1
|
Helfer VE, Gonzalez D, Kyler KE, Dunlap TC, Williams V, Radford J, Selvarangan R, Sasidharan A, Chan S, Artz N, Retke B, Toren P, Gibson K, Shakhnovich V. Exploring the Influence of Obesity and CYP2 C19 Genotype on Lansoprazole Pharmacokinetics in Pediatric Patients: A Population Modeling Approach. Clin Pharmacokinet 2025:10.1007/s40262-025-01517-0. [PMID: 40379961 DOI: 10.1007/s40262-025-01517-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND AND OBJECTIVE Pediatric pharmacokinetics pose unique challenges, particularly concerning drug dosing in the presence of obesity and genetic variability in drug-metabolizing enzymes such as cytochrome P450 (CYP) 2C19. This study aimed to develop a lansoprazole pediatric population pharmacokinetic (popPK) model considering obesity, obesity-associated changes in inflammatory cytokines and the liver, and CYP2C19 genotype, and to assess implications for dosing strategies. METHODS Pediatric subjects with and without obesity (6-21 years, n = 47) received one oral dose of 1.2 mg/kg fat-free mass (FFM), not exceeding 60 mg. Plasma concentrations were measured at multiple time points, and a popPK analysis using NONMEM with stepwise covariate modeling was performed. Simulations compared exposures between children without and with obesity (BMI percentile ≥ 95th) after two dosing strategies: U.S. Food and Drug Administration (FDA)-approved total body weight-tiered and FFM-based dosing. RESULTS A total of 537 lansoprazole concentrations were modeled using a two-compartment model with Weibull absorption and linear elimination. Parameters were allometrically scaled to FFM with fixed exponents of 0.75 for clearances and 1 for volumes. CYP2C19 was identified as a significant covariate for CL/F. No significant differences in lansoprazole exposure were observed between children with and without obesity, with both dosing approaches. Higher exposure was noted in poor/intermediate metabolizers of CYP2C19. FFM-based dosing led to similar levels of exposure between children (aged 6-11 years) and adolescents (aged 12-17 years). CONCLUSIONS Obesity was not associated with differences in lansoprazole pharmacokinetics in children. A FFM-based dosing approach could result in comparable exposure between children and adolescents. Dose adjustments are supported for poor/intermediate metabolizers of CYP2C19.
Collapse
Affiliation(s)
- Victória E Helfer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Daniel Gonzalez
- Division of Clinical Pharmacology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Kathryn E Kyler
- Children's Mercy Kansas City, Kansas City, MO, USA.
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.
| | - Tyler C Dunlap
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | | | | | - Rangaraj Selvarangan
- Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Sherwin Chan
- Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Nathan Artz
- Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Paul Toren
- Children's Mercy Kansas City, Kansas City, MO, USA
| | - Kim Gibson
- Children's Mercy Kansas City, Kansas City, MO, USA
| | | |
Collapse
|
2
|
Tang F, Ng CM, Horn J, Bada HS, Leggas M. Pharmacokinetic Modeling and Model-Based Hypothesis Generation for Dose Optimization of Clonidine in Neonates With Neonatal Opioid Withdrawal Syndrome. Clin Pharmacol Ther 2025; 117:1254-1263. [PMID: 39575611 PMCID: PMC11993295 DOI: 10.1002/cpt.3507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/10/2024] [Indexed: 04/14/2025]
Abstract
The No-POPPY study (NCT03396588), a double-blind, randomized trial compared morphine with clonidine therapy for neonatal opioid withdrawal syndrome (NOWS) and found that the duration of treatment was similar across groups. This is significant because perinatal use of morphine has the potential for neurodevelopmental consequences. Still, the clonidine group reached symptom stabilization (Finnegan score (FS) < 8) later than the morphine group and had a more significant number of patients who required adjunct therapy. However, the mean FS was consistently lower in the clonidine group after day 6. This prompted us to use pharmacokinetic (PK) and parametric time-to-event (TTE) modeling to simulate dosage schedules that may decrease the time to stabilization and reduce the need for adjunct therapy. Population PK (popPK) analysis was conducted, and the final model was a one-compartment model with first-order absorption and elimination, incorporating allometric scaling and age effect on apparent clearance (CL/F) and apparent volume (V/F). The population estimates for CL/F and V/F were 13.6 L/h/70 kg and 416 L/70 kg, respectively, similar to the reported values. A Weibull model described the TTE data best, followed by incorporating predicted average concentrations to yield the final Weibull accelerated failure time model. Simulations of dosing strategies showed that increasing both the starting and maximum doses could potentially shorten the time to stabilization, and thus, length of treatment and hospital stay. Given the hypothesis-generating nature of this analysis, the recommended dosing regimens should be tested prospectively to evaluate their benefits.
Collapse
Affiliation(s)
- Fei Tang
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | - Chee M. Ng
- NewGround Pharmaceutical Consulting LLCFoster CityCaliforniaUSA
| | - Jamie Horn
- St. Jude Children's Research HospitalMemphisTennesseeUSA
| | | | - Markos Leggas
- St. Jude Children's Research HospitalMemphisTennesseeUSA
| |
Collapse
|
3
|
Berezowska M, Hayden IS, Brandon AM, Zats A, Patel M, Barnett S, Ogungbenro K, Veal GJ, Taylor A, Suthar J. Recommended approaches for integration of population pharmacokinetic modelling with precision dosing in clinical practice. Br J Clin Pharmacol 2025; 91:1064-1079. [PMID: 39568428 PMCID: PMC11992666 DOI: 10.1111/bcp.16335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/22/2024] Open
Abstract
Current methods of dose determination have contributed to suboptimal and inequitable health outcomes in underrepresented patient populations. The persistent demand to individualise patient treatment, alongside increasing technological feasibility, is leading to a growing adoption of model-informed precision dosing (MIPD) at point of care. Population pharmacokinetic (popPK) modelling is a technique that supports treatment personalisation by characterising drug exposure in diverse patient groups. This publication addresses this important shift in clinical approach, by collating and summarising recommendations from literature. It seeks to provide standardised guidelines on best practices for the development of popPK models and their use in MIPD software tools, ensuring the safeguarding and optimisation of patient outcomes. Moreover, it consolidates guidance from key regulatory and advisory bodies on MIPD software deployment, as well as technical requirements for electronic health record integration. It also considers the future application and clinical impact of machine learning algorithms in popPK and MIPD. Ultimately, this publication aims to facilitate the incorporation of high-quality precision-dosing solutions into standard clinical workflows, thereby enhancing the effectiveness of individualised dose selection at point of care.
Collapse
Affiliation(s)
- Monika Berezowska
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| | - Isaac S. Hayden
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| | - Andrew M. Brandon
- Translational and Clinical Research InstituteNewcastle University Centre for CancerNewcastle upon TyneUK
| | - Arsenii Zats
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| | - Mehzabin Patel
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| | - Shelby Barnett
- Translational and Clinical Research InstituteNewcastle University Centre for CancerNewcastle upon TyneUK
| | - Kayode Ogungbenro
- Division of Pharmacy & Optometry, School of Health SciencesUniversity of ManchesterManchesterUK
| | - Gareth J. Veal
- Translational and Clinical Research InstituteNewcastle University Centre for CancerNewcastle upon TyneUK
| | - Alaric Taylor
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| | - Jugal Suthar
- Vesynta Ltd, Innovation Gateway, The London Cancer Hub, Cotswold Road, SuttonLondonUK
| |
Collapse
|
4
|
Tan WR, Irie K, McIntire C, Torres JL, Jones R, Gibson A, Mizuno T, Tang Girdwood S. Model-informed dose optimization for prophylactic piperacillin-tazobactam in perioperative pediatric critically ill patients. Antimicrob Agents Chemother 2025; 69:e0122724. [PMID: 39912647 PMCID: PMC11881557 DOI: 10.1128/aac.01227-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Piperacillin/tazobactam (PTZ) is frequently prescribed during the perioperative period as prophylaxis in critically ill patients. Current international guidelines recommend that the pediatric intraoperative dosing regimen for PTZ be 90-112.5 mg/kg (80-100 mg/kg as piperacillin [PIP]) administered every 2 hours (Q2H). Concerns have been raised not only about the risk of nephrotoxicity due to elevated PIP exposure but also regarding the practicality of adhering to a 2-h dosing interval in clinical settings. To address these concerns, we employed population pharmacokinetic (PK) modeling and simulation approaches to optimize PTZ dosing regimens in pediatric intraoperative patients. PIP plasma concentration data were obtained from 34 patients using an opportunistic sampling strategy. A two-compartment model was found to adequately describe the PK data. Creatinine clearance was identified as a significant covariate on clearance. The inclusion of inter-occasion variability significantly improved model fit. Simulations across body weights of 10-70 kg and creatinine clearance of 20-130 mL/min/1.73 m2 demonstrated that 6-15 mg/kg Q2H, or a 10 mg/kg loading dose followed by 1.0-2.75 mg/kg/h continuous infusion would achieve free PIP concentrations being above the minimum inhibitory concentration (MIC) for 100% of the dosing interval (100% fT >1× MIC). For achieving 100% fT >4× MIC, 25-55 mg/kg Q2H or a 20 mg/kg loading dose followed by 3.25-9.25 mg/kg/h continuous infusion was derived. The model-informed simulations indicated that both lower Q2H doses and continuous infusion methods are clinically viable options and potentially resolve current clinical challenges during intraoperative dosing.
Collapse
Affiliation(s)
- Wen Rui Tan
- Department of Pharmacology and System Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kei Irie
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carter McIntire
- Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Julie Luna Torres
- Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rhonda Jones
- Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Abigayle Gibson
- Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tomoyuki Mizuno
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sonya Tang Girdwood
- Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
5
|
Yang Y, Sokolov V, Volkova A, Liu X, Leon C, Kosinsky Y, Barker B, Zhang X, Ordentlich P, Sheng J, Chen X. Semimechanistic Population PK/PD Modeling of Axatilimab in Healthy Participants and Patients With Solid Tumors or Chronic Graft-Versus-Host Disease. Clin Pharmacol Ther 2025; 117:704-715. [PMID: 39704205 PMCID: PMC11835428 DOI: 10.1002/cpt.3503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024]
Abstract
Axatilimab, a high-affinity humanized immunoglobulin G4 monoclonal antibody against colony-stimulating factor 1 receptor (CSF-1R), is approved for the treatment of chronic graft-versus-host disease (cGVHD), and under investigation for idiopathic pulmonary fibrosis and solid tumors. The population pharmacokinetics (PK) and pharmacodynamics (PD) of axatilimab were characterized in healthy participants and patients with solid tumors or cGVHD using data from four clinical studies with 325 participants, including 278 patients with cGVHD. The model structure reflected the mechanism of action of axatilimab: blocking CSF-1R signaling with axatilimab reduces the circulating levels of cells in the mononuclear phagocytic cell lineage (including nonclassical monocytic cells (NCMCs) and Kupffer cells), resulting in increases in circulating enzymes owing to reduced clearance by Kupffer cells. The structural model consisted of a two-compartment axatilimab PK model and turnover PD models for CSF-1, NCMCs, aspartate transaminase (AST), and creatine phosphokinase (CPK). Axatilimab PK and CSF-1 equations also included saturable clearance components to reflect the competitive binding of axatilimab and CSF-1 to CSF-1R. Covariate search was conducted with the conditional sampling use for the stepwise approach based on correlation tests (COSSAC) approach. Covariate effects on model parameters, steady-state axatilimab exposure, and NCMC concentrations were assessed. The final population PK/PD model was mathematically described with 6 ordinary differential equations and 39 model parameters. Among the 11 statistically significant covariates, one (body weight) and two (participant population type and baseline CPK) covariates affected axatilimab steady-state exposure and steady-state NCMC levels by > 20%, respectively. These results informed the axatilimab dosing strategy in patients with cGVHD.
Collapse
MESH Headings
- Humans
- Male
- Adult
- Female
- Middle Aged
- Graft vs Host Disease/drug therapy
- Graft vs Host Disease/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Neoplasms/drug therapy
- Neoplasms/blood
- Models, Biological
- Aged
- Young Adult
- Healthy Volunteers
- Chronic Disease
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Adolescent
- Receptor, Macrophage Colony-Stimulating Factor
Collapse
Affiliation(s)
| | - Victor Sokolov
- M&S Decisions FZ LLCDubaiUnited Arab Emirates
- Marchuk Institute of Numerical Mathematics RASMoscowRussia
| | - Alina Volkova
- M&S Decisions FZ LLCDubaiUnited Arab Emirates
- Marchuk Institute of Numerical Mathematics RASMoscowRussia
| | - Xing Liu
- Incyte CorporationWilmingtonDelawareUSA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Wang J, Fang Q, Luo X, Jin L, Zhu H. Population pharmacokinetics and dosing optimization of imipenem in Chinese elderly patients. Front Pharmacol 2025; 15:1524272. [PMID: 39850576 PMCID: PMC11754279 DOI: 10.3389/fphar.2024.1524272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Objectives To assess the pharmacokinetics and pharmacodynamics of imipenem in a retrospective cohort of hospitalized Chinese older patients. Methods A population pharmacokinetic (PPK) model was constructed utilizing a nonlinear mixed-effects modeling approach. The final model underwent evaluation through bootstrap resampling and visual predictive checks. Additionally, a population pharmacokinetic and pharmacodynamic analysis was conducted employing Monte Carlo simulations to investigate the impact of commonly used dosing regimens (0.25 g every 6 h, 0.5 g every 6 h, 0.5 g every 8 h, 1 g every 6 h, 1 g every 8 h, and 1 g every 12 h) on the likelihood of achieving the target therapeutic outcomes. Results A total of 370 observations available from 142 patients were incorporated in the PPK model. A two-compartment PPK model with linear elimination best predicted the imipenem plasma concentrations, with the creatinine clearance as a significant covariate of clearance. Typical estimates for clearance, inter-compartmental clearance, central and peripheral volume were 13.1 L·h-1, 11.9 L·h-1, 11.7 L, 29.3 L, respectively. Conclusion The pharmacokinetics of imipenem in elderly patients were effectively characterized by the established PPK model, which includes creatinine clearance as a key covariate. This research will enhance our understanding of imipenem elimination and support precision dosing in this patient demographic.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Qiu Fang
- Department of Pharmacy, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuemei Luo
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Lu Jin
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Huaijun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
- Department of Pharmacy, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Melander E, Nielsen EI, Lindqvist A, Hovd M, Gandia P, Panchaud A, Guidi M, Annaert P, Baranczewski P, Spigset O, Nordeng H. Population pharmacokinetic modelling of cetirizine concentrations in human breast milk-A contribution from the ConcePTION project. Basic Clin Pharmacol Toxicol 2025; 136:e14100. [PMID: 39497323 DOI: 10.1111/bcpt.14100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 12/20/2024]
Abstract
Cetirizine is an antihistamine commonly used to treat allergic rhinitis and other allergic conditions. Cetirizine is often prescribed to breastfeeding mothers although there is limited information on infant exposure via breast milk. The aim of this study was to develop a popPK model based on data from a lactation study to predict cetirizine breast milk concentrations and estimate the relative infant dose (RID) in a breastfed infant. A popPK model was developed in NONMEM on data from a human lactation study including 35 women using cetirizine or levocetirizine while breastfeeding. Serial samples of breast milk were collected (n = 205) and the cetirizine concentrations quantified using a validated LC-MS/MS method. A one-compartment model of cetirizine in breast milk was developed and employed to calculate the relative infant dose (RID). Covariates related to the maternal characteristics and breastfeeding patterns were evaluated in the model; only milk sampling pumping duration was found to be a significant covariate, with an increasing pumping duration leading to an increased apparent milk volume of distribution (Vm). The mean RID was 1.99% with the highest RID being 3.36% at Cmax. PopPK modelling could be used to estimate infant exposure to cetirizine via breast milk. The low predicted exposure in infants supports that cetirizine is compatible with breastfeeding.
Collapse
Affiliation(s)
- Erik Melander
- Department of Pharmacy, Uppsala Drug Optimization and Pharmaceutical Profiling (UDOPP), Uppsala University, Uppsala, Sweden
| | - Elisabet I Nielsen
- Department of Pharmacy, Clinical Pharmacy research group, Uppsala University, Uppsala, Sweden
| | - Annika Lindqvist
- Department of Pharmacy, Uppsala Drug Optimization and Pharmaceutical Profiling (UDOPP), Uppsala University, Uppsala, Sweden
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Uppsala, Sweden
| | - Markus Hovd
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Peggy Gandia
- Pharmacokinetics and Toxicology laboratory, Toulouse University Hospital, Toulouse, France
- UMR1436-INTHERES, Toulouse National Veterinary School, Toulouse, France
| | - Alice Panchaud
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
- Service of Pharmacy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pawel Baranczewski
- Department of Pharmacy, Uppsala Drug Optimization and Pharmaceutical Profiling (UDOPP), Uppsala University, Uppsala, Sweden
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Uppsala, Sweden
| | - Olav Spigset
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hedvig Nordeng
- Department of Pharmacy, Pharmacoepidemiology and Drug Safety Research Group, University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Alasmari MS, Albusaysi S, Elhefnawy M, Ali AM, Altigani K, Almoslem M, Alharbi M, Alghamdi J, Alsultan A. Model-informed drug discovery and development approaches to inform clinical trial design and regulatory decisions: A primer for the MENA region. Saudi Pharm J 2024; 32:102207. [PMID: 39697476 PMCID: PMC11653594 DOI: 10.1016/j.jsps.2024.102207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Model-Informed Drug Discovery and Development (MID3) represents a transformative approach in pharmaceutical research, integrating quantitative models to inform and optimize decision-making throughout the drug development process. This review explores the current applications, challenges, and future prospects of MID3 within the Middle East and North Africa (MENA) region. By leveraging local data and advanced computational techniques, MID3 has the potential to significantly enhance the efficiency and success rates of drug development tailored to regional health priorities. We discussed successful case studies of applying MID3 at different phases of drug development and clinical trials. Furthermore, we emphasized the critical need for MENA countries to embrace MID3 by investing in workforce training, aligning regulatory frameworks, and fostering collaborative research initiatives. This call to action underscores the importance of a robust MID3 ecosystem, urging policymakers, academic institutions, and industry stakeholders to prioritize and support its integration into the MENA region's healthcare.
Collapse
Affiliation(s)
- Mohammed S. Alasmari
- Department of Pharmaceutical Services, Security Forces Hospital, Riyadh 11481, Saudi Arabia
| | - Salwa Albusaysi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | - Khalid Altigani
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Saudi Arabia
| | | | | | | | - Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
9
|
Cárdenas PA, Alves IA, De Araujo BV, Aragón DM. Effect of poly(ε-caprolactone) microspheres on population pharmacokinetic/pharmacodynamic model of a simple coumarin. J Microencapsul 2024; 41:739-753. [PMID: 39460601 DOI: 10.1080/02652048.2024.2418606] [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: 07/20/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
This study aims to evaluated the impact of poly(ε-caprolactone) (PCL) microspheres on the pharmacokinetics and pharmacodynamics (PopPK/PD) of 6-methylcoumarin (6MC). For this, PCL microspheres loaded with 6MC were prepared using the emulsification-evaporation method. Particle size, zeta potential, drug loading, and entrapment efficiency were characterised by dynamic light scattering and UV spectrophotometry. In vitro release and pharmacokinetics in Wistar rats were assessed for free and encapsulated 6MC. Anti-inflammatory activity was evaluated using the carrageenan-induced paw edoema model, with PopPK and PopPK/PD models developed. Microspheres showed diameters between 2.9 and 7.1 µm, zeta potentials of -10 to -15 mV, and drug loading of 0.24 mg/mg. Encapsulation efficiency was 45.5% to 75.9%. PopPK models showed enhanced absorption and distribution, with increased anti-inflammatory potency of encapsulated 6MC. PCL microspheres significantly improved the pharmacokinetic and pharmacodynamic profiles of 6MC, enhancing its therapeutic potential for lipophilic drugs.
Collapse
Affiliation(s)
- Paola A Cárdenas
- Departamento de Farmacia, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Izabel Almeida Alves
- Faculdade de Farmácia, Universidade Federal de Bahia, Salvador, BA, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Estado da Bahia, Salvador, BA, Brazil
| | - Bibiana Verlindo De Araujo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | |
Collapse
|
10
|
Iwama R, Nishida K, Ishii D, Iijima T. An integrated population pharmacokinetic model of febuxostat in pediatric patients with hyperuricemia including gout and adult population of healthy subjects and patients with renal dysfunction. Pharmacol Res Perspect 2024; 12:e70032. [PMID: 39523739 PMCID: PMC11551477 DOI: 10.1002/prp2.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024] Open
Abstract
The study objective was to validate febuxostat dosage and administration in pediatric patients with hyperuricemia including gout, using an integrated population pharmacokinetic (PopPK) analysis in the Japanese population. Integrated PopPK analysis of febuxostat used a nonlinear mixed-effects modeling (NONMEM) program on plasma febuxostat concentration data for 2611 samples from Japanese pediatric patients with hyperuricemia including gout (n = 29) and from adult subjects who are healthy or have renal dysfunction (n = 113). We described febuxostat pharmacokinetics using an integrated PopPK model applicable both to pediatric patients and to the adult population. The covariates of body weight and eGFR were identified for CL/F and the covariate of fasted/fed status for bioavailability. The range of steady-state exposures (Cmax,ss and AUCτ,ss) for 5, 10, 20, and 30 mg of febuxostat in fed pediatric patients weighing 20 to 40 kg was within that for 10, 20, 40, and 60 mg of febuxostat in fed pediatric patients and adults weighing 40 to 120 kg. Post hoc estimates of CL/F, adjusted by body weight, differed little between pediatric patients and the adult population in the renal function categories of normal, mild dysfunction, and moderate dysfunction. We successfully validated the febuxostat dose that provided the same level of exposure in pediatric patients as in the adult population: half the adult dose for pediatric patients weighing <40 kg and the full adult dose for pediatric patients weighing ≥40 kg. As in adults, the results support the use of febuxostat without dose adjustment in pediatric patients who have mild to moderate renal dysfunction.
Collapse
Affiliation(s)
- Ryutaro Iwama
- Translational Science Research DepartmentTeijin Institute for Bio‐medical Research, Teijin Pharma LimitedTokyoJapan
| | - Kimie Nishida
- Translational Science Research DepartmentTeijin Institute for Bio‐medical Research, Teijin Pharma LimitedTokyoJapan
| | - Daisuke Ishii
- Translational Science Research DepartmentTeijin Institute for Bio‐medical Research, Teijin Pharma LimitedTokyoJapan
| | - Takeshi Iijima
- Translational Science Research DepartmentTeijin Institute for Bio‐medical Research, Teijin Pharma LimitedTokyoJapan
| |
Collapse
|
11
|
Isla A, Alarcia-Lacalle A, Solinís MÁ, del Pozo-Rodríguez A, Abajo Z, Cabero M, Canut-Blasco A, Rodríguez-Gascón A. Population pharmacokinetics of oral fosfomycin calcium in healthy women. J Antimicrob Chemother 2024; 79:2837-2845. [PMID: 39205651 PMCID: PMC11531824 DOI: 10.1093/jac/dkae295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Fosfomycin is an antibiotic extensively used to treat uncomplicated urinary tract infections in women, and it is available in different salts and formulations. The European Medicines Agency (EMA) recommends further studies to characterize the pharmacokinetics of fosfomycin calcium for oral administration and to justify its dosage recommendation. OBJECTIVES A population pharmacokinetic model of fosfomycin calcium was developed after oral administration to healthy women. METHODS A clinical trial (a randomized, open-label, bioavailability study of single and multiple doses of 1000 mg capsules, single dose of 500 mg capsule and single dose of 250 mg/5 mL suspension of oral fosfomycin calcium under fasted conditions in healthy women volunteers, Code: PD7522.22, EudraCT: 2020-001664-28) was carried out at the Clinical Trial Unit, Araba University Hospital (Vitoria-Gasteiz, Spain). Twenty-four healthy women were included in the study, and plasma samples were collected at different times over a period of 24 h. The concentration-time data of fosfomycin in plasma were modelled by a population approach using a nonlinear mixed-effects modelling implemented by NONMEM 7.4 (ICON Clinical Research LLC, North Wales, PA, USA). RESULTS The pharmacokinetics of fosfomycin was best described by a two-compartment model. Creatinine clearance and body weight were identified as covariates for fosfomycin clearance and volume of distribution, respectively. CONCLUSIONS This study provides relevant information on the pharmacokinetic profile of fosfomycin in women after oral administration as calcium salt. This population model may be very useful for establishing dosage recommendations of fosfomycin calcium to treat urinary tract infections in women.
Collapse
Affiliation(s)
- Arantxa Isla
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
| | - Ana Alarcia-Lacalle
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
| | - Ana del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
| | - Zuriñe Abajo
- Bioaraba, Clinical Trials Unit, Vitoria-Gasteiz 01009, Spain
| | - María Cabero
- Bioaraba, Clinical Trials Unit, Vitoria-Gasteiz 01009, Spain
| | - Andrés Canut-Blasco
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
- Bioaraba, Microbiology Service, Araba University Hospital, Osakidetza Basque Health Service, Vitoria-Gasteiz 01009, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (Pharma Nano Gene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz 01009, Spain
| |
Collapse
|
12
|
Heerma van Voss MR, Notohardjo J, van Dodewaard-de Jong J, Bloemendal HJ, Ter Heine R. A sub-pharmacological test dose does not predict individual docetaxel exposure in prostate cancer patients. Cancer Chemother Pharmacol 2024; 94:437-441. [PMID: 38951305 PMCID: PMC11420247 DOI: 10.1007/s00280-024-04684-2] [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: 01/28/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024]
Abstract
PURPOSE Docetaxel is a cytotoxic drug used for first-line treatment of various malignancies. It has a narrow therapeutic index and shows wide interpatient variability in clearance and toxicity. Tools for individual dose optimization are needed to maximize efficacy and avoid toxicity. METHODS We performed a proof-of-concept study (EudraCT 2016-003785-77) to evaluate whether pharmacokinetics after a sub-pharmacological test dose of 1000 µg docetaxel (millidose) could be used to predict therapeutic dose exposure. Thirty prostate cancer patients eligible for treatment with docetaxel as part of routine clinical care were included. An intravenous docetaxel millidose was administered 1-7 days prior to therapeutic docetaxel. After both doses plasma docetaxel concentrations were measured by ultra- high performance liquid chromatography-tandem mass spectrometry. The docetaxel clearance was estimated with non-linear mixed effects modeling. RESULTS Geometric mean docetaxel clearance was 57.9 L/h (GCV 78.6%) after admission of a millidose and 40.3 L/h (GCV 60.7%) after admission of a therapeutic dose. The millidose and therapeutic dose in a single patient were not significantly correlated (Spearman's rho R = 0.02, P = 0.92). CONCLUSION Docetaxel pharmacokinetics at milli- and therapeutic dose level showed insufficient correlation for individual dose optimization. However, the clearance of a docetaxel millidose and full dose are within the same order of magnitude. Therefore, docetaxel millidose pharmacokinetics could potentially facilitate prediction of docetaxel pharmacokinetics at a population level in situations where therapeutic dose levels are impractical, such as pharmacokinetic drug-drug interaction studies or pediatric studies.
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands.
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Jessica Notohardjo
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Haiko J Bloemendal
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Watanabe F, Fujiwara K, Furuuchi K, Ito M, Hanada K, Kodama T, Aono A, Mitarai S, Yoshiyama T, Kurashima A, Ohta K, Morimoto K. Clofazimine serum concentration and safety/efficacy in nontuberculous mycobacterial pulmonary disease treatment. Respir Med 2024; 231:107718. [PMID: 38897551 DOI: 10.1016/j.rmed.2024.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Clofazimine (CFZ) has shown promising effects against Mycobacterium avium-intracellulare complex pulmonary disease (MAC-PD) and Mycobacterium abscessus species pulmonary disease (MABS-PD). However, the optimal CFZ dose remains unknown. We aimed to explore the relationship between steady-state CFZ concentration and its safety and efficacy in MAC-PD and MABS-PD. METHODS This prospective observational study focused on patients with MAC-PD and MABS-PD treated with CFZ (UMIN 000041053). To understand the safety and efficacy profile of CFZ and elucidate its optimal concentration, we analyzed CFZ-induced pigmentation grade, QTc interval, and culture conversion outcomes in relation to serum CFZ concentration using Student's t-test, a concentration-QTc model, and multivariable logistic regression analysis, respectively. In total, 64 patients (34 with MAC-PD; 30 with MABS-PD) were included. RESULTS The steady-state concentration of CFZ was higher in the moderate-to-severe pigmentation group than in the none-to-light pigmentation group (P < 0.001). At a CFZ concentration of 1 mg/L, the QTc interval was prolonged by 17.3 ms (95 % confidence interval [CI], 3.9-25.4) from baseline. Culture conversion was achieved in 33 (51.6 %) patients. The only significant predictor of culture conversion was surgery (adjusted odds ratio, 5.4; 95 % CI, 1.3-38.0). CFZ concentration and MIC of CFZ less than 0.25 mg/L were not associated with culture conversion in this study. CONCLUSION CFZ-induced pigmentation and QT interval prolongation are associated with serum CFZ concentrations. CFZ dosage may be optimized by monitoring serum CFZ concentration.
Collapse
Affiliation(s)
- Fumiya Watanabe
- Department of Pharmacometrics and Pharmacokinetics, Meiji Pharmaceutical University, Tokyo, Japan; Department of Pharmacy, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Keiji Fujiwara
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Koji Furuuchi
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Masashi Ito
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Kazuhiko Hanada
- Department of Pharmacometrics and Pharmacokinetics, Meiji Pharmaceutical University, Tokyo, Japan.
| | - Tatsuya Kodama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Akio Aono
- Department of Mycobacterium Reference and Research, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Satoshi Mitarai
- Department of Mycobacterium Reference and Research, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Takashi Yoshiyama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Atsuyuki Kurashima
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Ken Ohta
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Kozo Morimoto
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan; Division of Clinical Research, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| |
Collapse
|
14
|
Santos LO, Alves IA, Azeredo FJ. Pharmacokinetic Models of Tafenoquine: Insights for Optimal Malaria Treatment Strategies. Pharmaceutics 2024; 16:1124. [PMID: 39339162 PMCID: PMC11434791 DOI: 10.3390/pharmaceutics16091124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Tafenoquine (TQ) is a new 8-aminoquinoline antimalarial drug developed by the US Army for Plasmodium vivax malaria treatment. Modeling and simulation are essential tools for drug development and improving rationality in pharmacotherapy, and different modeling approaches are used. This study aims to summarize and explore the pharmacokinetic (PK) models available for tafenoquine in the literature. An integrative methodology was used to collect and review published data. Fifteen articles were identified using three modeling approaches: non-compartmental analysis (NCA), population pharmacokinetic analysis (popPK), and pharmacokinetic/pharmacodynamic analysis (PK/PD). An NCA was mainly used to describe the PK profile of TQ and to compare its PK profile alone to those obtained in association with other drugs. PopPK was used to assess TQ population PK parameters, covariates' impact, and dose selection. PK/PD helped understand the relationship between TQ concentrations, some adverse events common for 8-aminoquilones, and the efficacy assessment for Plasmodium falciparum. In summary, pharmacokinetic models were widely used during TQ development. However, there is still a need for different modeling approaches to support further therapeutic questions, such as treatment for special populations and potential drug-drug interactions.
Collapse
Affiliation(s)
- Luisa Oliveira Santos
- Laboratory of Pharmacokinetics and Pharmacometrics, Faculty of Pharmacy, Federal University of Bahia, Salvador 40170-110, Brazil
| | - Izabel Almeida Alves
- Laboratory of Pharmacokinetics and Pharmacometrics, Faculty of Pharmacy, Federal University of Bahia, Salvador 40170-110, Brazil
| | - Francine Johansson Azeredo
- Center for Pharmacometrics & System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| |
Collapse
|
15
|
Romański M, Staniszewska M, Dobosz J, Myslitska D, Paszkowska J, Kołodziej B, Romanova S, Banach G, Garbacz G, Sarcevica I, Huh Y, Purohit V, McAllister M, Wong SM, Danielak D. More Than a Gut Feeling─A Combination of Physiologically Driven Dissolution and Pharmacokinetic Modeling as a Tool for Understanding Human Gastric Motility. Mol Pharm 2024; 21:3824-3837. [PMID: 38958668 PMCID: PMC11345944 DOI: 10.1021/acs.molpharmaceut.4c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024]
Abstract
In vivo studies of formulation performance with in vitro and/or in silico simulations are often limited by significant gaps in our knowledge of the interaction between administered dosage forms and the human gastrointestinal tract. This work presents a novel approach for the investigation of gastric motility influence on dosage form performance, by combining biopredictive dissolution tests in an innovative PhysioCell apparatus with mechanistic physiology-based pharmacokinetic modeling. The methodology was based on the pharmacokinetic data from a large (n = 118) cohort of healthy volunteers who ingested a capsule containing a highly soluble and rapidly absorbed drug under fasted conditions. The developed dissolution tests included biorelevant media, varied fluid flows, and mechanical stress events of physiological timing and intensity. The dissolution results were used as inputs for pharmacokinetic modeling that led to the deduction of five patterns of gastric motility and their prevalence in the studied population. As these patterns significantly influenced the observed pharmacokinetic profiles, the proposed methodology is potentially useful to other in vitro-in vivo predictions involving immediate-release oral dosage forms.
Collapse
Affiliation(s)
- Michał Romański
- Department
of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznań, Poland
| | | | - Justyna Dobosz
- Physiolution
Polska, 74 Piłsudskiego
St., 50-020 Wrocław, Poland
| | - Daria Myslitska
- Physiolution
Polska, 74 Piłsudskiego
St., 50-020 Wrocław, Poland
| | | | | | | | - Grzegorz Banach
- Physiolution
Polska, 74 Piłsudskiego
St., 50-020 Wrocław, Poland
| | - Grzegorz Garbacz
- Physiolution
Polska, 74 Piłsudskiego
St., 50-020 Wrocław, Poland
| | - Inese Sarcevica
- Worldwide
Research and Development, Pfizer R&D
UK Ltd., Sandwich, CT13 9NJ, U.K.
| | - Yeamin Huh
- Worldwide
Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Vivek Purohit
- Worldwide
Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Mark McAllister
- Worldwide
Research and Development, Pfizer R&D
UK Ltd., Sandwich, CT13 9NJ, U.K.
| | - Suet M. Wong
- Worldwide
Research and Development, Pfizer R&D
UK Ltd., Sandwich, CT13 9NJ, U.K.
| | - Dorota Danielak
- Department
of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznań, Poland
| |
Collapse
|
16
|
Zhang LC, Li QY, Zhang YQ, Shan TC, Li Y, Li YH, Han H, Qin WD, Guo HP, Zhao W, Tang BH, Chen XM. Population pharmacokinetics of daptomycin in critically ill patients receiving extracorporeal membrane oxygenation. J Antimicrob Chemother 2024; 79:1697-1705. [PMID: 38814793 DOI: 10.1093/jac/dkae171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/10/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Daptomycin is widely used in critically ill patients for Gram-positive bacterial infections. Extracorporeal membrane oxygenation (ECMO) is increasingly used in this population and can potentially alter the pharmacokinetic (PK) behaviour of antibiotics. However, the effect of ECMO has not been evaluated in daptomycin. Our study aims to explore the effect of ECMO on daptomycin in critically ill patients through population pharmacokinetic (PopPK) analysis and to determine optimal dosage regimens based on both efficacy and safety considerations. METHODS A prospective, open-label PK study was carried out in critically ill patients with or without ECMO. The total concentration of daptomycin was determined by UPLC-MS/MS. NONMEM was used for PopPK analysis and Monte Carlo simulations. RESULTS Two hundred and ninety-three plasma samples were collected from 36 critically ill patients, 24 of whom received ECMO support. A two-compartment model with first-order elimination can best describe the PK of daptomycin. Creatinine clearance (CLCR) significantly affects the clearance of daptomycin while ECMO has no significant effect on the PK parameters. Monte Carlo simulations showed that, when the MICs for bacteria are ≥1 mg/L, the currently recommended dosage regimen is insufficient for critically ill patients with CLCR > 30 mL/min. Our simulations suggest 10 mg/kg for patients with CLCR between 30 and 90 mL/min, and 12 mg/kg for patients with CLCR higher than 90 mL/min. CONCLUSIONS This is the first PopPK model of daptomycin in ECMO patients. Optimal dosage regimens considering efficacy, safety, and pathogens were provided for critical patients based on pharmacokinetic-pharmacodynamic analysis.
Collapse
Affiliation(s)
- Li-Chen Zhang
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Qiu-Yue Li
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Qiu Zhang
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Ti-Chao Shan
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan Li
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-Hui Li
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Han
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wei-Dong Qin
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Hai-Peng Guo
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Pharmacy, The Second Hospital, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Pharmacy, The Second Hospital, Shandong University, Jinan, China
| | - Xiao-Mei Chen
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Uno M, Nakamaru Y, Yamashita F. Application of machine learning techniques in population pharmacokinetics/pharmacodynamics modeling. Drug Metab Pharmacokinet 2024; 56:101004. [PMID: 38795660 DOI: 10.1016/j.dmpk.2024.101004] [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: 11/16/2023] [Revised: 01/22/2024] [Accepted: 02/10/2024] [Indexed: 05/28/2024]
Abstract
Population pharmacokinetics/pharmacodynamics (pop-PK/PD) consolidates pharmacokinetic and pharmacodynamic data from many subjects to understand inter- and intra-individual variability due to patient backgrounds, including disease state and genetics. The typical workflow in pop-PK/PD analysis involves the determination of the structure model, selection of the error model, analysis based on the base model, covariate modeling, and validation of the final model. Machine learning is gaining considerable attention in the medical and various fields because, in contrast to traditional modeling, which often assumes linear or predefined relationships, machine learning modeling learns directly from data and accommodates complex patterns. Machine learning has demonstrated excellent capabilities for prescreening covariates and developing predictive models. This review introduces various applications of machine learning techniques in pop-PK/PD research.
Collapse
Affiliation(s)
- Mizuki Uno
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yuta Nakamaru
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
18
|
Sakaue T, Yamamoto K, Itohara K, Kitahiro Y, Endo T, Yokoyama N, Ishimura T, Omura T, Yano I. Population pharmacokinetics of everolimus in renal transplant recipients receiving long-term multiple immunosuppressive therapy. Drug Metab Pharmacokinet 2024; 56:101009. [PMID: 38547661 DOI: 10.1016/j.dmpk.2024.101009] [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: 01/08/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 06/24/2024]
Abstract
Everolimus is used for immunosuppression after renal transplantation. This study aimed to develop a population pharmacokinetic (PopPK) model of everolimus using therapeutic drug monitoring (TDM) data of patients under long-term multiple immunosuppressive therapy, including tacrolimus. To develop the model, 185 renal transplant recipients with 3358 everolimus blood concentrations during a median postoperative period of 35.3 months were included. The PopPK model is described as a one-compartment model with first-order absorption. The population mean of apparent clearance is 8.92 L/h (relative standard error = 3.6%), and this negatively correlated with the dose-normalized concentration (C/D) of tacrolimus and hematocrit value, and positively correlated with a daily dose of everolimus (i.e. TDM effect). The usefulness of dose adjustment using the final popPK model was assessed by a simulation study. The ratio of the first trough measurement within the therapeutic range of 3-8 ng/mL increased from 69.8% in the original dose to 87.9% in the individual dose calculated by the final PopPK model. The tacrolimus C/D ratio before initiating everolimus therapy and the hematocrit value were useful to estimate the initial dose of everolimus and can improve the safety and effectiveness of immunosuppressive therapy involving everolimus.
Collapse
Affiliation(s)
- Tomoyuki Sakaue
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kazuhiro Yamamoto
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kotaro Itohara
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yumi Kitahiro
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahito Endo
- Division of Urology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoki Yokoyama
- Division of Urology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takeshi Ishimura
- Division of Urology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomohiro Omura
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ikuko Yano
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
19
|
Wu W, Yang WS, Xu XY, Ge XL, Lu J, Wang GF, Wang Y, Li ZP. Population pharmacokinetics of oxcarbazepine active metabolite in Chinese paediatric patients with epilepsy: Model-based dose optimization. Basic Clin Pharmacol Toxicol 2024; 134:712-726. [PMID: 38506371 DOI: 10.1111/bcpt.14000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/07/2024] [Accepted: 02/19/2024] [Indexed: 03/21/2024]
Abstract
The pharmacological activity of oxcarbazepine (OXC) is primarily exerted through its active 10-monohydroxy metabolite (MHD). Nonetheless, there is limited pharmacokinetic information available regarding paediatric patients with epilepsy treated with OXC, especially in infants and toddlers. Concurrently, this drug exhibits substantial variability in pharmacokinetics and therapeutic response across different individuals. We aimed to develop a model to quantitatively investigate factors that affect MHD pharmacokinetics to formulate a dosage guideline for OXC in Chinese paediatric patients. A total of 297 MHD trough concentrations were obtained from 287 epileptic children. Six body weight (BW)-based allometric models were used for population pharmacokinetic modelling, while investigating the impact of other covariates on the apparent clearance. The one-compartment model and age cut-off model for the apparent clearance (CL/F) were established to describe the pharmacokinetics of MHD. The probability to obtain target trough concentration ranges (TTCRs) of MHD between 3 and 35 mg/L was determined by Monte Carlo simulations for doses ranging from 8 to 90 mg/kg/day. A new dose optimization strategy combining the dosage guidelines and Bayesian method provides a tailored approach for Chinese paediatric epileptic patients based on their individual BW and desired TTCRs of MHD, and also supports current dose recommendations, with the exception of children weighing ≤5 kg.
Collapse
Affiliation(s)
- Wei Wu
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Wen-Sheng Yang
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xiao-Yong Xu
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xi-Lin Ge
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jinmiao Lu
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Guang-Fei Wang
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Zhi-Ping Li
- Department of Clinical Pharmacy, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| |
Collapse
|
20
|
Ben Hassine K, Seydoux C, Khier S, Daali Y, Medinger M, Halter J, Heim D, Chalandon Y, Schanz U, Nair G, Cantoni N, Passweg JR, Satyanarayana Uppugunduri CR, Ansari M. Pharmacokinetic Modeling and Simulation with Pharmacogenetic Insights Support the Relevance of Therapeutic Drug Monitoring for Myeloablative Busulfan Dosing in Adult HSCT. Transplant Cell Ther 2024; 30:332.e1-332.e15. [PMID: 38081414 DOI: 10.1016/j.jtct.2023.12.003] [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: 09/17/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 01/01/2024]
Abstract
Therapeutic drug monitoring (TDM) of busulfan (Bu) is well-established in pediatric hematopoietic stem cell transplantation (HSCT), but its use in adults is limited due to a lack of clear recommendations and scarcity of evidence regarding its utility. GSTA1 promoter variants are reported to affect Bu clearance in both adults and pediatric patients. This study aimed to evaluate the value of preemptive genotyping GSTA1 and body composition (obesity) in individualizing Bu dosing in adults, through pharmacokinetic (PK) modeling and simulations. A population pharmacokinetic (PopPK) model was developed and validated with data from 60 adults who underwent HSCT. Simulations assessed different dosing scenarios based on body size metrics and GSTA1 genotypes. Due to the limited number of obese patients in the cohort, the effect of obesity on Bu pharmacokinetics (PK) was evaluated in silico using a physiologically-based pharmacokinetic (PBPK) model and relevant virtual populations from Simcyp software. Patients with at least 1 GSTA1*B haplotype had 17% lower clearance on average. PopPK simulations indicated that adjusting doses based on genotype increased the probability of achieving the target exposure (3.7 to 5.5 mg.h/L) from 53% to 60 % in GSTA1*A homozygous patients, and from 50% to 61% in *B carriers. Still, Approximately 40% of patients would not achieve this therapeutic window without TDM. A 2-sample optimal design was validated for routine model-based Bu first dose AUC0-∞ estimation, and the model was implemented in the Tucuxi user-friendly TDM software. PBPK simulations confirmed body surface area-based doses of 29 to 31 mg/m2/6h as the most appropriate, regardless of obesity status. This study emphasizes the importance of individualized Bu dosing strategies in adults to achieve therapeutic targets. Preemptive genotyping alone may not have a significant clinical impact, and routine TDM may be necessary for optimal transplantation outcomes.
Collapse
Affiliation(s)
- Khalil Ben Hassine
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claire Seydoux
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Sonia Khier
- Pharmacokinetic and Modeling Department, School of Pharmacy, Montpellier University, Montpellier, France; Probabilities and Statistics Department, Institut Montpelliérain Alexander Grothendieck (IMAG), CNRS, UMR 5149, Inria, Montpellier University, Montpellier, France
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, University Hospital of Geneva, Geneva, Switzerland; Faculty of Medicine & Sciences, University of Geneva, Geneva, Switzerland
| | - Michael Medinger
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Yves Chalandon
- Division of Hematology, Bone Marrow Transplant Unit, University Hospital of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Urs Schanz
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Gayathri Nair
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Nathan Cantoni
- Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Chakradhara Rao Satyanarayana Uppugunduri
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marc Ansari
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland.
| |
Collapse
|
21
|
Hansen JS, Boix F, Hasselstrøm JB, Sørensen L, Kjolby M, Gustavsen S, Hansen R, Petersen T, Sellebjerg F, Kasch H, Rasmussen PV, Finnerup NB, Sædder EA, Svendsen KB. Pharmacokinetics and pharmacodynamics of cannabis-based medicine in a patient population included in a randomized, placebo-controlled, clinical trial. Clin Transl Sci 2024; 17:e13685. [PMID: 38054364 PMCID: PMC10772478 DOI: 10.1111/cts.13685] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Information on the pharmacokinetics (PK) and pharmacodynamics (PD) of orally administered cannabis-based medicine (CBM) in capsule formulation in patient populations is sparse. In this exploratory study, we aimed to evaluate the PK and PD in a probable steady state of CBM in neuropathic pain and spasticity in a population of patients with multiple sclerosis (MS). Of 134 patients participating in a randomized, double-blinded, placebo-controlled, trial, 23 patients with MS (17 female) mean age 52 years (range 21-67) were enrolled in this substudy. They received oral capsules containing Δ9 -tetrahydrocannabinol (THC, n = 4), cannabidiol (CBD, n = 6), a combination (THC&CBD, n = 4), or placebo (n = 9). Maximum doses were 22.5 mg (THC) and 45 mg (CBD) a day divided into three administrations. PD parameters were evaluated for pain and spasticity. Blood samples were analyzed using an ultra-high-performance liquid chromatography-tandem mass spectrometer after protein precipitation and phospholipid removal. PK parameters were estimated using computerized modeling. The variation in daily dose and PK between individuals was considerable in a steady state, yet comparable with previous reports from healthy controls. Based on a simulation of the best model, the estimated PK parameters (mean) for THC (5 mg) were Cmax 1.21 ng/mL, Tmax 2.68 h, and half-life 2.75 h, and for CBD (10 mg) were Cmax 2.67 ng/mL, Tmax 0.10 h, and half-life 4.95 h, respectively. No effect was found on the PD parameters, but the placebo response was considerable. More immediate adverse events were registered in the active treatment groups compared with the placebo group.
Collapse
Affiliation(s)
- Julie Schjødtz Hansen
- Department of NeurologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Fernando Boix
- Section for Drug Abuse Research, Department of Forensic Sciences, Division of Laboratory MedicineOslo University HospitalOsloNorway
| | | | | | - Mads Kjolby
- Department of Clinical PharmacologyAarhus University HospitalAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Stefan Gustavsen
- Danish Multiple Sclerosis Center, Department of NeurologyCopenhagen University Hospital – RigshospitaletGlostrupDenmark
| | | | - Thor Petersen
- Department of NeurologyHospital of Southern Jutland and Research Unit in NeurologyAabenraaDenmark
- Department of Regional Health ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of NeurologyCopenhagen University Hospital – RigshospitaletGlostrupDenmark
| | - Helge Kasch
- Department of NeurologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | | | - Nanna Brix Finnerup
- Department of NeurologyAarhus University HospitalAarhusDenmark
- Danish Pain Research Centre, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Eva Aggerholm Sædder
- Department of Clinical PharmacologyAarhus University HospitalAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Kristina Bacher Svendsen
- Department of NeurologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
22
|
Kang SW, Jo HG, Kim D, Jeong K, Lee J, Lee HJ, Yang S, Park S, Rhie SJ, Chung EK. Population pharmacokinetics and model-based dosing optimization of teicoplanin in elderly critically ill patients with pneumonia. J Crit Care 2023; 78:154402. [PMID: 37634293 DOI: 10.1016/j.jcrc.2023.154402] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
PURPOSE To evaluate the population pharmacokinetics and pharmacodynamics of teicoplanin in elderly critically ill patients with pneumonia for optimal dosages. METHODS Fifteen critically ill patients (9 men) ≥ 60 years received teicoplanin 6 mg/kg for three doses followed by standard maintenance doses (6 mg/kg q24h) with renal dosing adjustment. Serial plasma samples from all patients were analyzed simultaneously by population pharmacokinetic modeling using NONMEM. Probability of target attainment (PTA) was calculated through Monte Carlo simulations for various dosing regimens to achieve adequate systemic exposures. RESULTS The median (interquartile range, IQR) age, body mass index, and creatinine clearance (CrCl) was 75 (64-78) years, 22.5 (20.8-25.4) kg/m2, and 64 (47-106) mL/min, respectively. The median (IQR) peak and trough concentration was 46.5 (42.7-51.0) and 8.7 (7.2-9.5) mg/L. The population pharmacokinetic model showed slower clearance (CL) and larger peripheral volume of distribution (V2) in patients with reduced CrCl: CL (L/h) = 0.629 × (CrCl/64)0.656, V2 (L) = 55.7 × (CrCl/64)-0.665. Model-based simulations showed PTAs ≥85% only for higher-dose regimens (12 mg/kg) up to an MIC of 0.5 mg/L. CONCLUSIONS Standard teicoplanin dosages for pneumonia may provide inadequate systemic exposures in elderly critically ill patients. High-dose regimens should be considered as empiric therapy or for less susceptible pathogens.
Collapse
Affiliation(s)
- Sung Wook Kang
- Department of Pulmonary and Critical Care Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Hyeong Geun Jo
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Donghyun Kim
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Institute of Regulatory Innovation through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyeoul Jeong
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaeok Lee
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hwa Jeong Lee
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seungwon Yang
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Institute of Regulatory Innovation through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sohyun Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea; Department of Pharmacy, National Medical Center, Seoul 04564, Republic of Korea
| | - Sandy Jeong Rhie
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Eun Kyoung Chung
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Institute of Regulatory Innovation through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea.
| |
Collapse
|
23
|
Taylor ZL, Poweleit EA, Paice K, Somers KM, Pavia K, Vinks AA, Punt N, Mizuno T, Girdwood ST. Tutorial on model selection and validation of model input into precision dosing software for model-informed precision dosing. CPT Pharmacometrics Syst Pharmacol 2023; 12:1827-1845. [PMID: 37771190 PMCID: PMC10725261 DOI: 10.1002/psp4.13056] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
There has been rising interest in using model-informed precision dosing to provide personalized medicine to patients at the bedside. This methodology utilizes population pharmacokinetic models, measured drug concentrations from individual patients, pharmacodynamic biomarkers, and Bayesian estimation to estimate pharmacokinetic parameters and predict concentration-time profiles in individual patients. Using these individualized parameter estimates and simulated drug exposure, dosing recommendations can be generated to maximize target attainment to improve beneficial effect and minimize toxicity. However, the accuracy of the output from this evaluation is highly dependent on the population pharmacokinetic model selected. This tutorial provides a comprehensive approach to evaluating, selecting, and validating a model for input and implementation into a model-informed precision dosing program. A step-by-step outline to validate successful implementation into a precision dosing tool is described using the clinical software platforms Edsim++ and MwPharm++ as examples.
Collapse
Affiliation(s)
- Zachary L. Taylor
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Ethan A. Poweleit
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of Biomedical InformaticsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Biomedical InformaticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Research in Patient ServicesCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Kelli Paice
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Katherine M. Somers
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Hematology and Oncology, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Kathryn Pavia
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Alexander A. Vinks
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Research in Patient ServicesCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Nieko Punt
- Department of Clinical Pharmacy and Pharmacology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
- MedimaticsMaastrichtThe Netherlands
| | - Tomoyuki Mizuno
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Sonya Tang Girdwood
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Hospital Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
24
|
Wang CB, Zhang YJ, Zhao MM, Zhao L. Dosage optimization of tacrolimus based on the glucocorticoid dose and pharmacogenetics in adult patients with systemic lupus erythematosus. Int Immunopharmacol 2023; 124:110866. [PMID: 37678026 DOI: 10.1016/j.intimp.2023.110866] [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: 06/26/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The purpose of the study was to develop a genotype-incorporated population pharmacokinetic (PPK) model of tacrolimus (TAC) in adults with systemic lupus erythematosus (SLE) to investigate the factors influencing TAC pharmacokinetics and to develop an individualized dosing regimen based on the model. In addition, a non-genotype-incorporated model was also established to assess its predictive performance compared to the genotype-incorporated model. METHODS A total of 365 trough concentrations from 133 adult SLE patients treated with TAC were collected to develop a genotype-incorporated PPK model and a non-genotype-incorporated PPK model of TAC using a nonlinear mixed-effects model (NONMEM). External validation of the two models was performed using data from an additional 29 patients. Goodness-of-fit diagnostic plots, bootstrap method, and normalized predictive distribution error test were used to validate the predictive performance and stability of the final models. The goodness-of-fit of the two final models was compared using the Akaike information criterion (AIC). The dosing regimen was optimized using Monte Carlo simulations based on the developed optimal model. RESULTS The typical value of the apparent clearance (CL/F) of TAC estimated in the final genotype-incorporated model was 14.3 L h-1 with inter-individual variability of 27.6%. CYP3A5 polymorphism and coadministered medication were significant factors affecting TAC-CL/F. CYP3A5 rs776746 GG genotype carriers had only 77.3% of the TAC-CL/F of AA or AG genotype carriers. Omeprazole reduced TAC-CL/F by 3.7 L h-1 when combined with TAC, while TAC-CL/F increased nonlinearly as glucocorticoid dose increased. Similar findings were demonstrated in the non-genotype-incorporated PPK model. Comparing these two models, the genotype-incorporated PPK model was superior to the non-genotype-incorporated PPK model (AIC = 643.19 vs. 657.425). Monte Carlo simulation based on the genotype-incorporated PPK model indicated that CYP3A5 rs776746 AA or AG genotype carriers required a 1/2-1 fold higher dose of TAC than GG genotype carriers to achieve the target concentration. And as the daily dose of prednisone increases, the dose of TAC required to reach the target concentration increases appropriately. CONCLUSIONS We developed the first pharmacogenetic-based PPK model of TAC in adult patients with SLE and proposed a dosing regimen based on glucocorticoid dose and CYP3A5 genotype according to the model, which could facilitate individualized dosing for TAC.
Collapse
Affiliation(s)
- Cheng-Bin Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu-Jia Zhang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ming-Ming Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
25
|
Tang Z, Guan J, Mao JH, Han L, Zhang JJ, Chen R, Jiao Z. Quantitative risk-benefit profiles of oral contraceptives, insulin sensitizers and antiandrogens for women with polycystic ovary syndrome: A model-based meta-analysis. Eur J Pharm Sci 2023; 190:106577. [PMID: 37666459 DOI: 10.1016/j.ejps.2023.106577] [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: 02/26/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Oral contraceptives (OCs), insulin sensitizers, and antiandrogens (AAs), alone or in combination, are commonly used for treating non-fertility indications in polycystic ovary syndrome (PCOS). However, unclear risk-benefit profiles jeopardize their appropriate clinical applications. This study aimed to quantitatively evaluate the effects of the aforementioned medications and to compare their risk-benefit profiles. Randomized controlled trials published until 14th March 2022 were searched in PubMed and Embase. A model-based meta-analysis was developed to examine the time-effect profiles of each medication. The maximal percentage change of the effect (Emax) and time to achieve half of Emax (T50) were estimated. Primary outcomes included menstruation, hirsutism score, free androgen index (FAI), body mass index (BMI), insulin sensitivity, and lipid profiles. Overall, 200 studies (9,685 patients and 385 arms) were identified for modeling. OCs performed exceptionally well in improving menstruation (Emax: 149%; T50: 7.44 weeks), hirsutism score (Emax: 66.2%; T50: 26.2 weeks), and FAI (Emax: 75.7%; T50: 0.51 weeks). However, OCs elevated the triglyceride (TG) level (Emax: 12.6%; T50:1.19 weeks). After 12-week OC treatment, the TG level of approximately 30% of patients, whose baselines were normal, exceeded the reference limit. This suggested that OC-induced dyslipidemia should be routinely monitored. The maximal BMI-lowering effect of metformin was similar to that of placebo (Emax: 3.80%); however, metformin had a shorter T50 (6.67 weeks versus 12.9 weeks). Further, active lifestyle intervention plus placebo significantly decreased BMI (Emax: 8.78%). Adding metformin to active lifestyle intervention accelerated the BMI-lowering effect within 24 weeks, whereas with the extension of this addition beyond 24 weeks, BMI did not reduce further, which indicated that benefits were limited from this prolonged addition. AAs were less potent in reducing hirsutism score (Emax: 40.2% versus 66.2%) and FAI (Emax: 34.5% versus 75.7%) compared to OCs. OC plus metformin combined OC-derived androgen-suppressing effects and metformin-derived insulin-sensitizing effects, and partially relieved the OC-induced TG increase (Emax: 9.76%). Baseline dependency was found in most clinical responses, implying that pharmacotherapies tailored based on baselines achieved more clinical improvements. This study presents new quantitative evidence on pharmacotherapies for PCOS. Currently, long-term risk-benefit profiles and emerging therapies are inadequately reported and require more further research.
Collapse
Affiliation(s)
- Zhe Tang
- Department of Pharmacy, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Department of Pharmacy, Shanghai Jiao Tong University Affiliated Chest Hospital, 241 Huai-hai West Road, Shanghai 200030, PR China
| | - Jing Guan
- Department of Pharmacy, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China
| | - Jue-Hui Mao
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Chest Hospital, 241 Huai-hai West Road, Shanghai 200030, PR China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lu Han
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Chest Hospital, 241 Huai-hai West Road, Shanghai 200030, PR China; School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Juan-Juan Zhang
- Center of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China
| | - Rui Chen
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Chest Hospital, 241 Huai-hai West Road, Shanghai 200030, PR China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Chest Hospital, 241 Huai-hai West Road, Shanghai 200030, PR China.
| |
Collapse
|
26
|
van Wijk RC, Imperial MZ, Savic RM, Solans BP. Pharmacokinetic analysis across studies to drive knowledge-integration: A tutorial on individual patient data meta-analysis (IPDMA). CPT Pharmacometrics Syst Pharmacol 2023; 12:1187-1200. [PMID: 37303132 PMCID: PMC10508576 DOI: 10.1002/psp4.13002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Answering challenging questions in drug development sometimes requires pharmacokinetic (PK) data analysis across different studies, for example, to characterize PKs across diverse regions or populations, or to increase statistical power for subpopulations by combining smaller size trials. Given the growing interest in data sharing and advanced computational methods, knowledge integration based on multiple data sources is increasingly applied in the context of model-informed drug discovery and development. A powerful analysis method is the individual patient data meta-analysis (IPDMA), leveraging systematic review of databases and literature, with the most detailed data type of the individual patient, and quantitative modeling of the PK processes, including capturing heterogeneity of variance between studies. The methodology that should be used in IPDMA in the context of population PK analysis is summarized in this tutorial, highlighting areas of special attention compared to standard PK modeling, including hierarchical nested variability terms for interstudy variability, and handling between-assay differences in limits of quantification within a single analysis. This tutorial is intended for any pharmacological modeler who is interested in performing an integrated analysis of PK data across different studies in a systematic and thorough manner, to answer questions that transcend individual primary studies.
Collapse
Affiliation(s)
- Rob C. van Wijk
- University of California San Francisco Schools of Pharmacy and MedicineSan FranciscoCaliforniaUSA
- UCSF Center for Tuberculosis, University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Marjorie Z. Imperial
- University of California San Francisco Schools of Pharmacy and MedicineSan FranciscoCaliforniaUSA
- UCSF Center for Tuberculosis, University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Radojka M. Savic
- University of California San Francisco Schools of Pharmacy and MedicineSan FranciscoCaliforniaUSA
- UCSF Center for Tuberculosis, University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Belén P. Solans
- University of California San Francisco Schools of Pharmacy and MedicineSan FranciscoCaliforniaUSA
- UCSF Center for Tuberculosis, University of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
27
|
Mizuno K, Capparelli EV, Fukuda T, Dong M, Adamson PC, Blumer JL, Cnaan A, Clark PO, Reed MD, Shinnar S, Vinks AA, Glauser TA. Model-Informed Precision Dosing Guidance of Ethosuximide Developed from a Randomized Controlled Clinical Trial of Childhood Absence Epilepsy. Clin Pharmacol Ther 2023; 114:459-469. [PMID: 37316457 DOI: 10.1002/cpt.2965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
Ethosuximide was identified as the optimal option for new-onset childhood absence epilepsy (CAE) in a randomized, two-phase dose escalation comparative effectiveness trial of ethosuximide, lamotrigine, and valproic acid. However, 47% of ethosuximide initial monotherapy participants experienced short-term treatment failure. This study aimed to characterize the initial monotherapy ethosuximide exposure-response relationship and to propose model-informed precision dosing guidance. Dose titration occurred over a 16-20-week period until patients experienced seizure freedom or intolerable side effects. Subjects with initial monotherapy failure were randomized to one of the other two medications and dose escalation was repeated. A population pharmacokinetic model was created using plasma concentration data (n = 1,320), collected at 4-week intervals from 211 unique participants during both the initial and second monotherapy phases. A logistic regression analysis was performed on the initial monotherapy cohort (n = 103) with complete exposure-response data. Eighty-four participants achieved seizure freedom with a wide range of ethosuximide area under the curves (AUC) ranging from 420 to 2,420 μg·h/mL. AUC exposure estimates for achieving a 50% and 75% probability of seizure freedom were 1,027 and 1,489 μg·h/mL, respectively, whereas the corresponding cumulative frequency of intolerable adverse events was 11% and 16%. Monte Carlo Simulation indicated a daily dose of 40 and 55 mg/kg to achieve 50% and 75% probability of seizure freedom in the overall population, respectively. We identified the need for adjusted mg/kg dosing in different body weight cohorts. This ethosuximide proposed model-informed precision dosing guidance to achieve seizure freedom carries promise to optimize initial monotherapy success for patients with CAE.
Collapse
Affiliation(s)
- Kana Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Edmund V Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Tsuyoshi Fukuda
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Peter C Adamson
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jeffery L Blumer
- Rainbow Clinical Research Center, Rainbow Babies and Children's Hospital, and Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Avital Cnaan
- Children's National Health System, Washington, DC, USA
| | - Peggy O Clark
- Comprehensive Epilepsy Center, Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michael D Reed
- Rainbow Clinical Research Center, Rainbow Babies and Children's Hospital, and Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shlomo Shinnar
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Tracy A Glauser
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- Comprehensive Epilepsy Center, Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Chen J, Huang X, Yu L, Li J, Yang R, Li L, Zhou J, Yao H, Bu S. Vancomycin population pharmacokinetics analysis in Chinese paediatric patients with varying degrees of renal function and ages: development of new practical dosing recommendations. J Antimicrob Chemother 2023:dkad202. [PMID: 37379498 PMCID: PMC10393882 DOI: 10.1093/jac/dkad202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023] Open
Abstract
OBJECTIVES To describe the pharmacokinetics of vancomycin in a large Chinese paediatric cohort with varying degrees of renal function and ages and to develop practical dosing guidelines. PATIENTS AND METHODS We conducted a retrospective population pharmacokinetic study using data from paediatric patients who received vancomycin between June 2013 and June 2022. A non-linear mixed-effect modelling approach with a one-compartment model structure was applied. Monte Carlo simulations were used to stimulate an optimal dosage regimen to achieve the target of AUC24/MIC between 400 and 650. RESULTS We analysed a total of 673 paediatric patients and 1547 vancomycin serum concentrations. Covariate analysis revealed that physiological maturation, renal function, albumin and cardiothoracic surgery (CTS) significantly affected vancomycin pharmacokinetics. The typical clearance and volume of distribution, standardized to 70 kg, were 7.75 L/h (2.3% relative standard error, RSE) and 36.2 L (1.7% RSE), respectively. Based on the model, we proposed an optimal dosing regimen that considers the patient's age and estimate glomerular filtration rate (eGFR) to achieve a target AUC24/MIC for CTS and non-CTS patients. We also found that a loading dose of 20 mg/kg can help patients with an eGFR of <60 mL/min/1.73 m2 achieve the target AUC on the first day of treatment. CONCLUSIONS We established vancomycin pharmacokinetic parameters in Chinese paediatric patients and proposed a dosing guideline integrating eGFR, age and CTS status, potentially improving clinical outcomes and reducing nephrotoxicity risk.
Collapse
Affiliation(s)
- Jihui Chen
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohui Huang
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liting Yu
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiru Li
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Yang
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lixia Li
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Zhou
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huijuan Yao
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuhong Bu
- Department of Clinical Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Zwart TC, Bezstarosti S, Achini FR, Reinders MEJ, Schilham MW, Heidt S, Guchelaar HJ, de Fijter JW, Moes DJAR. Population pharmacokinetics of subcutaneous alemtuzumab in kidney transplantation. Br J Clin Pharmacol 2023; 89:1471-1485. [PMID: 36408784 DOI: 10.1111/bcp.15608] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 10/30/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
AIM Alemtuzumab is a monoclonal antibody used as induction immunosuppressive therapy in kidney transplantation. It targets CD52 on lymphocytes, inducing profound immune cell depletion upon administration. Owing to its off-label status in kidney transplantation, its pharmacokinetic characteristics are largely unknown in this setting, and its current fixed dosing algorithm originates from other populations. We developed a population pharmacokinetic model for alemtuzumab in kidney transplant recipients and investigated the potential of personalized alemtuzumab therapy. METHODS In total, 362 pharmacokinetic observations drawn 0-165 days after transplantation were available from 61 adult kidney transplant recipients who received two consecutive doses of 15 mg alemtuzumab subcutaneously. A population pharmacokinetic model was developed using nonlinear mixed-effects modelling and applied to simulate various dosing regimens. RESULTS The alemtuzumab concentration-time data were best described by a two-compartmental model with first-order absorption and parallel first-order and time-varying concentration-dependent elimination, with between-subject variability on the first-order elimination (39.6%) and central distribution volume (39.6%). Alemtuzumab pharmacokinetics varied with body size, rendering lighter individuals exposed to lympholytic alemtuzumab concentrations (>0.1 mg/L) for prolonged durations as compared to their heavier peers. This between-subject variability could be reduced through lean bodyweight-adjusted dosing, showing a twofold to threefold reduction in the slope of the median alemtuzumab exposure over the bodyweight range. CONCLUSION Alemtuzumab displays substantial pharmacokinetic variability in kidney transplant recipients, which may warrant a personalized treatment strategy. Lean bodyweight-adjusted dosing poses an option for individualized dosing, but further evaluation of its potential clinical benefit is warranted.
Collapse
Affiliation(s)
- Tom C Zwart
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne Bezstarosti
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Federica R Achini
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine (Nephrology) and LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco W Schilham
- Department of Paediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan W de Fijter
- Department of Internal Medicine (Nephrology) and LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Caputo A, Racine A, Paule I, Tariot PN, Langbaum JB, Coello N, Riviere ME, Ryan JM, Lopez CL, Graf A. Rationale for the selection of dual primary endpoints in prevention studies of cognitively unimpaired individuals at genetic risk for developing symptoms of Alzheimer's disease. Alzheimers Res Ther 2023; 15:45. [PMID: 36879340 PMCID: PMC9987044 DOI: 10.1186/s13195-023-01183-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND There is a critical need for novel primary endpoints designed to detect early and subtle changes in cognition in clinical trials targeting the asymptomatic (preclinical) phase of Alzheimer's disease (AD). The Alzheimer's Prevention Initiative (API) Generation Program, conducted in cognitively unimpaired individuals at risk of developing AD (e.g., enriched by the apolipoprotein E (APOE) genotype), used a novel dual primary endpoints approach, whereby demonstration of treatment effect in one of the two endpoints is sufficient for trial success. The two primary endpoints were (1) time to event (TTE)-with an event defined as a diagnosis of mild cognitive impairment (MCI) due to AD and/or dementia due to AD-and (2) change from baseline to month 60 in the API Preclinical Composite Cognitive (APCC) test score. METHODS Historical observational data from three sources were used to fit models to describe the TTE and the longitudinal APCC decline, both in people who do and do not progress to MCI or dementia due to AD. Clinical endpoints were simulated based on the TTE and APCC models to assess the performance of the dual endpoints versus each of the two single endpoints, with the selected treatment effect ranging from a hazard ratio (HR) of 0.60 (40% risk reduction) to 1 (no effect). RESULTS A Weibull model was selected for TTE, and power and linear models were selected to describe the APCC score for progressors and non-progressors, respectively. Derived effect sizes in terms of reduction of the APCC change from baseline to year 5 were low (0.186 for HR = 0.67). The power for the APCC alone was consistently lower compared to the power of TTE alone (58% [APCC] vs 84% [TTE] for HR = 0.67). Also, the overall power was higher for the 80%/20% distribution (82%) of the family-wise type 1 error rate (alpha) between TTE and APCC compared to 20%/80% (74%). CONCLUSIONS Dual endpoints including TTE and a measure of cognitive decline perform better than the cognitive decline measure as a single primary endpoint in a cognitively unimpaired population at risk of AD (based on the APOE genotype). Clinical trials in this population, however, need to be large, include older age, and have a long follow-up period of at least 5 years to be able to detect treatment effects.
Collapse
Affiliation(s)
| | - Amy Racine
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | - Ines Paule
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | | | | | - Neva Coello
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | | | | | | | - Ana Graf
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| |
Collapse
|
31
|
Valentin T, Lambert M, Chaltiel L, Allal B, Mseddi M, Yakoubi M, Chevreau C, Toulmonde M, Firmin N, Filleron T, Chatelut E. Population pharmacokinetic analysis reveals no impact of aprepitant on the pharmacokinetics of ifosfamide, 2-dechloroifosfamide, and 3-dechloroifosfamide. Eur J Pharm Sci 2023; 185:106420. [PMID: 36882147 DOI: 10.1016/j.ejps.2023.106420] [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: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
PURPOSE Several case reports and retrospective series have clearly pointed to the role of aprepitant, an antiemetic drug, in the development of encephalopathy when used with ifosfamide. Described as an inhibitor of several CYP metabolic pathways, aprepitant is suspected of drug-drug-interaction on ifosfamide pharmacokinetics. The pharmacokinetics of ifosfamide and two of its metabolites (2-dechloroifosfamide and 3-dechloroifosfamide) was studied in patients with soft tissue sarcomas to evaluate the impact of aprepitant administration. METHODS A population pharmacokinetic approach was applied to analyze data obtained in 42 patients at cycle 1 (without aprepitant) and cycle 2 (with aprepitant for 34 of them). RESULTS A previously published pharmacokinetic model including a time-dependency process well fit the data. Aprepitant had no impact on ifosfamide or its two metabolite pharmacokinetic parameters. CONCLUSION This study suggests that aprepitant does not lead to a significant modification of ifosfamide metabolization, even though other metabolites such as 4 hydroxyifosfamide and chloroacetaldehyde were not monitored in this study.
Collapse
Affiliation(s)
| | - Marie Lambert
- Institut Claudius-Regaud, IUCT-Oncopole, Toulouse, France; CRCT, Université de Toulouse, Inserm
| | | | - Ben Allal
- Institut Claudius-Regaud, IUCT-Oncopole, Toulouse, France; CRCT, Université de Toulouse, Inserm
| | - Mourad Mseddi
- Institut Claudius-Regaud, IUCT-Oncopole, Toulouse, France; CRCT, Université de Toulouse, Inserm
| | - Malika Yakoubi
- Institut Claudius-Regaud, IUCT-Oncopole, Toulouse, France; CRCT, Université de Toulouse, Inserm
| | | | | | - Nelly Firmin
- Medical Oncology, Montpellier Cancer Institute, France
| | | | - Etienne Chatelut
- Institut Claudius-Regaud, IUCT-Oncopole, Toulouse, France; CRCT, Université de Toulouse, Inserm.
| |
Collapse
|
32
|
Colman RJ, Mizuno T, Fukushima K, Haslam DB, Hyams JS, Boyle B, Noe JD, D’Haens GR, Limbergen JV, Chun K, Yang J, Denson LA, Ollberding NJ, Vinks AA, Minar P. Real world population pharmacokinetic study in children and young adults with inflammatory bowel disease discovers novel blood and stool microbial predictors of vedolizumab clearance. Aliment Pharmacol Ther 2023; 57:524-539. [PMID: 36314265 PMCID: PMC9931651 DOI: 10.1111/apt.17277] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/04/2022] [Accepted: 10/15/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Vedolizumab for inflammatory bowel disease (IBD) is often intensified based on distinct pharmacokinetics in children. Prior adult-specific population pharmacokinetic models have identified limited covariates of drug clearance. AIMS To establish a population pharmacokinetic model for children and young adults to identify novel covariates of drug clearance to better account for paediatric-specific inter-patient variability in vedolizumab pharmacokinetics; a key secondary exploratory aim was to identify microbial signatures of pharmacokinetic outcomes in a subset of patients. METHODS The study included data from 463 observed vedolizumab concentrations (59 peaks and 404 troughs) from 74 patients with IBD (52 with Crohn's disease and 22 with ulcerative colitis or unclassified IBD, median age 16 years). Pharmacokinetic analysis was conducted with non-linear mixed effects modelling. For the evaluation of the exposure-response relationship, clinical outcomes were evaluated by trough levels, clearance and vedolizumab exposure. Whole-genome metagenomic sequencing was conducted at baseline and week 2. RESULTS A two-compartment population pharmacokinetic model was identified with a clear correlation between CL and weight, erythrocyte sedimentation rate, and hypoalbuminemia. Trough concentrations before infusion 3 (37 μg/ml) and before infusion 4 (20 μg/ml) best predicted steroid-free clinical remission at infusion 4. Using faecal metagenomics, we identified an early (baseline and week 2) abundance of butyrate-producing species and pathways that were associated with an infusion 4 trough concentration >20 μg/ml. CONCLUSIONS This novel paediatric vedolizumab pharmacokinetic model could inform precision dosing. While additional studies are needed, an abundance of faecal butyrate producers is associated with early response to vedolizumab, suggesting that microbial analysis may be beneficial to biological selection.
Collapse
Affiliation(s)
- Ruben J. Colman
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Keizo Fukushima
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center
| | - David B. Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Jeffrey S. Hyams
- Division of Digestive Diseases, Hepatology and Nutrition, Connecticut Children’s Medical Center
| | - Brendan Boyle
- Division of Gastroenterology, Hepatology and Nutrition, Nationwide Children’s Hospital
| | - Joshua D. Noe
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Wisconsin
| | - Geert R. D’Haens
- Gastroenterology and Hepatology, Amsterdam University Medical Centers – location University of Amsterdam, Amsterdam, the Netherlands
| | - Johan Van Limbergen
- Department of Pediatric Gastroenterology and Nutrition, Amsterdam University Medical Centers – Location University of Amsterdam, Emma Children’s Hospital, Amsterdam, the Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | | | | | - Lee A. Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Nicholas J. Ollberding
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center
| | - Alexander A. Vinks
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Phillip Minar
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
33
|
Li Y, Wang ML, Guo Y, Cao YF, Zhao MM, Zhao LM. Population pharmacokinetics and dosing regimen optimization of levetiracetam in epilepsy during pregnancy. Br J Clin Pharmacol 2023; 89:1152-1161. [PMID: 36260320 DOI: 10.1111/bcp.15572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/14/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022] Open
Abstract
AIMS The pharmacokinetics of levetiracetam (LEV) significantly changed during pregnancy. It is a great challenge to predict the adjusted doses of LEV to reach the preconception target concentrations. This study aimed to establish a population pharmacokinetic model of LEV in women with epilepsy (WWE) during pregnancy to analyse the factors of pharmacokinetic variability and to develop a model-based individualized dosing regimen. METHODS A total of 166 concentration-time points from 37 WWE during pregnancy treated with LEV were collected to analyse LEV pharmacokinetics with nonlinear mixed-effects modelling. The dosing regimen was optimized by Monte Carlo simulations based on the final model. RESULTS The LEV pharmacokinetics in pregnant WWE were best described by a 1-compartment model of first-order absorption and elimination. The population typical value of apparent clearance (CL/F) in the final model was estimated to be 3.82 L/h (95% confidence interval 3.283-4.357 L/h) with a relative standard error of 7.2%. Both total body weight (TBW) and trimester of pregnancy were significantly associated with LEV-CL/F during pregnancy; LEV-CL/F increased by 42.72% when TBW increased from 55 to 65 kg from the first trimester to the second trimester. Monte Carlo simulations showed that dosing regimens for LEV should be individualized based on the patient's TBW and trimester of pregnancy to maximize the likelihood of achieving the therapeutic range. CONCLUSION This first population pharmacokinetic study of LEV in WWE during pregnancy supports the use of a weight-based and pregnancy-based dosing regimen and can lay a foundation for further optimizing the individualized dosing regimens.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ming-Lu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Guo
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yun-Feng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, NHC Key Laboratory of Reproduction Regulation, Shanghai, China
| | - Ming-Ming Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Li-Mei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
34
|
Population Pharmacokinetics of CMAB008 (an Infliximab Biosimilar) and Remicade ® in Healthy Subjects and Patients with Moderately to Severely Active Rheumatoid Arthritis. Adv Ther 2023; 40:1005-1018. [PMID: 36607544 DOI: 10.1007/s12325-022-02396-8] [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: 10/19/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION CMAB008 is a monoclonal antibody developed as a biosimilar to infliximab (Remicade®, Janssen). The pharmacokinetic characteristics of CMAB008 and Remicade® in healthy subjects and patients with moderately to severely active rheumatoid arthritis (RA) were investigated using a population modeling approach, and the pharmacokinetic similarity of CMAB008 to Remicade® was assessed. METHODS The population pharmacokinetic model was developed on the basis of intensive pharmacokinetic data from a phase 1 study in healthy male subjects and combined intensive and sparse pharmacokinetic data from a phase 3 study in patients with RA. RESULTS A two-compartment model with first-order elimination adequately described CMAB008 and Remicade® concentration data in healthy subjects and patients with RA. The analysis of covariates identified anti-drug antibody (ADA), neutralizing antibody (NAB), real-time body weight (BWT), and real-time albumin (ALB) as significant covariates on clearance, and BWT was also a significant covariate for the central volume of distribution. The treatment type (CMAB008 versus Remicade®) and the study population (healthy subjects versus patients with RA) were not identified as significant covariates on the pharmacokinetics of infliximab, demonstrating pharmacokinetic similarity between CMAB008 and Remicade® in both study populations. The effect of BWT and ALB changes on exposures to infliximab was within the acceptable range, suggesting that the 3 mg/kg regimen is appropriate in clinical practice for patients with RA and BTW and ALB distribution within the range evaluated in the current analysis. CONCLUSIONS The pharmacokinetic characteristics were similar between CMAB008 and Remicade® in healthy subjects and patients with RA. CMAB008 can be considered bioequivalent to Remicade®. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT04779892, NCT03478111.
Collapse
|
35
|
Wang C, Chen J, Yang B, Li S, Zhang Y, Chen L, Wang T, Dong Y. Determination of vancomycin exposure target and individualized dosing recommendations for critically ill patients undergoing continuous renal replacement therapy. Pharmacotherapy 2023; 43:180-188. [PMID: 36714991 DOI: 10.1002/phar.2771] [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: 09/15/2022] [Revised: 12/18/2022] [Accepted: 12/25/2022] [Indexed: 01/31/2023]
Abstract
STUDY OBJECTIVE Few studies have been conducted to quantify the exposure target of vancomycin in intensive care unit (ICU) patients undergoing continuous renal replacement therapy (CRRT) and provide optimized dosage regimens. We aimed to determine vancomycin exposure target and dosing recommendations using data from an open database in critically ill patients undergoing CRRT. DESIGN A retrospective observational cohort study. DATA SOURCE A large public database. PATIENTS The adult patients who received intravenous vancomycin and CRRT treatment in the database between 2017 and 2019 were reviewed to determine eligibility. A total of 180 patients with 1186 observations were included in the population pharmacokinetic (PPK) model development. The clinical efficacy of vancomycin was analyzed in 159 eligible patients. METHODS A PPK model was developed to estimate individual pharmacokinetic (PK) parameters. The area under the concentration-time curve (AUC) was estimated by a Bayesian approach based on individual vancomycin concentrations. Multivariate logistic regression analyses were performed to identify the factors of clinical outcomes. Threshold of vancomycin exposure in predicting efficacy was identified via receiver operating characteristic (ROC) curve. Dosing recommendations were designed using Monte Carlo Simulations (MCS) based on the optimized exposure target. MEASUREMENTS AND MAIN RESULTS On covariate analysis, CRRT intensity significantly affected vancomycin PK. The AUC above 427 mg*h/L was the only significant predictor of clinical efficacy (adjusted odds ratio (aOR): 1.008, 95% confidence interval (CI): 1.004-1.011, p = 0.000). MCS indicated that vancomycin dosage regimens of 5 mg/kg q12h or 7.5 mg/kg q12h were recommended for patients with CRRT intensities of 20-25 mL/kg/h or 25.1-45 mL/kg/h, respectively. CONCLUSIONS An AUC threshold of 427 mg*h/L (assuming the minimal inhibitory concentration (MIC) = 1 mg/L) was a recommended efficacy exposure target of vancomycin for critically ill patients undergoing CRRT. Vancomycin 5-7.5 mg/kg q12h is recommended as the initial dosage regimens for ICU patients undergoing CRRT.
Collapse
Affiliation(s)
- Chuhui Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaojiao Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bo Yang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sihan Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiran Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Lei Chen
- Department of Hemodialysis, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Taotao Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
36
|
Lee TY, Pierrillas PB, Lin YW, de Greef R, Zandvliet AS, Schindler E, Migoya E. Population PK and Semimechanistic PK/PD Modeling and Simulation of Relugolix Effects on Testosterone Suppression in Men with Prostate Cancer. Clin Pharmacol Ther 2023; 113:124-134. [PMID: 36073238 PMCID: PMC10091729 DOI: 10.1002/cpt.2743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/21/2022] [Indexed: 12/24/2022]
Abstract
Relugolix, the first orally active, nonpeptide gonadotropin-releasing hormone receptor antagonist, is approved in the United States and the European Union for the treatment of adult patients with advanced prostate cancer. The recommended dosing regimen is a 360-mg loading dose followed by a 120-mg daily dose. Relugolix and testosterone concentration data and clinical information from two phase I studies, two phase II studies, and the phase III safety and efficacy study (HERO) were used to develop a population pharmacokinetic (PopPK) model and a semimechanistic population pharmacokinetic/pharmacodynamic (PopPK/PD) model that characterized relugolix exposure and its relationship to testosterone concentrations. Age, body weight, and Black/African American race had at most minimal effects on relugolix exposure or testosterone concentrations with no clinical relevance. Simulations using the PopPK/PD model confirmed the recommended dosing regimen of relugolix, with the median simulated testosterone concentrations predicted to achieve castration levels (< 50 ng/dL) and profound castration levels (< 20 ng/dL) by day 2 and day 9, respectively, and demonstrated that 97.3% and 85.5% of the patients remained at castration levels (< 50 ng/dL) upon temporary interruption of treatment for 7 days and 14 days, respectively. Collectively, simulations based on the PopPK and PopPK/PD models were consistent with actual data from clinical studies, reflecting the high predictiveness of the models and supporting the reliability of model-based simulations. These models can be used to provide guidance regarding dosing recommendations under various circumstances (e.g., temporary interruption of treatment, if needed) for relugolix.
Collapse
Affiliation(s)
- Tien-Yi Lee
- Myovant Sciences, Inc., Brisbane, California, USA
| | | | | | | | | | - Emilie Schindler
- Certara, Princeton, New Jersey, USA.,F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | |
Collapse
|
37
|
A Survey of Population Pharmacokinetic Reports Submitted to the USFDA: An Analysis of Common Issues in NDA and BLA from 2012 to 2021. Clin Pharmacokinet 2022; 61:1697-1703. [PMID: 36316621 DOI: 10.1007/s40262-022-01182-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Population pharmacokinetic (PopPK) analysis is one of the important components of regulatory submission. The purpose of this study was to survey PopPK analysis reports in new drug applications (NDAs) and biological licensing applications (BLAs) submitted to the US Food and Drug Administration (FDA) and to retrieve the information regarding PopPK-related issues in the US FDA review reports. METHODS We surveyed NDAs and BLAs over the period from 2012 to 2022 from Drug@FDA databases and extracted the review reports from the website. We explored the issues identified in these reports and sorted them into four categories: (i) data used for PopPK analysis, (ii) base model development, (iii) final model development, and (iv) model evaluation. The percentage of the issues in each category and the total issues were calculated. RESULTS Data from a total of 317 applications were analyzed. Of these, 122 applications had at least one PopPK-related issue, and the count went up to 168 issues. The proportion of issues for each category was ranked as follows: model evaluation (39%, 65 of 168), final model development (34%, 57 of 168), data used for PopPK analysis (14%, 24 of 168), and base model development (13%, 22 of 168). The most common issues were related to the goodness-of-fit plots, covariate selection, and high shrinkage of ETA, accounting for 17.9%, 15.5%, and 11.3% of the total issues, respectively. CONCLUSION The findings from this study may help the applicant understand the FDA's thinking on the PopPK analysis.
Collapse
|
38
|
van der Hoek S, Mulder DJ, Willemsen AT, Visser T, Heeres A, Slart RH, Elsinga PH, Heerspink HJ, Stevens J. Studying Telmisartan Plasma Exposure, Kidney Distribution, Receptor Occupancy, and Response in Patients With Type 2 Diabetes Using [ 11 C]Telmisartan. Clin Pharmacol Ther 2022; 112:1264-1270. [PMID: 36070078 PMCID: PMC9827889 DOI: 10.1002/cpt.2744] [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: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 01/31/2023]
Abstract
The angiotensin receptor blocker telmisartan slows progression of kidney disease in patients with type 2 diabetes (T2D), yet many patients remain at high risk for progressive kidney function loss. The underlying mechanisms for this response variation might be attributed to differences in angiotensin-1 receptor occupancy (RO), resulting from individual variation in plasma drug exposure, tissue drug exposure, and receptor availability. Therefore, we first assessed the relationship between plasma telmisartan exposure and urinary-albumin-to-creatinine-ratio (UACR) in 10 patients with T2D and albuminuria (mean age 66 years, median UACR 297 mg/g) after 4 weeks treatment with 80 mg telmisartan once daily. Increasing telmisartan exposure associated with a larger reduction in UACR (Pearson correlation coefficient (PCC) = -0.64, P = 0.046, median change UACR: -40.1%, 95% confidence interval (CI): -22.9 to -77.4%, mean telmisartan area under the curve (AUC) = 2927.1 ng·hour/mL, 95% CI: 723.0 to 6501.6 ng·hour/mL). Subsequently, we assessed the relation among plasma telmisartan exposure, kidney distribution, and angiotensin-1 RO in five patients with T2D (mean age 60 years, median UACR 72 mg/g) in a separate positron emission tomography imaging study with [11 C]Telmisartan. Individual plasma telmisartan exposure correlated with telmisartan distribution to the kidneys (PCC = 0.976, P = 0.024). A meaningful RO could be calculated in three patients receiving 120 mg oral telmisartan, and although high exposure seems related to higher RO, with AUC0-last of 31, 840, and 274 ng·hour/mL and corresponding RO values 5.5%, 44%, and 59%, this was not significant (P = 0.64). Together these results indicate, for the first time, a relationship among interindividual differences in plasma exposure, kidney tissue distribution, RO, and ultimately UACR response after telmisartan administration.
Collapse
Affiliation(s)
- Sjoukje van der Hoek
- Department of Clinical Pharmacy and PharmacologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Douwe J. Mulder
- Division of Vascular Medicine, Department of Internal MedicineUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Antoon T.M. Willemsen
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | | | - Andre Heeres
- SymeresGroningenThe Netherlands,Hanze University of Applied SciencesGroningenThe Netherlands
| | - Riemer H.J.A. Slart
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Hiddo J.L. Heerspink
- Department of Clinical Pharmacy and PharmacologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Jasper Stevens
- Department of Clinical Pharmacy and PharmacologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| |
Collapse
|
39
|
Guo J, Lin W, Weng Y, Chen Y, Zeng S, Lin J, Zheng X, Li X, Lin M, Yu X, Chen Q. Optimal exposure to docetaxel in adjuvant chemotherapy for early-stage breast cancer. J Clin Pharm Ther 2022; 47:2205-2213. [PMID: 36418195 DOI: 10.1111/jcpt.13793] [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: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/26/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Drug-induced neutropenia is the main reason for the dose limitation of docetaxel in patients with breast cancer. The area under the drug concentration-time curve (AUC) of docetaxel is associated with neutropenia. However, the optimal exposure to docetaxel for receiving postoperative adjuvant chemotherapy remains unclear. Therefore, we aimed to evaluate the relationship between the docetaxel AUC and neutropenia, identify potential influencing factors, and explore the best monitoring target for docetaxel when treating patients with early-stage breast cancer using a population pharmacokinetic (PopPK) model. METHODS Docetaxel plasma concentration, demographics, clinical data, and related laboratory data were collected. PopPK analyses were performed using a nonlinear mixed-effect modelling program. The docetaxel AUC was determined using the maximum a posteriori Bayesian (MAPB) method. The docetaxel exposure-toxicity threshold measured from the AUC for neutropenia was determined using the receiver operating characteristic (ROC) curve. The correlation between docetaxel exposure and neutropenia was analysed using multivariable logistic regression. RESULTS Among the 70 participants, 47 (67.1%) developed severe neutropenia. The PopPK analysis showed that the typical drug clearance (CL) rate was 37.4 L/h. Age was a significant covariate of CL rate, and aspartate aminotransferase and albumin levels were covariables of the volume of distribution. The multivariable regression analysis showed that AUC >3.0 mg.h/L (odds ratio [OR], 5.940; 95% confidence interval [CI], 1.693-20.843; P = 0.005), platinum use (OR, 0.156; 95% CI, 0.043-0.562; P = 0.005) and baseline haemoglobin level (OR, 0.938; 95% CI, 0.887-0.993; P = 0.027) were significant factors influencing the occurrence of grade 3/4 neutropenia. The AUC of first cycle may not predict the occurrence rates of grade 3/4 neutropenia in later cycles. WHAT IS NEW AND CONCLUSION We developed a docetaxel PopPK model for patients with early-stage breast cancer. Age and AST and ALB levels were significant covariates. AUC estimated using the MAPB method can predict the toxicity of docetaxel in patients with breast cancer. Docetaxel AUC >3.0 mg.h/L, absence of platinum use and low baseline haemoglobin level were risk factors for docetaxel-induced grade 3/4 neutropenia. STUDY REGISTRATION Chinese Clinical Trial Center Registry (ChiCTR2200056460).
Collapse
Affiliation(s)
- Jujiang Guo
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Weijie Lin
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yiyin Weng
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yao Chen
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shaowu Zeng
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Juli Lin
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiuluan Zheng
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiuqing Li
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Min Lin
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xuefen Yu
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Quanyao Chen
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
40
|
Shen X, Chen X, Lu J, Chen Q, Li W, Zhu J, He Y, Guo H, Xu C, Fan X. Pharmacogenetics-based population pharmacokinetic analysis and dose optimization of valproic acid in Chinese southern children with epilepsy: Effect of ABCB1 gene polymorphism. Front Pharmacol 2022; 13:1037239. [PMID: 36506519 PMCID: PMC9733833 DOI: 10.3389/fphar.2022.1037239] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022] Open
Abstract
Objective: The aim of this study was to establish a population pharmacokinetic (PPK) model of valproic acid (VPA) in pediatric patients with epilepsy in southern China, and provide guidance for individualized medication of VPA therapy. Methods: A total of 376 VPA steady-state trough concentrations were collected from 103 epileptic pediatric patients. The PPK parameter values for VPA were calculated by using the nonlinear mixed-effects modeling (NONMEM) method, and a one-compartment model with first-order absorption and elimination processes was applied. Covariates included demographic information, concomitant medications and selected gene polymorphisms. Goodness-of-fit (GOF), bootstrap analysis, and visual predictive check (VPC) were used for model evaluation. In addition, we used Monte Carlo simulations to propose dose recommendations for different subgroup patients. Results: A significant effect of the patient age and ABCB1 genotypes was observed on the VPA oral clearance (CL/F) in the final PPK model. Compared with patients with the ABCB1 rs3789243 AA genotype, CL/F in patients with GG and AG genotypes was increased by 8% and reduced by 4.7%, respectively. The GOF plots indicated the satisfactory predictive performance of the final model, and the evaluation by bootstrap and VPC showed that a stable model had been developed. A table of individualized dosing regimens involving age and ABCB1 genotype was constructed based on the final PPK model. Conclusion: This study quantitatively investigated the effects of patient age and ABCB1 rs3789243 variants on the pharmacokinetic variability of VPA. The PPK models could be beneficial to individual dose optimization in epileptic children on VPA therapy.
Collapse
Affiliation(s)
- Xianhuan Shen
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Xinyi Chen
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jieluan Lu
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Qing Chen
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Wenzhou Li
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Jiahao Zhu
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Yaodong He
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Huijuan Guo
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Chenshu Xu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Xiaomei Fan
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
41
|
Population Pharmacokinetic Modeling and Probability of Target Attainment of Ceftaroline in Brain and Soft Tissues. Antimicrob Agents Chemother 2022; 66:e0074122. [PMID: 36005769 PMCID: PMC9487611 DOI: 10.1128/aac.00741-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ceftaroline, approved to treat skin infections and pneumonia due to methicillin-resistant Staphylococcus aureus (MRSA), has been considered for the treatment of central nervous system (CNS) infections. A population pharmacokinetic (popPK) model was developed to describe ceftaroline soft tissue and cerebrospinal fluid (CSF) distributions and investigate the probability of target attainment (PTA) of the percentage of the dosing interval that the unbound drug concentration exceeded the MIC (%fT>MIC) to treat MRSA infections. Healthy subjects' plasma and microdialysate concentrations from muscle and subcutaneous tissue following 600 mg every 12 h (q12h) and q8h and neurosurgical patients' plasma and CSF concentrations following single 600-mg dosing were used. Plasma concentrations were described by a two-compartment model, and tissue concentrations were incorporated as three independent compartments linked to the central compartment by bidirectional transport (clearance in [CLin] and CLout). Apparent volumes were fixed to physiological interstitial values. Healthy status and body weight were identified as covariates for the volume of the central compartment, and creatinine clearance was identified for clearance. The CSF glucose concentration (GLUC) was inversely correlated with CLin,CSF. Simulations showed a PTA of >90% in plasma and soft tissues for both regimens assuming an MIC of 1 mg/L and a %fT>MIC of 28.8%. Using the same target, patients with inflamed meninges (0.5 < GLUC ≤ 2 mmol/L) would reach PTAs of 99.8% and 97.2% for 600 mg q8h and q12h, respectively. For brain infection with mild inflammation (2 < GLUC ≤ 3.5 mmol/L), the PTAs would be reduced to 34.3% and 9.1%, respectively. Ceftaroline's penetration enhanced by meningeal inflammation suggests that the drug could be a candidate to treat MRSA CNS infections.
Collapse
|
42
|
Pharmacokinetics and Adverse Effects of Clofazimine in the Treatment of Pulmonary Non-Tuberculous Mycobacterial Infection. Antimicrob Agents Chemother 2022; 66:e0044122. [PMID: 35862744 PMCID: PMC9380550 DOI: 10.1128/aac.00441-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Clofazimine (CFZ) is used to treat pulmonary non-tuberculous mycobacterial (NTM) infection; however, its pharmacokinetics remain unexplored in patients with pulmonary NTM, and the relationship between CFZ serum concentration and adverse effects has not been investigated. The objectives of this study were to characterize the pharmacokinetics of CFZ in pulmonary NTM disease treatment and to investigate the relationship between the steady-state CFZ serum concentration and adverse effects. A prospective observational study was conducted on 45 patients with pulmonary NTM treated with CFZ (UMIN000041053). A maximum of five serum samples per patient were taken at the CFZ trough, and serum concentration was measured using high-performance liquid chromatography-mass spectrometry (HPLC-MS). The pharmacokinetics of CFZ were analyzed using a nonlinear mixed effect model. The relationships among steady-state CFZ serum concentration and adverse effects, pigmentation, and heart rate-corrected QT (QTc) interval were investigated. Twenty-six patients had M. avium or M. intracellulare infection and nineteen had M. abscessus infection. The primary CFZ dosage was 50 mg/day. The estimated apparent CFZ clearance, apparent volume of distribution, and half-life were 2.4 L/h, 2,960 L, and 36 days, respectively. The combined use of rifampicin and CFZ significantly reduced CFZ exposure by 22%. Although there was no relationship between CFZ serum concentration and pigmentation intensity, the QTc interval was significantly correlated with CFZ serum concentration. The estimation of accurate pharmacokinetics for CFZ required approximately 5 months of monitoring. The relationship between the serum concentration and specific adverse effects of CFZ confirmed that CFZ serum concentration was not associated with pigmentation but did affect the QTc interval.
Collapse
|
43
|
Langeskov EK, Kristensen K. Population pharmacokinetic of paracetamol and atorvastatin with co-administration of semaglutide. Pharmacol Res Perspect 2022; 10:e00962. [PMID: 35799471 PMCID: PMC9263537 DOI: 10.1002/prp2.962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 04/02/2022] [Indexed: 11/09/2022] Open
Abstract
Semaglutide is a glucagon-like-peptide-1 (GLP-1) analogue marketed for once-weekly subcutaneous administration for type 2 diabetes mellitus. Like other long-acting GLP-1 analogues, semaglutide reduces gastric emptying and, potentially, alters the rate of absorption of orally co-administered drugs. The objective of the current analysis was to evaluate the effects on the gastric emptying rate caused by semaglutide on pharmacokinetic model parameters of paracetamol and atorvastatin in healthy subjects. Non-linear mixed effect modeling was used to estimate population pharmacokinetic model parameters of paracetamol and atorvastatin after single doses with or without semaglutide. The absorption rate (ka) of paracetamol decreased by 53% when co-administered with semaglutide. For atorvastatin, ka and transit compartment rate (ktr) decreased by 72% and 91%, respectively. Thus, gastric emptying, measured as T50, i.e., drug disappearance from the absorption compartments, showed an additional 5-min delay for paracetamol and a 67-min delay for atorvastatin when co-administered with semaglutide. Semaglutide affected pharmacokinetic model parameters of paracetamol and atorvastatin, and minor quantitative differences in gastric emptying between placebo vs. semaglutide administration were observed. However, these effects of semaglutide were considered not to be of clinical relevance.
Collapse
Affiliation(s)
- Emilie K Langeskov
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Kristensen
- Discovery & Development PKPD, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
44
|
Population pharmacokinetic modelling of imatinib in healthy subjects receiving a single dose of 400 mg. Cancer Chemother Pharmacol 2022; 90:125-136. [PMID: 35831644 PMCID: PMC9360108 DOI: 10.1007/s00280-022-04454-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/20/2022] [Indexed: 11/06/2022]
Abstract
Purpose Imatinib is indicated for treatment of CML, GIST, etc. The population pharmacokinetics (popPK) of imatinib in patients under long-term treatment are reported in literature. Data obtained from bioequivalence trials for healthy subjects were used to evaluate the influence of demographic and pharmacogenetic factors on imatinib pharmacokinetics (PK) in a collective without concurrent drugs, organ dysfunction, inflammation etc. In addition, the differences in PK between the healthy subjects and a patient cohort was examined to identify possible disease effects. Methods 26 volunteers were administered orally with single dose of 400 mg imatinib. 16–19 plasma samples per volunteer were collected from 0.5 up to 72 h post-dose. The popPK was built and post hoc estimates were compared with previously published PK parameters evaluated by non-compartmental analysis in the same cohort. The predictivity of the model for data collected from 40 patients with gastrointestinal stromal tumors at steady state was evaluated. Results The popPK was best described by a two-compartment transit model with first-order elimination. No significant covariates were identified, probably due to the small cohort and the narrow range of demographic covariates; CYP3A5 phenotypes appeared to have some influence on the clearance of imatinib. Good agreement between non-compartment and popPK analyses was observed with the differences of the geometric means/ median of PK estimates below 10%. The model indicated lower clearance for patients compared to healthy volunteers (p value < 0.01). Conclusion The two-compartment transit model adequately describes the absorption and distribution of imatinib in healthy volunteers. For patients, a lower clearance of imatinib compared to healthy volunteer was estimated by the model. The model can be applied for dose individualization based on trough concentrations assuming no significant differences in absorption between patients and healthy volunteers. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04454-y.
Collapse
|
45
|
Jin ZB, Wu Z, Cui YF, Liu XP, Liang HB, You JY, Wang CY. Population Pharmacokinetics and Dosing Regimen of Lithium in Chinese Patients With Bipolar Disorder. Front Pharmacol 2022; 13:913935. [PMID: 35860024 PMCID: PMC9289112 DOI: 10.3389/fphar.2022.913935] [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: 04/06/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Lithium is an effective medication approved for the treatment of bipolar disorder (BD). It has a narrow therapeutic index (TI) and requires therapeutic drug monitoring. This study aimed to conduct a population pharmacokinetics (PPK) analysis of lithium and investigate the appropriateness of the dosing regimen according to different patient characteristics. Methods: A total of 476 lithium concentrations from 268 patients with bipolar disorder were analyzed using nonlinear mixed-effects modeling. Monte Carlo simulations were employed to investigate the influence of covariates, such as weight, creatinine clearance, and daily doses of lithium concentrations, and to determine the individualized dosing regimens for patients. Results: Lithium PK was described by a one-compartment model with first-order absorption and elimination processes. The typical estimated apparent clearance was 0.909 L/h−1 with 16.4% between-subject variability in the 62 kg patients with 116 ml/min creatinine clearance and 600 mg daily doses. To achieve a target trough concentration (0.4–0.8 mmol/L) in the maintenance phase, the regimen of 500 mg than 750 mg daily dose was recommended for patients with renal insufficiency and weighing 100 kg. Conclusion: A PPK model for lithium was developed to determine the influence of patient characteristics on lithium pharmacokinetics. Weight, creatinine clearance, and total daily dose of lithium can affect the drug’s clearance. These results demonstrate the nonlinear renal excretion of lithium; hence, dosage adjustments are recommended for patients with renal insufficiency.
Collapse
Affiliation(s)
- Zi-bin Jin
- Department of Medical Psychology, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhuo Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-fan Cui
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xue-peng Liu
- Department of Medical Psychology, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hong-bo Liang
- Department of Medical Psychology, The Affiliated Xuzhou Eastern Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jia-yong You
- Department of Medical Service, Xuzhou Civil Affairs Psychiatric Hospital, Xuzhou, China
| | - Chen-yu Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Chen-yu Wang, , orcid.org/0000-0003-1808-361X
| |
Collapse
|
46
|
Wen H, Yin L, Wang J, Zhang L, Sun T, Xu F, Zhang M, Liu L, Zhang R, Liu X, Meng X, Xing Y, Lu H, Jiao Z, Zhang L. Population pharmacokinetics and model-informed precision dosing of lamivudine in Chinese HIV-infected patients with mild and moderate impaired renal function. Expert Rev Clin Pharmacol 2022; 15:647-655. [DOI: 10.1080/17512433.2022.2078306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Haini Wen
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lin Yin
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jiangrong Wang
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tao Sun
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Feng Xu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, SuZhou, Jiangsu, China
| | - Minxin Zhang
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force of PLA, Fuzhou, Fujian, China
| | - Li Liu
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Renfang Zhang
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaoqian Liu
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xianmin Meng
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yaru Xing
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lijun Zhang
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Bräm DS, Parrott N, Hutchinson L, Steiert B. Introduction of an artificial neural network–based method for concentration‐time predictions. CPT Pharmacometrics Syst Pharmacol 2022; 11:745-754. [PMID: 35582964 PMCID: PMC9197537 DOI: 10.1002/psp4.12786] [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: 09/01/2021] [Revised: 02/26/2022] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Pharmacometrics and the application of population pharmacokinetic (PK) modeling play a crucial role in clinical pharmacology. These methods, which describe data with well‐defined equations and estimate physiologically interpretable parameters, have not changed substantially during the past decades. Although the methods have proven their usefulness, they are often resource intensive and require a high level of expertise. We investigated whether a method based on artificial neural networks (ANNs) may provide an alternative approach for the prediction of concentration‐time curve to supplement the gold standard methods. In this work, we used simulated data to overcome the requirement for a large clinical training data set, implemented a pharmacologically reasonable network architecture to improve extrapolation to different dosing schemes, and used transfer learning to quickly adapt the predictions to new patient groups. We demonstrate that ANNs are able to learn the shape of concentration‐time curves and make individual predictions based on a short sequence of PK measurements. Furthermore, an ANN trained on simulated data was applied to real clinical data and was demonstrated to extrapolate to different dosing schemes. We also adapted the ANN trained on simulated healthy subjects to simulated hepatic impaired patients through transfer learning. In summary, we demonstrate how ANNs could be leveraged in a PK workflow to efficiently make individual concentration‐time predictions, and we discuss the current limitations and advantages of such an ANN‐based method.
Collapse
Affiliation(s)
- Dominic Stefan Bräm
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Basel Switzerland
- Pediatric Pharmacology and Pharmacometrics University Children's Hospital Basel Basel Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Basel Switzerland
| | - Lucy Hutchinson
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Basel Switzerland
| | - Bernhard Steiert
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Basel Basel Switzerland
| |
Collapse
|
48
|
Perlstein I, Merenlender Wagner A, Gomeni R, Lamson M, Harary E, Spiegelstein O, Kalmanczhelyi A, Tiver R, Loupe P, Levi M, Elgart A. Population Pharmacokinetic Modeling and Simulation of TV-46000: A Long-Acting Injectable Formulation of Risperidone. Clin Pharmacol Drug Dev 2022; 11:865-877. [PMID: 35245409 PMCID: PMC9315033 DOI: 10.1002/cpdd.1078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/29/2021] [Indexed: 12/05/2022]
Abstract
TV‐46000 is a long‐acting subcutaneous antipsychotic that uses a novel copolymer drug delivery technology in combination with a well‐characterized molecule, risperidone, that is in clinical development as a treatment for schizophrenia. A population pharmacokinetic (PPK) modeling and simulation approach was implemented to identify TV‐46000 doses and dosing schedules for clinical development that would provide the best balance between clinical efficacy and safety. The PPK model was created by applying pharmacokinetic data from a phase 1 study of 97 patients with a diagnosis of schizophrenia or schizoaffective disorder who received either single or repeated doses of TV‐46000. The PPK model was used to characterize the complex release profile of the total active moiety (TAM; the sum of the risperidone and 9‐OH risperidone concentrations) concentration following subcutaneous injections of TV‐46000. The PK profile was best described by a double Weibull function of the in vivo release rate and by a 2‐compartment disposition and elimination model. Simulations were performed to determine TV‐46000 doses and dosing schedules that maintained a median profile of TAM concentrations similar to published TAM exposure following oral risperidone doses that have been correlated to a 40% to 80% dopamine‐D2 receptor occupancy therapeutic window. The simulations showed that therapeutic dose ranges for TV‐46000 are 50 to 125 mg for once‐monthly and 100 to 250 mg for the once every 2 months regimens. This PPK model provided a basis for prediction of patient‐specific exposure and dopamine‐D2 receptor occupancy estimates to support further clinical development and dose selection for the phase 3 studies.
Collapse
Affiliation(s)
- Itay Perlstein
- Magic Wand Research LLC, Philadelphia, Pennsylvania, USA
| | | | | | | | - Eran Harary
- Teva Branded Pharmaceutical Products Inc. Research and Development, Netanya, Israel
| | - Ofer Spiegelstein
- Teva Branded Pharmaceutical Products Inc. Research and Development, Netanya, Israel
| | | | - Ryan Tiver
- Teva Branded Pharmaceutical Products Inc., West Chester, Pennsylvania, USA
| | - Pippa Loupe
- Teva Branded Pharmaceutical Products Inc., West Chester, Pennsylvania, USA
| | - Micha Levi
- Formerly Teva Branded Pharmaceutical Products, currently Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Anna Elgart
- Teva Branded Pharmaceutical Products Inc. Research and Development, Netanya, Israel
| |
Collapse
|
49
|
R-praziquantel integrated population pharmacokinetics in preschool- and school-aged African children infected with Schistosoma mansoni and S. haematobium and Lao adults infected with Opisthorchis viverrini. J Pharmacokinet Pharmacodyn 2022; 49:293-310. [PMID: 35024995 DOI: 10.1007/s10928-021-09791-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022]
Abstract
Racemic praziquantel (PZQ) is the standard treatment for schistosomiasis and liver fluke infections (opisthorchiasis and clonorchiasis). The development of an optimal pediatric formulation and dose selection would benefit from a population pharmacokinetic (popPK) model. A popPK model was developed for R-PZQ, the active enantiomer of PZQ, in 664 subjects, 493 African children (2-15 years) infected with Schistosoma mansoni and S. haematobium, and 171 Lao adults (15-78 years) infected with Opisthorchis viverrini. Racemate tablets were administered as single doses of 20, 40 and 60 mg/kg in children and 30, 40 and 50 mg/kg in 129 adults, and as 3 × 25 mg/kg apart in 42 adults. Samples collected by the dried-blood-spot technique were assayed by LC-MS/MS. A two-compartment disposition model, with allometric scaling and dual first-order and transit absorption, was developed using Phoenix™ software. Inversely parallel functions of age described the apparent oral bioavailability (BA) and clearance maturation in children and ageing in adults. BA decreased slightly in children with dose increase, and by 35% in adults with multiple dosing. Crushing tablets for preschool-aged children increased the first-order absorption rate by 64%. The mean transit absorption time was 70% higher in children. A popPK model for R-PZQ integrated African children over 2 years of age with schistosomiasis and Lao adults with opisthorchiasis, and should be useful to support dose optimization in children. In vitro hepatic and intestinal metabolism data would help refining and validating the model in younger children as well as in target ethnic pediatric and adult groups.
Collapse
|
50
|
Eculizumab precision dosing algorithm for thrombotic microangiopathy in children and young adults undergoing HSCT. Blood Adv 2022; 6:1454-1463. [PMID: 35008105 PMCID: PMC8905690 DOI: 10.1182/bloodadvances.2021006523] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022] Open
Abstract
This large study examined eculizumab PK/PD in bleeding and nonbleeding patients with TA-TMA. PK/PD model-based eculizumab dosing is needed for bleeding in TA-TMA, whereas fixed-dose regimens are effective in nonbleeding patients.
Transplant-associated thrombotic microangiopathy (TA-TMA) is a fatal posttransplant complication of hematopoietic stem cell transplantation. We recently reported that survival for TA-TMA has been improved by early intervention with eculizumab, a complement C5 inhibitor, guided by pharmacokinetic/pharmacodynamic (PK/PD) model-informed precision dosing. However, patients with gastrointestinal bleeding showed poor survival, even when treated with more frequent doses. Our objective was to develop separate models in bleeding and nonbleeding patients with TA-TMA and to propose precision dosing algorithms. Eculizumab PK/PD was analyzed in 19 bleeding and 38 nonbleeding patients (0.5-29.9 years of age). A complement activation biomarker (sC5b-9) and body weight were identified as significant determinants of eculizumab clearance regardless of bleeding. Eculizumab clearance after the first dose was higher in bleeding than in nonbleeding patients (83.8 vs 61.3 mL/h per 70 kg; P = .07). The high clearance was maintained over treatment doses in bleeding patients, whereas nonbleeding patients showed a time-dependent decrease in clearance. sC5b-9 levels were highest before the first dose and decreased over time, regardless of bleeding complications. A Monte Carlo Simulation analysis showed that the current dosing protocols recommended for atypical hemolytic uremic syndrome had <15% probability of attaining the target concentration of >100 μg/mL eculizumab in nonbleeding patients. We identified an intensified loading protocol to reach 80% target attainment. Our data clearly showed the need for individualized dosing for patients with significant bleeding and for ongoing dose adjustments to optimize outcomes. The developed models will be incorporated into a clinical decision guideline for precision dosing to improve outcomes in children and young adults with TA-TMA.
Collapse
|