1
|
Ravix A, Gotta V, Pfister M, Berger C, Glauser A, Paioni P, Csajka C, Guidi M. Dose Evaluation and Optimization of Amoxicillin in Children Treated for Lyme Disease. J Clin Pharmacol 2025. [PMID: 39866024 DOI: 10.1002/jcph.6190] [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: 10/08/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025]
Abstract
Amoxicillin is commonly used to treat erythema migrans in the first stage of Lyme disease in children, with a recommended dose of 50 mg/kg/day, administered three times a day (q8h). This model-based simulation study aimed to determine whether splitting the same daily dose into two administrations (q12h) would provide comparable drug exposure. A pharmacokinetic model suitable for a pediatric population (age: 1 month to 18 years, weight: 4-80 kg) was selected through a literature review. Simulations were performed with 15,000 virtual patients receiving 16.67 mg/kg/dose q8h, 25 mg/kg/dose q12h, or other q12h dosing variations. The target therapeutic level was defined by the percentage of time that the unbound drug concentration remained above the minimum inhibitory concentration (% fT > MIC) specific to Borrelia burgdorferi, with MICs of 0.06, 0.25, 1, 2, and 4 mg/L, requiring at least 40% and 50% of time for effective treatment. Probability of target attainment (PTA) was considered acceptable if it exceeded 50%, allowing for comparison of dosing schedules. Results indicated that the 50 mg/kg/day divided q12h regimen provided similar drug exposure to the q8h regimen for MICs below 2 mg/L (PTAs >50%). For a MIC of 2 mg/L, PTA was achieved with a higher dose of 30 mg/kg/dose q12h. However, for a MIC of 4 mg/L, the PTA criterion was not met. These findings suggest that a twice-daily dosing of 25 mg/kg/dose provides comparable bactericidal activity to the thrice-daily regimen for MICs between 0.06 and 1 mg/L. This simplified regimen may improve adherence and treatment implementation in children.
Collapse
Affiliation(s)
- Anne Ravix
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Verena Gotta
- Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
- SwissPedDose/SwissPedNet Collaboration Expert Team, Zurich, Switzerland
| | - Marc Pfister
- Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
- SwissPedDose/SwissPedNet Collaboration Expert Team, Zurich, Switzerland
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
- SwissPedDose, Zürich, Switzerland
| | | | - Paolo Paioni
- SwissPedDose/SwissPedNet Collaboration Expert Team, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Chantal Csajka
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- SwissPedDose/SwissPedNet Collaboration Expert Team, Zurich, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva & Lausanne, Switzerland
| | - Monia Guidi
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva & Lausanne, Switzerland
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Centanni M, Zaher O, Elhad D, Karlsson MO, Friberg LE. Physiologically-based pharmacokinetic models versus allometric scaling for prediction of tyrosine-kinase inhibitor exposure from adults to children. Cancer Chemother Pharmacol 2024; 94:297-310. [PMID: 38782791 PMCID: PMC11390758 DOI: 10.1007/s00280-024-04678-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Model-based methods can predict pediatric exposure and support initial dose selection. The aim of this study was to evaluate the performance of allometric scaling of population pharmacokinetic (popPK) versus physiologically based pharmacokinetic (PBPK) models in predicting the exposure of tyrosine kinase inhibitors (TKIs) for pediatric patients (≥ 2 years), based on adult data. The drugs imatinib, sunitinib and pazopanib were selected as case studies due to their complex PK profiles including high inter-patient variability, active metabolites, time-varying clearances and non-linear absorption. METHODS Pediatric concentration measurements and adult popPK models were derived from the literature. Adult PBPK models were generated in PK-Sim® using available physicochemical properties, calibrated to adult data when needed. PBPK and popPK models for the pediatric populations were translated from the models for adults and were used to simulate concentration-time profiles that were compared to the observed values. RESULTS Ten pediatric datasets were collected from the literature. While both types of models captured the concentration-time profiles of imatinib, its active metabolite, sunitinib and pazopanib, the PBPK models underestimated sunitinib metabolite concentrations. In contrast, allometrically scaled popPK simulations accurately predicted all concentration-time profiles. Trough concentration (Ctrough) predictions from the popPK model fell within a 2-fold range for all compounds, while 3 out of 5 PBPK predictions exceeded this range for the imatinib and sunitinib metabolite concentrations. CONCLUSION Based on the identified case studies it appears that allometric scaling of popPK models is better suited to predict exposure of TKIs in pediatric patients ≥ 2 years. This advantage may be attributed to the stable enzyme expression patterns from 2 years old onwards, which can be easily related to adult levels through allometric scaling. In some instances, both methods performed comparably. Understanding where discrepancies between the model methods arise, can further inform model development and ultimately support pediatric dose selection.
Collapse
Affiliation(s)
- Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Omar Zaher
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - David Elhad
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden.
| |
Collapse
|
3
|
Decker RL, Steven Ernest C, Radtke DB, Wang R, Araújo J, Keller SY, Zhang X. A population pharmacokinetic model using allometric scaling for baricitinib in patients with juvenile idiopathic arthritis. CPT Pharmacometrics Syst Pharmacol 2024; 13:970-981. [PMID: 38532270 PMCID: PMC11179695 DOI: 10.1002/psp4.13131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 03/28/2024] Open
Abstract
Baricitinib is approved for the treatment of rheumatoid arthritis (RA) in more than 70 countries, and juvenile idiopathic arthritis (JIA) in the European Union. Population pharmacokinetic (PK) models were developed in a phase 3 trial to characterize PK in pediatric patients with JIA and identify weight-based dosing regimens. The phase 3, randomized, double-blind, placebo-controlled withdrawal, efficacy and safety trial, JUVE-BASIS, enrolled patients (aged 2 to <18 years) with polyarticular course JIA. During a safety/PK period, baricitinib concentration data from age-based dose cohorts were compared to concentrations from adult patients receiving 4-mg QD. PK data were used to develop a population PK model with allometric scaling to determine a weight-based posology in pediatric patients with JIA that matched the adult 4-mg exposure. Baricitinib plasma concentrations from 217 pediatric patients were used to characterize PK. Based on the adult model, pediatric PK was best described using a 2-compartment model with allometric scaling on clearance and volume of distribution and renal function (estimated with glomerular filtration rate [GFR], a known covariate affecting PK of baricitinib) on clearance. The PK modeling suggested the optimal dosing regimen based on weight for pediatric patients as: 2-mg QD for patients 10 to <30 kg and 4-mg QD for patients ≥30 kg. The use of a population PK model of baricitinib treatment in adult patients with RA, with the addition of allometric scaling for weight on clearance and volume terms, was useful to predict exposures and identify weight-based dosing in pediatric patients with JIA.
Collapse
Affiliation(s)
| | | | | | - Rona Wang
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | | | - Xin Zhang
- Eli Lilly and CompanyIndianapolisIndianaUSA
| |
Collapse
|
4
|
Ye J, Bi Y, Ting N. How to select the initial dose for a pediatric study? J Biopharm Stat 2023; 33:844-858. [PMID: 36476267 DOI: 10.1080/10543406.2022.2149770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
In typical clinical development programs, a new drug is first developed for the adult use. Drugs are often approved for adult use or in the process of obtaining approval in adults in the target indication before pediatric development is initiated. In designing the first pediatric clinical trial, one of the challenges is to select the initial dose to be tested. The ICH E11 R1 guidance advises that chronologic age alone may not always be the most appropriate categorical determinant to define developmental subgroups in pediatric studies. In this manuscript, the approaches to utilize available data in adults related to those factors beyond age to inform the starting dose selection in pediatric drug development are discussed. Practical considerations and approaches are provided for informing pediatric starting dose. Additional considerations to use pre-clinical information are provided in the case when adult information is limited or not available.
Collapse
Affiliation(s)
- Jingjing Ye
- Global Statistics and Data Science (GSDS), Fulton, MD, USA
| | - Youwei Bi
- Division of Pharmacometrics, Office of Translational Sciences (OTS), Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Naitee Ting
- Biostatistics and Data Science, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| |
Collapse
|
5
|
Lim A, Sharma P, Stepanov O, Reddy VP. Application of Modelling and Simulation Approaches to Predict Pharmacokinetics of Therapeutic Monoclonal Antibodies in Pediatric Population. Pharmaceutics 2023; 15:pharmaceutics15051552. [PMID: 37242793 DOI: 10.3390/pharmaceutics15051552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Ethical regulations and limited paediatric participants are key challenges that contribute to a median delay of 6 years in paediatric mAb approval. To overcome these barriers, modelling and simulation methodologies have been adopted to design optimized paediatric clinical studies and reduce patient burden. The classical modelling approach in paediatric pharmacokinetic studies for regulatory submissions is to apply body weight-based or body surface area-based allometric scaling to adult PK parameters derived from a popPK model to inform the paediatric dosing regimen. However, this approach is limited in its ability to account for the rapidly changing physiology in paediatrics, especially in younger infants. To overcome this limitation, PBPK modelling, which accounts for the ontogeny of key physiological processes in paediatrics, is emerging as an alternative modelling strategy. While only a few mAb PBPK models have been published, PBPK modelling shows great promise demonstrating a similar prediction accuracy to popPK modelling in an Infliximab paediatric case study. To facilitate future PBPK studies, this review consolidated comprehensive data on the ontogeny of key physiological processes in paediatric mAb disposition. To conclude, this review discussed different use-cases for pop-PK and PBPK modelling and how they can complement each other to increase confidence in pharmacokinetic predictions.
Collapse
Affiliation(s)
- Andrew Lim
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
- Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Pradeep Sharma
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| | - Oleg Stepanov
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| | - Venkatesh Pilla Reddy
- Clinical Pharmacology and Pharmacometrics, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB2 8PA, UK
| |
Collapse
|
6
|
Chung A, Carroll M, Almeida P, Petrova A, Isaac D, Mould D, Wine E, Huynh H. Early Infliximab Clearance Predicts Remission in Children with Crohn's Disease. Dig Dis Sci 2022; 68:1995-2005. [PMID: 36562887 DOI: 10.1007/s10620-022-07783-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Children with Crohn's disease have lower response rates to infliximab, lower infliximab levels, and higher infliximab clearance on weight-based dosing than adults. We hypothesize infliximab clearance is a predictive of later outcomes on infliximab in children with Crohn's disease. METHODS In this single-center retrospective study, data were collected from charts on diagnosis, anthropometry, routine labs, infliximab therapeutic drug monitoring, infliximab dosing, disease activity, and other treatments. With these data we generated a population pharmacokinetic model using non-linear mixed effects modeling and calculated infliximab clearance for each patient over time. Patients were classified as in remission, responder-only or non-responder at 5, 10 and 16 months. Regression and ROC analyses were used to assess for early predictors of remission and response to infliximab. RESULTS Eighty-five subjects were included, with a median follow-up of 22.3 months (IQR 10.1-36.8). Our pharmacokinetic model showed infliximab clearance was positively associated with CRP and weight, while negatively associated with albumin. In regression analyses, early infliximab clearance was the only significant, consistent predictor of remission. A 0.1 L/day increase in infliximab clearance predicted remission with an OR between 0.179 and 0.426. Differences in dosing did not account for differences in outcome. Infliximab clearance alone had moderate predictive accuracy of remission, with an AUC between 0.682 and 0.738. CONCLUSIONS Early infliximab clearance is strongly associated with remission in children with Crohn's disease. It may be useful as a marker of response in proactive therapeutic drug monitoring to guide early dose optimization and/or changes in treatment for betterment of long-term outcomes.
Collapse
Affiliation(s)
- Aaron Chung
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Matthew Carroll
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Patricia Almeida
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Alexandra Petrova
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Daniela Isaac
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | | | - Eytan Wine
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Hien Huynh
- Division of Pediatric Gastroenterology, Faculty of Medicine, University of Alberta, Edmonton, Canada. .,Division of Pediatric GI Nutrition, Department of Pediatrics, University of Alberta Faculty of Medicine & Dentistry, ECHA 4-579 11405 87 Ave NW, Edmonton, AB, T6G1C9, Canada.
| |
Collapse
|
7
|
Bouazza N, Dokoumetzidis A, Knibbe CAJ, de Wildt SN, Ambery C, De Cock PA, Gasthuys E, Foissac F, Urien S, Hamberg AK, Poggesi I, Zhao W, Vermeulen A, Standing JF, Tréluyer JM. General clinical and methodological considerations on the extrapolation of pharmacokinetics and optimization of study protocols for small molecules and monoclonal antibodies in children. Br J Clin Pharmacol 2022; 88:4985-4996. [PMID: 36256514 DOI: 10.1111/bcp.15571] [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: 10/21/2021] [Revised: 09/05/2022] [Accepted: 09/20/2022] [Indexed: 12/01/2022] Open
Abstract
Pharmacometric modelling plays a key role in both the design and analysis of regulatory trials in paediatric drug development. Studies in adults provide a rich source of data to inform the paediatric investigation plans, including knowledge on drug pharmacokinetics (PK), safety and efficacy. In children, drug disposition differs widely from birth to adolescence but extrapolating adult to paediatric PK, safety and efficacy either with pharmacometric or physiologically based approaches can help design or in some cases reduce the need for clinical studies. Aspects to consider when extrapolating PK include the maturation of drug metabolizing enzyme expression, glomerular filtration, drug excretory systems, and the expression and activity of specific transporters in conjunction with other drug properties such as fraction unbound. Knowledge of these can be used to develop extrapolation tools such as allometric scaling plus maturation functions or physiologically based PK. PK/pharmacodynamic approaches and well-designed clinical trials in children are of key importance in paediatric drug development. In this white paper, state-of-the-art of current methods used for paediatric extrapolation will be discussed. This paper is part of a conect4children implementation of innovative methodologies including pharmacometric and physiologically based PK modelling in clinical trial design/paediatric drug development through dissemination of expertise and expert advice. The suggestions arising from this white paper should define a minimum set of standards in paediatric modelling and contribute to the regulatory science.
Collapse
Affiliation(s)
- Naïm Bouazza
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | | | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Intensive Care and Paediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Claire Ambery
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline plc, London, UK
| | - Pieter A De Cock
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium.,Department of Pharmacy, Ghent University Hospital, Ghent, Belgium.,Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium
| | - Elke Gasthuys
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, University of Ghent, Ghent, Belgium
| | - Frantz Foissac
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Saïk Urien
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Anna-Karin Hamberg
- Department of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
| | - Italo Poggesi
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Beerse, Belgium
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, University of Ghent, Ghent, Belgium
| | - Joseph F Standing
- Infection, Inflammation and Immunology, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Pharmacy, Great Ormond Street Hospital for Children, London, UK
| | - Jean-Marc Tréluyer
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | | |
Collapse
|
8
|
Damoiseaux D, Li W, Martínez-Chávez A, Beijnen JH, Schinkel AH, Huitema ADR, Dorlo TPC. Predictiveness of the Human-CYP3A4-Transgenic Mouse Model (Cyp3aXAV) for Human Drug Exposure of CYP3A4-Metabolized Drugs. Pharmaceuticals (Basel) 2022; 15:ph15070860. [PMID: 35890158 PMCID: PMC9322370 DOI: 10.3390/ph15070860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/23/2022] [Accepted: 07/03/2022] [Indexed: 11/24/2022] Open
Abstract
The extrapolation of drug exposure between species remains a challenging step in drug development, contributing to the low success rate of drug approval. As a consequence, extrapolation of toxicology from animal models to humans to evaluate safe, first-in-human (FIH) doses requires high safety margins. We hypothesized that a human-CYP3A4-expressing transgenic (Cyp3aXAV) mouse is a more predictive model for human drug exposure of CYP3A4-metabolized small-molecule drugs. Population pharmacokinetic models based on wild-type (WT) and Cyp3aXAV mouse pharmacokinetic data of oral lorlatinib, brigatinib, ribociclib and fisogatinib were allometrically scaled and compared to human exposure. Extrapolation of the Cyp3aXAV mouse model closely predicted the observed human exposure for lorlatinib and brigatinib with a 1.1-fold and 1.0-fold difference, respectively, compared to a 2.1-fold and 1.9-fold deviation for WT-based extrapolations of lorlatinib and brigatinib, respectively. For ribociclib, the extrapolated WT mouse model gave better predictions with a 1.0-fold deviation compared to a 0.3-fold deviation for the extrapolated Cyp3aXAV mouse model. Due to the lack of a human population pharmacokinetic model for fisogatinib, only median maximum concentration ratios were calculated, resulting in ratios of 1.0 and 0.6 for WT and Cyp3aXAV mice extrapolations, respectively. The more accurate predictions of human exposure in preclinical research based on the Cyp3aXAV mouse model can ultimately result in FIH doses associated with improved safety and efficacy and in higher success rates in drug development.
Collapse
Affiliation(s)
- David Damoiseaux
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (D.D.); (J.H.B.); (A.D.R.H.)
| | - Wenlong Li
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (W.L.); (A.M.-C.); (A.H.S.)
| | - Alejandra Martínez-Chávez
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (W.L.); (A.M.-C.); (A.H.S.)
| | - Jos H. Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (D.D.); (J.H.B.); (A.D.R.H.)
- Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Alfred H. Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (W.L.); (A.M.-C.); (A.H.S.)
| | - Alwin D. R. Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (D.D.); (J.H.B.); (A.D.R.H.)
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Thomas P. C. Dorlo
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (D.D.); (J.H.B.); (A.D.R.H.)
- Correspondence:
| |
Collapse
|
9
|
Hu TM. A General Biphasic Bodyweight Model for Scaling Basal Metabolic Rate, Glomerular Filtration Rate, and Drug Clearance from Birth to Adulthood. AAPS J 2022; 24:67. [PMID: 35538161 DOI: 10.1208/s12248-022-00716-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
The objective of this study is to propose a unified, continuous, and bodyweight-only equation to quantify the changes of human basal metabolic rate (BMR), glomerular filtration rate (GFR), and drug clearance (CL) from infancy to adulthood. The BMR datasets were retrieved from a comprehensive historical database of male and female subjects (0.02 to 64 years). The CL datasets for 17 drugs and the GFR dataset were generated from published maturation and growth models with reported parameter values. A statistical approach was used to simulate the model-generated CL and GFR data for a hypothetical population with 26 age groups (from 0 to 20 years). A biphasic equation with two power-law functions of bodyweight was proposed and evaluated as a general model using nonlinear regression and dimensionless analysis. All datasets universally reveal biphasic curves with two distinct linear segments on log-log plots. The biphasic equation consists of two reciprocal allometric terms that asymptotically determine the overall curvature. The fitting results show a superlinear scaling phase (asymptotic exponent >1; ca. 1.5-3.5) and a sublinear scaling phase (asymptotic exponent <1; ca. 0.5-0.7), which are separated at the phase transition bodyweight ranging from 5 to 20 kg with a mean value of 10 kg (corresponding to 1 year of age). The dimensionless analysis generalizes and offers quantitative realization of the maturation and growth process. In conclusion, the proposed mixed-allometry equation is a generic model that quantitatively describes the phase transition in the human maturation process of diverse human functions.
Collapse
Affiliation(s)
- Teh-Min Hu
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
10
|
Lu J, Deng K, Zhang X, Liu G, Guan Y. Neural-ODE for pharmacokinetics modeling and its advantage to alternative machine learning models in predicting new dosing regimens. iScience 2021; 24:102804. [PMID: 34308294 PMCID: PMC8283337 DOI: 10.1016/j.isci.2021.102804] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/07/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022] Open
Abstract
Forecasting pharmacokinetics (PK) for individual patients is a fundamental problem in clinical pharmacology. One key challenge is that PK models constructed using data from one dosing regimen must predict PK data for different dosing regimen(s). We propose a deep learning approach based on neural ordinary differential equations (neural-ODE) and tested its generalizability against a variety of alternative models. Specifically, we used the PK data from two different treatment regimens of trastuzumab emtansine. The models performed similarly when the training and the test sets come from the same dosing regimen. However, for predicting a new treatment regimen, the neural-ODE model showed substantially better performance. To date, neural-ODE is the most accurate PK model in predicting untested treatment regimens. This study represents the first time neural-ODE has been applied to PK modeling and the results suggest it is a widely applicable algorithm with the potential to impact future studies.
Collapse
Affiliation(s)
- James Lu
- Modeling & Simulation/Clinical Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kaiwen Deng
- Ann Arbor Algorithms Inc, 3001 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Xinyuan Zhang
- Ann Arbor Algorithms Inc, 3001 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Gengbo Liu
- Modeling & Simulation/Clinical Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Nguyen D, Shaik JS, Tai G, Tiffany C, Perry C, Dumont E, Gardiner D, Barth A, Singh R, Hossain M. Comparison between physiologically based pharmacokinetic and population pharmacokinetic modelling to select paediatric doses of gepotidacin in plague. Br J Clin Pharmacol 2021; 88:416-428. [PMID: 34289143 PMCID: PMC9293063 DOI: 10.1111/bcp.14996] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/09/2021] [Indexed: 12/31/2022] Open
Abstract
Aims To develop physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PopPK) models to predict effective doses of gepotidacin in paediatrics for the treatment of pneumonic plague (Yersinia pestis). Methods A gepotidacin PBPK model was constructed using a population‐based absorption, distribution, metabolism and excretion simulator, Simcyp®, with physicochemical and in vitro data, optimized with clinical data from a dose‐escalation intravenous (IV) study and a human mass balance study. A PopPK model was developed with pooled PK data from phase 1 studies with IV gepotidacin in healthy adults. Results For both the PopPK and PBPK models, body weight was found to be a key covariate affecting gepotidacin clearance. With PBPK, ~90% of the predicted PK for paediatrics fell between the 5th and 95th percentiles of adult values except for subjects weighing ≤5 kg. PopPK‐simulated paediatric means for Cmax and AUC(0‐τ) were similar to adult exposures across various weight brackets. The proposed dosing regimens were weight‐based for subjects ≤40 kg and fixed‐dose for subjects >40 kg. Comparison of observed and predicted exposures in adults indicated that both PBPK and PopPK models achieved similar AUC and Cmax for a given dose, but the Cmax predictions with PopPK were slightly higher than with PBPK. The two models differed on dose predictions in children <3 months old. The PopPK model may be suboptimal for low age groups due to the absence of maturation characterization of drug‐metabolizing enzymes involved with clearance in adults. Conclusions Both PBPK and PopPK approaches can reasonably predict gepotidacin exposures in children.
Collapse
Affiliation(s)
- Dung Nguyen
- GlaxoSmithKline, Collegeville, PA, United States
| | | | - Guoying Tai
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | | | | | | - Aline Barth
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | |
Collapse
|
12
|
Mahmood I, Tegenge MA. Spreadsheet-Based Minimal Physiological Models for the Prediction of Clearance of Therapeutic Proteins in Pediatric Patients. J Clin Pharmacol 2021; 61 Suppl 1:S108-S116. [PMID: 34185903 DOI: 10.1002/jcph.1846] [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: 12/06/2020] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
There is a growing interest in the use of physiologically based pharmacokinetic (PBPK) models as clinical pharmacology drug development tools. In PBPK modeling, not every organ or physiological parameter is required, leading to the development of a minimal PBPK (mPBPK) model, which is simple and efficient. The objective of this study was to streamline mPBPK modeling approaches and enable straightforward prediction of clearance of protein-based products in children. Four mPBPK models for scaling clearance from adult to children were developed and evaluated on Excel spreadsheets using (1) liver and kidneys; (2) liver, kidneys, and skin; (3) liver, kidneys, skin, and lymph; and (4) interstitial, lymph, and plasma volume. There were 35 therapeutic proteins with a total of 113 observations across different age groups (premature neonates to adolescents). For monoclonal and polyclonal antibodies, more than 90% of observations were within a 0.5- to 2-fold prediction error for all 4 methods. For nonantibodies, 79% to 100% of observations were within the 0.5- to 2-fold prediction error for the 4 different methods. Methods 1 and 4 provided the best results, >90% of the total observations were within the 0.5- to 2-fold prediction error for all 3 classes of protein-based products across a wide age range. The precision of clearance prediction was comparatively lower in children ≤2 years of age vs older children (>2 years of age) with methods 1 and 4 predicting 80% to 100% and 75% to 90% of observations within the 0.5- to 2-fold prediction error, respectively. The results of the study indicated that mPBPK models can be developed on spreadsheets, with acceptable performance for prediction of clearance.
Collapse
Affiliation(s)
- Iftekhar Mahmood
- Mahmood Clinical Pharmacology Consultancy, Rockville, Maryland, USA
| | - Million A Tegenge
- Division of Clinical Evaluation and Pharmacology/Toxicology, Center for Biologics Evaluation and Research (CBER), Office of Tissues and Advanced Therapies (OTAT), Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| |
Collapse
|
13
|
Vinks AA, Barrett JS. Model-Informed Pediatric Drug Development: Application of Pharmacometrics to Define the Right Dose for Children. J Clin Pharmacol 2021; 61 Suppl 1:S52-S59. [PMID: 34185897 DOI: 10.1002/jcph.1841] [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: 12/29/2020] [Accepted: 02/16/2021] [Indexed: 12/26/2022]
Abstract
One of the biggest challenges in pediatric drug development is defining a safe and effective dose in pediatric populations, which span across a wide age and development range from neonates to adolescents. Model-informed drug development approaches are particularly suited to address knowledge gaps including data leveraging to increase the success of pediatric studies. Considering the often limited number of patients available for study and logistic difficulties to collect the necessary data in pediatric populations, the application of pharmacometrics and modeling and simulation techniques can improve clinical trial efficiency, increase the probability of regulatory success, and optimize therapeutic individualization in support of dedicated trials. This review describes the state of pediatric model-informed drug development to define the right dose for children and provides suggestions for future development.
Collapse
Affiliation(s)
- Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jeffrey S Barrett
- Quantitative Medicine, Critical Path Institute, Tucson, Arizona, USA
| |
Collapse
|
14
|
Bi Y, Liu J, Li F, Yu J, Bhattaram A, Bewernitz M, Li RJ, Ahn J, Earp J, Ma L, Zhuang L, Yang Y, Zhang X, Zhu H, Wang Y. Model-Informed Drug Development in Pediatric Dose Selection. J Clin Pharmacol 2021; 61 Suppl 1:S60-S69. [PMID: 34185906 DOI: 10.1002/jcph.1848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/18/2021] [Indexed: 01/12/2023]
Abstract
Model-informed drug development (MIDD) has been a powerful and efficient tool applied widely in pediatric drug development due to its ability to integrate and leverage existing knowledge from different sources to narrow knowledge gaps. The dose selection is the most common MIDD application in regulatory submission related to pediatric drug development. This article aims to give an overview of the 3 broad categories of use of MIDD in pediatric dose selection: leveraging from adults to pediatric patients, leveraging from animals to pediatric patients, and integrating mechanism in infants and neonates. Population pharmacokinetic analyses with allometric scaling can reasonably predict the clearance in pediatric patients aged >5 years. A mechanistic-based approach, such as physiologically based pharmacokinetic accounting for ontogeny, or an allometric model with age-dependent exponent, can be applied to select the dose in pediatric patients aged ≤2 years. The exposure-response relationship from adults or from other drugs in the same class may be useful in aiding the pediatric dose selection and benefit-risk assessment. Increasing application and understanding of use of MIDD have contributed greatly to several policy developments in the pediatric field. With the increasing efforts of MIDD under the Prescription Drug User Fee Act VI, bigger impacts of MIDD approaches in pediatric dose selection can be expected. Due to the complexity of model-based analyses, early engagement between drug developers and regulatory agencies to discuss MIDD issues is highly encouraged, as it is expected to increase the efficiency and reduce the uncertainty.
Collapse
Affiliation(s)
- Youwei Bi
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Fang Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jingyu Yu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Atul Bhattaram
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael Bewernitz
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ruo-Jing Li
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jihye Ahn
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Justin Earp
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lian Ma
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Luning Zhuang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yuching Yang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
15
|
Malik PRV, Temrikar ZH, Chelle P, Edginton AN, Meibohm B. Pediatric Dose Selection for Therapeutic Proteins. J Clin Pharmacol 2021; 61 Suppl 1:S193-S206. [PMID: 34185910 DOI: 10.1002/jcph.1829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
In selecting optimal dosing regimens in support of the clinical use of monoclonal antibodies and other therapeutic proteins in pediatric indications, the unique pharmacokinetic properties of this class of biologics, as well as the underlying physiologic and pathophysiologic processes and their modulation by childhood growth and development, needs to be appreciated. During drug development, first-in-pediatric dose selection is a capstone event in the pediatric investigation plan that relies heavily on extrapolation of pharmacokinetic and pharmacodynamic data from adult to pediatric populations. It is facilitated by combinations of pharmacometric approaches, including allometry, physiologically based pharmacokinetic modeling, and population pharmacokinetic analyses, although data on reliability and qualification of some of these tools in the context of therapeutic proteins are still limited but emerging. Presented data suggest nonlinear relationships between body weight and both clearance and volume of distribution for therapeutic proteins in pediatric populations, with allometric exponents of 0.75 and 0.8, respectively. For newborns and infants (<1 year), even higher nonlinearity seems to occur. Translation of the quantitative characterization of the pediatric pharmacokinetics of therapeutic proteins into dosing regimens for the drug label requires compromising between precision dosing and clinical practicability, with tiered dosing algorithms based on size or age strata being the currently most frequently applied methodology.
Collapse
Affiliation(s)
- Paul R V Malik
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Zaid H Temrikar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Pierre Chelle
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
16
|
Cui C, Valerie Sia JE, Tu S, Li X, Dong Z, Yu Z, Yao X, Hatley O, Li H, Liu D. Development of a physiologically based pharmacokinetic (PBPK) population model for Chinese elderly subjects. Br J Clin Pharmacol 2021; 87:2711-2722. [PMID: 33068053 PMCID: PMC8359847 DOI: 10.1111/bcp.14609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/31/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Aims This study aims to develop and verify a physiologically based pharmacokinetic (PBPK) population model for the Chinese geriatric population in Simcyp. Methods Firstly, physiological information for the Chinese geriatric population was collected and later employed to develop the Chinese geriatric population model by recalibration of corresponding physiological parameters in the Chinese adult population model available in Simcyp (i.e., Chinese healthy volunteer model). Secondly, drug‐dependent parameters were collected for six drugs with different elimination pathways (i.e., metabolized by CYP1A2, CYP3A4 or renal excretion). The drug models were then developed and verified by clinical data from Chinese adults, Caucasian adults and Caucasian elderly subjects to ensure that drug‐dependent parameters are correctly inputted. Finally, the tested drug models in combination with the newly developed Chinese geriatric population model were applied to simulate drug concentration in Chinese elderly subjects. The predicted results were then compared with the observations to evaluate model prediction performance. Results Ninety‐eight per cent of predicted AUC, 95% of predicted Cmax, and 100% of predicted CL values were within two‐fold of the observed values, indicating all drug models were properly developed. The drug models, combined with the newly developed population model, were then used to predict pharmacokinetics in Chinese elderly subjects aged 60–93. The predicted AUC, Cmax, and CL values were all within two‐fold of the observed values. Conclusion The population model for the Chinese elderly subjects appears to adequately predict the concentration of the drug that was metabolized by CYP1A2, CYP3A4 or eliminated by renal clearance.
Collapse
Affiliation(s)
- Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China
| | - Jie En Valerie Sia
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siqi Tu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.,School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Xiaobei Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.,School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Zhongqi Dong
- Janssen China R&D Center, Shanghai, 200233, China
| | - Zhiheng Yu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China
| | - Xueting Yao
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China
| | - Oliver Hatley
- Certara UK Ltd, Simcyp Division, Sheffield, S1 2BJ, UK
| | - Haiyan Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
17
|
Yao X, Liu X, Tu S, Li X, Lei Z, Hou Z, Yu Z, Cui C, Dong Z, Salem F, Li H, Liu D. Development of a Virtual Chinese Pediatric Population Physiological Model Targeting Specific Metabolism and Kidney Elimination Pathways. Front Pharmacol 2021; 12:648697. [PMID: 34045960 PMCID: PMC8145459 DOI: 10.3389/fphar.2021.648697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Physiologically based pharmacokinetic (PBPK) modeling and simulating may be a powerful tool in predicting drug behaviors in specific populations. It is a mathematical model that relates the pharmacokinetic (PK) profile of a compound with human anatomical characteristics, physiological characteristics, and biochemical parameters. Predictions using PBPK models offer a promising way to guide drug development and can be used to optimize clinical dosing regimens. However, PK data of new drugs in the pediatric population are too limited to guide clinical therapy, which may lead to frequent adverse events or insufficient efficacy for pediatric patients, particularly in neonates and infants. Objective: The objective of this study was to establish a virtual Chinese pediatric population based on the physiological parameters of Chinese children that could be utilized in PBPK models. Methods: A Chinese pediatric PBPK model was developed in Simcyp Simulator by collecting published Chinese pediatric physiological and anthropometric data to use as system parameters. This pediatric population model was then evaluated in the Chinese pediatric population by predicting the pharmacokinetic characteristics of four probe drugs: theophylline (major CYP1A2 substrate), fentanyl (major CYP3A4 substrate), vancomycin, and ceftazidime (renal-eliminated). Results: The predicted maximum concentration (Cmax), area under the curve of concentration-time (AUC), and clearance (CL) for theophylline (CYP1A2 metabolism pathway) and fentanyl (CYP3A4 metabolism pathway) were within two folds of the observed data. For drugs mainly eliminated by renal clearance (vancomycin and ceftazidime) in the Chinese pediatric population, the ratio of prediction to observation for major PK parameters was within a 2-fold error range. Conclusion: The model is a supplement to the previous Chinese population PBPK model. We anticipate the model to be a better representative of the pediatric Chinese population for drugs PK, offering greater clinical precision for medication given to the pediatric population, ultimately advancing clinical development of pediatric drugs. We can refine this model further by collecting more physiological parameters of Chinese children.
Collapse
Affiliation(s)
- Xueting Yao
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Xuanlin Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Siqi Tu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing, China
| | - Xiaobei Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing, China
| | - Zihan Lei
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhe Hou
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhiheng Yu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | | | - Farzaneh Salem
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom
| | - Haiyan Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China.,Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
18
|
Germovsek E, Cheng M, Giragossian C. Allometric scaling of therapeutic monoclonal antibodies in preclinical and clinical settings. MAbs 2021; 13:1964935. [PMID: 34530672 PMCID: PMC8463036 DOI: 10.1080/19420862.2021.1964935] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/19/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Constant technological advancement enabled the production of therapeutic monoclonal antibodies (mAbs) and will continue to contribute to their rapid expansion. Compared to small-molecule drugs, mAbs have favorable characteristics, but also more complex pharmacokinetics (PK), e.g., target-mediated nonlinear elimination and recycling by neonatal Fc-receptor. This review briefly discusses mAb biology, similarities and differences in PK processes across species and within human, and provides a detailed overview of allometric scaling approaches for translating mAb PK from preclinical species to human and extrapolating from adults to children. The approaches described here will remain vital in mAb drug development, although more data are needed, for example, from very young patients and mAbs with nonlinear PK, to allow for more confident conclusions and contribute to further growth of this field. Improving mAb PK predictions will facilitate better planning of (pediatric) clinical studies and enable progression toward the ultimate goal of expediting drug development.
Collapse
Affiliation(s)
- Eva Germovsek
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Ming Cheng
- Development Biologicals, Drug Metabolism And Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| | - Craig Giragossian
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| |
Collapse
|
19
|
Prediction of Clearance in Children from Adults Following Drug-Drug Interaction Studies: Application of Age-Dependent Exponent Model. Drugs R D 2020; 20:47-54. [PMID: 32056156 PMCID: PMC7067713 DOI: 10.1007/s40268-020-00295-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background and Objective Pharmacokinetic drug–drug interaction (DDI) studies are conducted in adult subjects during drug development but there are limited studies that have characterized pharmacokinetic DDI studies in children. The objective of this study was to evaluate if the DDI clearance values from adults can be allometrically extrapolated from adults to children. Methods Fifteen drugs were included in this study and the age of the children ranged from premature neonates to adolescents (30 observations across the age groups). The age-dependent exponent (ADE) model was used to predict the clearance of drugs in children from adults following DDI studies. Results The prediction error of drug clearances following DDIs in children ranged from 4 to 67%. Of 30 observations, 17 (57%) and 27 (90%) observations had a prediction error ≤ 30% and ≤ 50%, respectively. Conclusion This study indicates that it is possible to predict the clearance of drugs with reasonable accuracy in children from adults following DDI studies using an ADE model. The method is simple, robust, and reliable and can replace other complex empirical models.
Collapse
|
20
|
Extrapolation of Drug Clearance in Children ≤ 2 Years of Age from Empirical Models Using Data from Children (> 2 Years) and Adults. Drugs R D 2020; 20:1-10. [PMID: 31820365 PMCID: PMC7067721 DOI: 10.1007/s40268-019-00291-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The application of modeling and simulation approaches in clinical pharmacology studies has gained momentum over the last 20 years. OBJECTIVES The objective of this study was to develop six empirical models from clearance data obtained from children aged > 2 years and adults to evaluate the suitability of the models to predict drug clearance in children aged ≤ 2 years (preterm, term, and infants). METHODS Ten drugs were included in this study and administered intravenously: alfentanil, amikacin, busulfan, cefetamet, meperidine, oxycodone, propofol, sufentanil, theophylline, and tobramycin. These drugs were selected according to the availability of individual subjects' weight, age, and clearance data (concentration-time data for these drugs were not available to the author). The chosen drugs are eliminated by extensive metabolism by either the renal route or both the renal and hepatic routes. The six empirical models were (1) age and body weight-dependent sigmoidal maximum possible effect (Emax) maturation model, (2) body weight-dependent sigmoidal Emax model, (3) uridine 5'-diphospho [body weight-dependent allometric exponent model (BDE)], (4) age-dependent allometric exponent model (ADE), (5) a semi-physiological model, and (6) an allometric model developed from children aged > 2 years to adults. The model-predicted clearance values were compared with observed clearance values in an individual child. In this analysis, a prediction error of ≤ 50% for mean or individual clearance values was considered acceptable. RESULTS Across all age groups and the ten drugs, data for 282 children were compared between observed and model-predicted clearance values. The validation data consisted of 33 observations (sum of different age groups for ten drugs). Only three of the six models (body weight-dependent sigmoidal Emax model, ADE, and semi-physiological model) provided reasonably accurate predictions of clearance (> 80% observation with ≤ 50% prediction error) in children aged ≤ 2 years. In most instances, individual predicted clearance values were erratic (as indicated by % error) and were not in agreement with the observed clearance values. CONCLUSIONS The study indicated that simple empirical models can provide more accurate results than complex empirical models.
Collapse
|
21
|
Xu Y, Langevin BA, Zhou H, Xu Z. Model‐Aided Adults‐to‐Children Pharmacokinetic Extrapolation and Empirical Body Size‐Based Dosing Exploration for Therapeutic Monoclonal Antibodies—Is Allometry a Reasonable Choice? J Clin Pharmacol 2020; 60:1573-1584. [DOI: 10.1002/jcph.1677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Yan Xu
- Clinical Pharmacology and Pharmacometrics Janssen Research & Development, LLC Spring House Pennsylvania USA
| | - Brooke A. Langevin
- Clinical Pharmacology and Pharmacometrics Janssen Research & Development, LLC Spring House Pennsylvania USA
- Chemical & Biomolecular Engineering Johns Hopkins University Baltimore Maryland USA
| | - Honghui Zhou
- Clinical Pharmacology and Pharmacometrics Janssen Research & Development, LLC Spring House Pennsylvania USA
| | - Zhenhua Xu
- Clinical Pharmacology and Pharmacometrics Janssen Research & Development, LLC Spring House Pennsylvania USA
| |
Collapse
|
22
|
Wu Q, Taboureau O, Audouze K. Development of an adverse drug event network to predict drug toxicity. Curr Res Toxicol 2020; 1:48-55. [PMID: 34345836 PMCID: PMC8320634 DOI: 10.1016/j.crtox.2020.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 11/28/2022] Open
Abstract
Despite of their therapeutic effects, drug's exposure may have negative effects on human health such as adverse drug reaction (ADR) and side effects (SE). Adverse drug events (ADEs), that correspond to an event occurring during the drug treatment (i.e. ADR and SE), is not necessarily caused by the drug itself, as this is the case with medical errors and social factors. Due to the complexity of the biological systems, not all ADEs are known for marketed drugs. Therefore, new and effective methods are needed to determine potential risks, including the development of computational strategies. We present an ADE association network based on 90,827 drug-ADE associations between 930 unique drug and 6221 unique ADE, on which we implemented a scoring system based on a pull-down approach for prediction of drug-ADE combination. Based on our network, ADEs proposed for three drugs, safinamide, sonidegib, rufinamide are further discussed. The model was able to identify, already known drug-ADE associations that are supported by the literature and FDA reports, and also to predict uncharacterized associations such as dopamine dysregulation syndrome, or nicotinic acid deficiency for the drugs safinamide and sonidegib respectively, illustrating the power of such integrative toxicological approach.
Collapse
Key Words
- ADE, adverse drug event
- ADR, adverse drug reaction
- AOP, adverse outcome pathway
- Adverse event network
- Computational toxicology
- FAERS, FDA Adverse Event Reporting System
- FDA, Food and Drug Administration
- HMS-PCI, high-throughput mass spectrometric protein complex identification
- LRT, Likelihood Ratio Test
- MedDRA, Medical Dictionary for Regulatory Activities
- Network science
- PPAN, protein-protein association network
- PT, Preferred Term
- Predictive toxicity
- QSAR, Quantitative structure-activity relationships
- SE, side effect
- SOC, System Organ Class
- System toxicology
- TAP–MS, tandem-affinity-purification method coupled to mass spectrometry
- pullS, pull-down score
- wS, weighted score
Collapse
Affiliation(s)
- Qier Wu
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| | - Olivier Taboureau
- Université de Paris, BFA, CNRS UMR 8251, ERL Inserm U1133, CNRS UMR 8251, F-75013 Paris, France
| | - Karine Audouze
- Université de Paris, T3S, Inserm UMR S-1124, F-75006 Paris, France
| |
Collapse
|
23
|
Ye PP, Zheng Y, Du B, Liu XT, Tang BH, Kan M, Zhou Y, Hao GX, Huang X, Su LQ, Wang WQ, Yu F, Zhao W. First dose in neonates: pharmacokinetic bridging study from juvenile mice to neonates for drugs metabolized by CYP3A. Xenobiotica 2020; 50:1275-1284. [PMID: 32400275 DOI: 10.1080/00498254.2020.1768454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
First dose prediction is challenging in neonates. Our objective in this proof-of-concept study was to perform a pharmacokinetic (PK) bridging study from juvenile mice to neonates for drugs metabolized by CYP3A. We selected midazolam and clindamycin as model drugs. We developed juvenile mice population PK models using NONMEM. The PK parameters of these two drugs in juvenile mice were used to bridge PK parameters in neonates using different correction methods. The bridging results were evaluated by the fold-error of 0.5- to 1.5-fold. Simple allometry with and without a correction factor for maximum lifespan potential could be used for a bridging of clearance (CL) and volume of distribution (Vd), respectively, from juvenile mice to neonates. Simulation results demonstrated that for midazolam, 100% of clinical studies for which both the predictive CL and Vd were within 0.5- to 1.5-fold of the observed. For clindamycin, 75% and 100% of clinical studies for which the predictive CL and Vd were within 0.5- to 1.5-fold of the observed. A PK bridging of drugs metabolized by CYP3A is feasible from juvenile mice to neonates. It could be a complement to the ADE and PBPK models to support the first dose in neonates.
Collapse
Affiliation(s)
- Pan-Pan Ye
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Du
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xi-Ting Liu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Kan
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Le-Qun Su
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wen-Qi Wang
- Clinical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Feng Yu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Clinical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Department of Pediatrics, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
24
|
Malik PRV, Edginton AN. Integration of Ontogeny Into a Physiologically Based Pharmacokinetic Model for Monoclonal Antibodies in Premature Infants. J Clin Pharmacol 2019; 60:466-476. [DOI: 10.1002/jcph.1540] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Paul R. V. Malik
- School of PharmacyUniversity of Waterloo Kitchener Ontario Canada
| | | |
Collapse
|
25
|
Malik PRV, Edginton AN. Physiologically-Based Pharmacokinetic Modeling vs. Allometric Scaling for the Prediction of Infliximab Pharmacokinetics in Pediatric Patients. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:835-844. [PMID: 31343836 PMCID: PMC6875711 DOI: 10.1002/psp4.12456] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
The comparative performances of physiologically‐based pharmacokinetic (PBPK) modeling and allometric scaling for predicting the pharmacokinetics (PKs) of large molecules in pediatrics are unknown. Therefore, both methods were evaluated for accuracy in translating knowledge of infliximab PKs from adults to children. PBPK modeling was performed using the base model for large molecules in PK‐Sim version 7.4 with modifications in Mobi. Eight population PK models from literature were reconstructed and scaled by allometry to pediatrics. Evaluation data included seven pediatric studies (~4–18 years). Both methods performed comparably with 66.7% and 68.6% of model‐predicted concentrations falling within twofold of the observed concentrations for PBPK modeling and allometry, respectively. Considerable variability was noted among the allometric models. Therefore, pediatric clinical trial planning would benefit from using approaches that require predictions depending on the specific question i.e., PBPK modeling and allometry.
Collapse
Affiliation(s)
- Paul R V Malik
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Andrea N Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| |
Collapse
|