontario , Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo , Waterloo , Ontario , Canada
| |
Collapse
|
17
|
Smits A, De Cock P, Vermeulen A, Allegaert K. Physiologically based pharmacokinetic (PBPK) modeling and simulation in neonatal drug development: how clinicians can contribute. Expert Opin Drug Metab Toxicol 2018; 15:25-34. [PMID: 30554542 DOI: 10.1080/17425255.2019.1558205] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Legal initiatives to stimulate neonatal drug development should be accompanied by development of valid research tools. Physiologically based (PB)-pharmacokinetic (PK) modeling and simulation are established tools, accepted by regulatory authorities. Consequently, PBPK holds promise to be a strong research tool to support neonatal drug development. Area covered: The currently available PBPK models still have poor predictive performance in neonates. Using an illustrative approach on distinct PK processes of absorption, distribution, metabolism, excretion, and real-world data in neonates, we provide evidence on the need to further refine available PBPK system parameters through generation and integration of new knowledge. This necessitates cross talk between clinicians and modelers to integrate knowledge (PK datasets, system knowledge, maturational physiology) or test and refine PBPK models. Expert opinion: Besides refining these models for 'small molecules', PBPK model development should also be more widely applied for therapeutic proteins and to determine exposure through breastfeeding. Researchers should also be aware that PBPK modeling in combination with clinical observations can also be used to elucidate age-related changes that are almost impossible to study based on in vivo or in vitro data. This approach has been explored for hepatic biliary excretion, renal tubular activity, and central nervous system exposure.
Collapse
Affiliation(s)
- Anne Smits
- a Neonatal Intensive Care Unit , University Hospitals Leuven , Leuven , Belgium.,b Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Pieter De Cock
- c Department of Pharmacy , Ghent University Hospital , Ghent , Belgium.,d Heymans Institute of Pharmacology , Ghent University , Ghent , Belgium.,e Department of Pediatric Intensive Care , Ghent University , Ghent , Belgium
| | - An Vermeulen
- f Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium
| | - Karel Allegaert
- b Department of Development and Regeneration , KU Leuven , Leuven , Belgium.,g Department of Pediatrics, Division of Neonatology , Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam , Rotterdam , The Netherlands
| |
Collapse
|
18
|
T'jollyn H, Vermeulen A, Van Bocxlaer J. PBPK and its Virtual Populations: the Impact of Physiology on Pediatric Pharmacokinetic Predictions of Tramadol. AAPS JOURNAL 2018; 21:8. [PMID: 30498862 DOI: 10.1208/s12248-018-0277-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/09/2018] [Indexed: 11/30/2022]
Abstract
In pediatric PBPK models, age-related changes in the body are known to occur. Given the sparsity of and the variability associated with relevant physiological parameters, different PBPK software providers may vary in their system's data. In this work, three commercially available PBPK software packages (PK-Sim®, Simcyp®, and Gastroplus®) were investigated regarding their differences in system-related information, possibly affecting clearance prediction. Three retrograde PBPK clearance models were set up to enable prediction of pediatric tramadol clearance. These models were qualified in terms of total, CYP2D6, and renal clearance in adults. Tramadol pediatric clearance predictions from PBPK were compared with a pooled popPK model covering clearance ranging from neonates to adults. Fold prediction errors were used to evaluate the results. Marked differences in liver clearance prediction between PBPK models were observed. In general, the prediction bias of total clearance was greatest at the youngest population and decreased with age. Regarding CYP2D6 and renal clearance, important differences exist between PBPK software tools. Interestingly, the PBPK model with the shortest CYP2D6 maturation half-life (PK-Sim) agreed best with the in vivo CYP2D6 maturation model. Marked differences in physiological data explain the observed differences in hepatic clearance prediction in early life between the various PBPK software providers tested. Consensus on the most suited pediatric data to use should harmonize and optimize pediatric clearance predictions. Moreover, the combination of bottom-up and top-down approaches, using a convenient probe substrate, has the potential to update system-related parameters in order to better represent pediatric physiology.
Collapse
Affiliation(s)
- Huybrecht T'jollyn
- A Division of Janssen Pharmaceutica NV, Quantitative Sciences, Janssen Research and Development, Beerse, Belgium.
| | - An Vermeulen
- A Division of Janssen Pharmaceutica NV, Quantitative Sciences, Janssen Research and Development, Beerse, Belgium.,Faculty of Pharmaceutical Sciences, Laboratory of Medical Biochemistry and Clinical Analysis, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Jan Van Bocxlaer
- Faculty of Pharmaceutical Sciences, Laboratory of Medical Biochemistry and Clinical Analysis, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| |
Collapse
|
19
|
Clopidogrel Pharmacokinetics in Malaysian Population Groups: The Impact of Inter-Ethnic Variability. Pharmaceuticals (Basel) 2018; 11:ph11030074. [PMID: 30049953 PMCID: PMC6161187 DOI: 10.3390/ph11030074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/08/2018] [Indexed: 12/13/2022] Open
Abstract
Malaysia is a multi-ethnic society whereby the impact of pharmacogenetic differences between ethnic groups may contribute significantly to variability in clinical therapy. One of the leading causes of mortality in Malaysia is cardiovascular disease (CVD), which accounts for up to 26% of all hospital deaths annually. Clopidogrel is used as an adjunct treatment in the secondary prevention of cardiovascular events. CYP2C19 plays an integral part in the metabolism of clopidogrel to the active metabolite clopi-H4. However, CYP2C19 genetic polymorphism, prominent in Malaysians, could influence target clopi-H4 plasma concentrations for clinical efficacy. This study addresses how inter-ethnicity variability within the Malaysian population impacts the attainment of clopi-H4 target plasma concentration under different CYP2C19 polymorphisms through pharmacokinetic (PK) modelling. We illustrated a statistically significant difference (P < 0.001) in the clopi-H4 Cmax between the extensive metabolisers (EM) and poor metabolisers (PM) phenotypes with either Malay or Malaysian Chinese population groups. Furthermore, the number of PM individuals with peak clopi-H4 concentrations below the minimum therapeutic level was partially recovered using a high-dose strategy (600 mg loading dose followed by a 150 mg maintenance dose), which resulted in an approximate 50% increase in subjects attaining the minimum clopi-H4 plasma concentration for a therapeutic effect.
Collapse
|
20
|
Nguyen PTT, Parvez MM, Kim MJ, Ho Lee J, Ahn S, Ghim JL, Shin JG. Development of a Physiologically Based Pharmacokinetic Model of Ethionamide in the Pediatric Population by Integrating Flavin-Containing Monooxygenase 3 Maturational Changes Over Time. J Clin Pharmacol 2018; 58:1347-1360. [PMID: 29878384 DOI: 10.1002/jcph.1133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 11/06/2022]
Abstract
Currently, ethionamide is the most frequently prescribed second-line antituberculosis drug in children. After extensive metabolism by flavin-containing monooxygenase (FMO) isoform 3 in the liver, the drug may exert cytotoxic effects. The comparison of children in different age groups revealed a significant age-related increase in ethionamide elimination in vivo. However, to date, the exact mechanism underlying this dynamic increase in ethionamide elimination has not been elucidated. We hypothesized that the age-dependent changes in ethionamide elimination were predominantly a result of the progressive increases in the expression and metabolic capacity of FMO3 during childhood. To test this hypothesis, a full physiologically based pharmacokinetic (PBPK) model of ethionamide was established and validated in adults through incorporation of comprehensive metabolism and transporter profiles, then expanded to the pediatric population through integration of FMO3 maturational changes over time. Thus, a good prediction PBPK model was validated successfully both in adults and children and applied to demonstrate the critical contribution of FMO3 in the mechanistic elimination pathway of ethionamide. In addition, a significant correlation between clearance and age was observed in children by accounting for ethionamide maturation, which could offer a mechanistic understanding of the age-associated changes in ethionamide elimination. In conclusion, this study underlines the benefits of in vitro-in vivo extrapolation and a quantitative PBPK approach for the investigation of transporter-enzyme interplay in ethionamide disposition and the demonstration of FMO3 ontogeny in children.
Collapse
Affiliation(s)
- Phuong Thi Thu Nguyen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Faculty of Pharmacy, Hai Phong University of Medicine and Pharmacy, Haiphong, Vietnam
| | - Md Masud Parvez
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Min Jung Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jung Ho Lee
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Sangzin Ahn
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jong-Lyul Ghim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| |
Collapse
|
21
|
Martinez MN, Gehring R, Mochel JP, Pade D, Pelligand L. Population variability in animal health: Influence on dose-exposure-response relationships: Part II: Modelling and simulation. J Vet Pharmacol Ther 2018; 41:E68-E76. [PMID: 29806231 DOI: 10.1111/jvp.12666] [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: 02/07/2018] [Accepted: 04/15/2018] [Indexed: 11/29/2022]
Abstract
During the 2017 Biennial meeting, the American Academy of Veterinary Pharmacology and Therapeutics hosted a 1-day session on the influence of population variability on dose-exposure-response relationships. In Part I, we highlighted some of the sources of population variability. Part II provides a summary of discussions on modelling and simulation tools that utilize existing pharmacokinetic data, can integrate drug physicochemical characteristics with species physiological characteristics and dosing information or that combine observed with predicted and in vitro information to explore and describe sources of variability that may influence the safe and effective use of veterinary pharmaceuticals.
Collapse
Affiliation(s)
- Marilyn N Martinez
- Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland
| | - Ronette Gehring
- Utrecht Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jonathan P Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| | | | - Ludovic Pelligand
- Department of Clinical Services and Sciences and Department of Comparative Biomedical Sciences, The Royal Veterinary College, Hatfield, UK
| |
Collapse
|
22
|
Rodieux F, Vutskits L, Posfay-Barbe KM, Habre W, Piguet V, Desmeules JA, Samer CF. When the Safe Alternative Is Not That Safe: Tramadol Prescribing in Children. Front Pharmacol 2018; 9:148. [PMID: 29556194 PMCID: PMC5844975 DOI: 10.3389/fphar.2018.00148] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/13/2018] [Indexed: 01/10/2023] Open
Abstract
Children represent a vulnerable population in which management of nociceptive pain is complex. Drug responses in children differ from adults due to age-related differences. Moreover, therapeutic choices are limited by the lack of indication for a number of analgesic drugs due to the challenge of conducting clinical trials in children. Furthermore the assessment of efficacy as well as tolerance may be complicated by children's inability to communicate properly. According to the World Health Organization, weak opioids such as tramadol and codeine, may be used in addition to paracetamol and ibuprofen for moderate nociceptive pain in both children and adults. However, codeine prescription has been restricted for the last 5 years in children because of the risk of fatal overdoses linked to the variable activity of cytochrome P450 (CYP) 2D6 which bioactivates codeine. Even though tramadol has been considered a safe alternative to codeine, it is well established that tramadol pharmacodynamic opioid effects, efficacy and safety, are also largely influenced by CYP2D6 activity. For this reason, the US Food and Drug Administration recently released a boxed warning regarding the use of tramadol in children. To provide safe and effective tramadol prescription in children, a personalized approach, with dose adaptation according to CYP2D6 activity, would certainly be the safest method. We therefore recommend this approach in children requiring chronic or recurrent nociceptive pain treatment with tramadol. In case of acute inpatients nociceptive pain management, prescribing tramadol at the minimal effective dose, in a child appropriate dosage form and after clear instructions are given to the parents, remains reasonable based on current data. In all other situations, morphine should be preferred for moderate to severe nociceptive pain conditions.
Collapse
Affiliation(s)
- Frédérique Rodieux
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Laszlo Vutskits
- Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,Department of Basic Neuroscience, Faculty of Medicine, University of GenevaGeneva, Switzerland.,Division of Anesthesiology, Unit for Pediatric Anesthesia, Children's Hospitals of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Klara M Posfay-Barbe
- Pediatric Infectious Diseases Unit, Department of Pediatrics, Children's Hospital of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Walid Habre
- Division of Anesthesiology, Unit for Pediatric Anesthesia, Children's Hospitals of Geneva, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,Anesthesiological Investigations Unit, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Valérie Piguet
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| | - Jules A Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of LausanneGeneva, Switzerland
| | - Caroline F Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, University of GenevaGeneva, Switzerland
| |
Collapse
|
23
|
Templeton IE, Jones NS, Musib L. Pediatric Dose Selection and Utility of PBPK in Determining Dose. AAPS JOURNAL 2018; 20:31. [DOI: 10.1208/s12248-018-0187-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/04/2018] [Indexed: 11/30/2022]
|
24
|
Zhang T, Zhang K, Ma L, Li Z, Wang J, Zhang Y, Lu C, Zhu M, Zhuang X. Metabolic Pathway of Icotinib In Vitro: The Differential Roles of CYP3A4, CYP3A5, and CYP1A2 on Potential Pharmacokinetic Drug-Drug Interaction. J Pharm Sci 2017; 107:979-983. [PMID: 29247736 DOI: 10.1016/j.xphs.2017.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/13/2017] [Accepted: 12/06/2017] [Indexed: 11/15/2022]
Abstract
Icotinib is the first self-developed small molecule drug in China for targeted therapy of non-small cell lung cancer. To date, systematic studies on the pharmacokinetic drug-drug interaction of icotinib were limited. By identifying metabolite generated in human liver microsomes and revealing the contributions of major cytochromes P450 (CYPs) in the formation of major metabolites, the aim of the present work was to understand the mechanisms underlying pharmacokinetic and pharmacological variability in clinic. A liquid chromatography/UV/high-resolution mass spectrometer method was developed to characterize the icotinib metabolites. The formation of 6 major metabolites was studied in recombinant CYP isozymes and human liver microsomes with specific inhibitors to identify the CYPs responsible for icotinib metabolism. The metabolic pathways observed in vitro are consistent with those observed in human. Results demonstrated that the metabolites are predominantly catalyzed by CYP3A4 (77%∼87%), with a moderate contribution from CYP3A5 (5%∼15%) and CYP1A2 (3.7%∼7.5%). The contribution of CYP2C8, 2C9, 2C19, and 2D6 is insignificant. Based on our observations, to minimize drug-drug interaction risk in clinic, coprescription of icotinib with strong CYP3A inhibitors or inducers must be weighed. CYP1A2, a highly inducible enzyme in the smoking population, may also represent a determinant of pharmacokinetic and pharmacological variability of icotinib, especially in lung cancer patients with smoking history.
Collapse
Affiliation(s)
- TianHong Zhang
- Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | | | - Li Ma
- Pharmacetucial Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey 08540
| | - Zheng Li
- Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Juan Wang
- Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - YunXia Zhang
- Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Chuang Lu
- Sanofi Company, Waltham, Massachusetts 02451
| | - Mingshe Zhu
- Pharmacetucial Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey 08540; MassDefect Technologies, Princeton, New Jersey 08540.
| | - XiaoMei Zhuang
- Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
25
|
Zhou W, Johnson TN, Bui KH, Cheung SYA, Li J, Xu H, Al-Huniti N, Zhou D. Predictive Performance of Physiologically Based Pharmacokinetic (PBPK) Modeling of Drugs Extensively Metabolized by Major Cytochrome P450s in Children. Clin Pharmacol Ther 2017; 104:188-200. [PMID: 29027194 DOI: 10.1002/cpt.905] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
The accuracy of physiologically based pharmacokinetic (PBPK) model prediction in children, especially those younger than 2 years old, has not been systematically evaluated. The aim of this study was to characterize the pediatric predictive performance of the PBPK approach for 10 drugs extensively metabolized by CYP1A2 (theophylline), CYP2C8 (desloratidine, montelukast), CYP2C9 (diclofenac), CYP2C19 (esomeprazole, lansoprazole), CYP2D6 (tramadol), and CYP3A4 (itraconazole, ondansetron, sufentanil). Model performance in children was evaluated by comparing simulated plasma concentration-time profiles with observed clinical results for each drug and age group. PBPK models reasonably predicted the pharmacokinetics of desloratadine, diclofenac, itraconazole, lansoprazole, montelukast, ondansetron, sufentanil, theophylline, and tramadol across all age groups. Collectively, 58 out of 67 predictions were within 2-fold and 43 out of 67 predictions within 1.5-fold of observed values. Developed PBPK models can reasonably predict exposure in children age 1 month and older for an array of predominantly CYP metabolized drugs.
Collapse
Affiliation(s)
- Wangda Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | | | - Khanh H Bui
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - S Y Amy Cheung
- Quantitative Clinical Pharmacology, AstraZeneca, Cambridge, UK
| | - Jianguo Li
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Hongmei Xu
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Nidal Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Diansong Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| |
Collapse
|
26
|
Duan P, Fisher JW, Yoshida K, Zhang L, Burckart GJ, Wang J. Physiologically Based Pharmacokinetic Prediction of Linezolid and Emtricitabine in Neonates and Infants. Clin Pharmacokinet 2017; 56:383-394. [PMID: 27596256 DOI: 10.1007/s40262-016-0445-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Modeling and simulation approaches are increasingly being utilized in pediatric drug development. Physiologically based pharmacokinetic (PBPK) modeling offers an enhanced ability to predict age-related changes in pharmacokinetics in the pediatric population. METHODS In the current study, adult PBPK models were developed for the renally excreted drugs linezolid and emtricitabine. PBPK models were then utilized to predict pharmacokinetics in pediatric patients for various age groups from the oldest to the youngest patients in a stepwise approach. RESULTS Pharmacokinetic predictions for these two drugs in the pediatric population, including infants and neonates, were within a twofold range of clinical observations. Based on this study, linezolid and emtricitabine pediatric PBPK models incorporating the ontogeny in renal maturation describe the pharmacokinetic differences between adult and pediatric populations, even though the contribution of renal clearance to the total clearance of two drugs was very different (30 % for linezolid vs. 86 % for emtricitabine). CONCLUSION These results suggest that PBPK modeling may provide one option to help predict the pharmacokinetics of renally excreted drugs in neonates and infants.
Collapse
Affiliation(s)
- Peng Duan
- Office of New Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jeffrey W Fisher
- National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Kenta Yoshida
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Building 51, Rm 2154, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Building 51, Rm 2154, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Building 51, Rm 2154, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jian Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Building 51, Rm 2154, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
27
|
Marsousi N, Desmeules JA, Rudaz S, Daali Y. Usefulness of PBPK Modeling in Incorporation of Clinical Conditions in Personalized Medicine. J Pharm Sci 2017; 106:2380-2391. [DOI: 10.1016/j.xphs.2017.04.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/14/2022]
|
28
|
Nicolas JM, Bouzom F, Hugues C, Ungell AL. Oral drug absorption in pediatrics: the intestinal wall, its developmental changes and current tools for predictions. Biopharm Drug Dispos 2017; 38:209-230. [PMID: 27976409 PMCID: PMC5516238 DOI: 10.1002/bdd.2052] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 11/21/2016] [Accepted: 11/30/2016] [Indexed: 12/14/2022]
Abstract
The dissolution, intestinal absorption and presystemic metabolism of a drug depend on its physicochemical characteristics but also on numerous physiological (e.g. gastrointestinal pH, volume, transit time, morphology) and biochemical factors (e.g. luminal enzymes and flora, intestinal wall enzymes and transporters). Over the past decade, evidence has accumulated indicating that these factors may differ in children and adults resulting in age-related changes in drug exposure and drug response. Thus, drug dosage may require adjustment for the pediatric population to ensure the desired therapeutic outcome and to avoid side-effects. Although tremendous progress has been made in understanding the effects of age on intestinal physiology and function, significant knowledge gaps remain. Studying and predicting pharmacokinetics in pediatric patients remains challenging due to ethical concerns associated with clinical trials in this vulnerable population, and because of the paucity of predictive in vitro and in vivo animal assays. This review details the current knowledge related to developmental changes determining intestinal drug absorption and pre-systemic metabolism. Supporting experimental approaches as well as physiologically based pharmacokinetic modeling are also discussed together with their limitations and challenges. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jean-Marie Nicolas
- Non-Clinical Development Department, UCB Biopharma sprl, Braine-l'Alleud, Belgium
| | - François Bouzom
- Non-Clinical Development Department, UCB Biopharma sprl, Braine-l'Alleud, Belgium
| | - Chanteux Hugues
- Non-Clinical Development Department, UCB Biopharma sprl, Braine-l'Alleud, Belgium
| | - Anna-Lena Ungell
- Non-Clinical Development Department, UCB Biopharma sprl, Braine-l'Alleud, Belgium
| |
Collapse
|
29
|
Elder DP, Holm R, Kuentz M. Medicines for Pediatric Patients-Biopharmaceutical, Developmental, and Regulatory Considerations. J Pharm Sci 2016; 106:950-960. [PMID: 28041968 DOI: 10.1016/j.xphs.2016.12.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 01/19/2023]
Abstract
This commentary reflects current developments in pediatric medicine. The underpinning legislation in both Europe and the United States has led to the initiation of an increased number of clinical trials in the pediatric population, but there are still a number of outstanding issues within this field. These include the differences in the physiology between adults and the very heterogeneous nature of pediatric patients. There is an ongoing scientific debate on the applicability of a Pediatric Biopharmaceutical Classification System to define when waivers for bioequivalence studies can be supported by in vitro dissolution. However, a challenge is that in vitro models should adequately mimic the physiology of different pediatric age-groups and dose definition is another critical aspect. There is a tendency for off-label use of established adult medicines, resulting in increased adverse events and decreased efficacy in the target population. Recent advances in physiologically based pharmacokinetic modelling may be used to provide valuable input into these discussions, but there are currently still many knowledge gaps. It is encouraging that there is a global recognition of these deficiencies and substantial funding in the field of basic research is being provided, for example, within Europe the Innovative Medicines Initiative consortium.
Collapse
Affiliation(s)
- David P Elder
- GlaxoSmithKline, Park Road, Ware, Hertfordshire SG12 0DP, UK
| | - René Holm
- Drug Product Development, Jansen Research & Development, Johnson & Johnson, Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Martin Kuentz
- University of Applied Sciences and Arts Northwestern Switzerland, Institute of Pharmaceutical Technology, Muttenz CH-4132, Switzerland.
| |
Collapse
|
30
|
Gadkar K, Kirouac D, Parrott N, Ramanujan S. Quantitative systems pharmacology: a promising approach for translational pharmacology. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:57-65. [PMID: 27978989 DOI: 10.1016/j.ddtec.2016.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022]
Abstract
Biopharmaceutical companies have increasingly been exploring Quantitative Systems Pharmacology (QSP) as a potential avenue to address current challenges in drug development. In this paper, we discuss the application of QSP modeling approaches to address challenges in the translational of preclinical findings to the clinic, a high risk area of drug development. Three cases have been highlighted with QSP models utilized to inform different questions in translational pharmacology. In the first, a mechanism based asthma model is used to evaluate efficacy and inform biomarker strategy for a novel bispecific antibody. In the second case study, a mitogen-activated protein kinase (MAPK) pathway signaling model is used to make translational predictions on clinical response and evaluate novel combination therapies. In the third case study, a physiologically based pharmacokinetic (PBPK) model it used to guide administration of oseltamivir in pediatric patients.
Collapse
Affiliation(s)
- K Gadkar
- Development Sciences, Genentech, United States.
| | - D Kirouac
- Development Sciences, Genentech, United States
| | - N Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - S Ramanujan
- Development Sciences, Genentech, United States
| |
Collapse
|
31
|
Liu T, Ghafoori P, Gobburu JVS. Allometry Is a Reasonable Choice in Pediatric Drug Development. J Clin Pharmacol 2016; 57:469-475. [PMID: 27649629 DOI: 10.1002/jcph.831] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/15/2016] [Indexed: 11/10/2022]
Abstract
Pharmacokinetics (PK) plays a key role in bridging drug efficacy and safety from adults to pediatric patients. The principal purpose of projecting dosing in pediatrics is to guide trial design, not to waive the study per se. This research was designed to evaluate whether the allometric scaling (AS) approach is a satisfactory method to design PK studies in pediatric patients aged 2 years and older. We systematically evaluated drugs that had pediatric label information updated from 1998 to 2015. Only intravenous (IV) or oral administration drugs with available PK information in both children and adults from FDA-approved labels were included. The allometric scaling approach was used to extrapolate adult clearance to pediatric clearance. The relative difference between the observed and the allometric scaling approach-predicted clearance was summarized and used to evaluate the predictive power of the allometric scaling approach. A total of 36 drugs eliminated by a metabolic pathway and 10 drugs by the renal pathway after intravenous (IV) or oral administration were included. Regardless of the administration route, elimination pathway, and age group, the allometric scaling approach can predict clearance in pediatric patients within a 2-fold difference; 18 of the included drugs were predicted within a 25% difference, and 31 drugs within a 50% difference. The allometric scaling approach can adequately design PK studies in pediatric subjects 2 years and older.
Collapse
Affiliation(s)
- Tao Liu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Parima Ghafoori
- Department of Pharmacy Practice and Science, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Jogarao V S Gobburu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| |
Collapse
|