1
|
Arsène S, Parès Y, Tixier E, Granjeon-Noriot S, Martin B, Bruezière L, Couty C, Courcelles E, Kahoul R, Pitrat J, Go N, Monteiro C, Kleine-Schultjann J, Jemai S, Pham E, Boissel JP, Kulesza A. In Silico Clinical Trials: Is It Possible? Methods Mol Biol 2024; 2716:51-99. [PMID: 37702936 DOI: 10.1007/978-1-0716-3449-3_4] [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] [Indexed: 09/14/2023]
Abstract
Modeling and simulation (M&S), including in silico (clinical) trials, helps accelerate drug research and development and reduce costs and have coined the term "model-informed drug development (MIDD)." Data-driven, inferential approaches are now becoming increasingly complemented by emerging complex physiologically and knowledge-based disease (and drug) models, but differ in setup, bottlenecks, data requirements, and applications (also reminiscent of the different scientific communities they arose from). At the same time, and within the MIDD landscape, regulators and drug developers start to embrace in silico trials as a potential tool to refine, reduce, and ultimately replace clinical trials. Effectively, silos between the historically distinct modeling approaches start to break down. Widespread adoption of in silico trials still needs more collaboration between different stakeholders and established precedence use cases in key applications, which is currently impeded by a shattered collection of tools and practices. In order to address these key challenges, efforts to establish best practice workflows need to be undertaken and new collaborative M&S tools devised, and an attempt to provide a coherent set of solutions is provided in this chapter. First, a dedicated workflow for in silico clinical trial (development) life cycle is provided, which takes up general ideas from the systems biology and quantitative systems pharmacology space and which implements specific steps toward regulatory qualification. Then, key characteristics of an in silico trial software platform implementation are given on the example of jinkō.ai (nova's end-to-end in silico clinical trial platform). Considering these enabling scientific and technological advances, future applications of in silico trials to refine, reduce, and replace clinical research are indicated, ranging from synthetic control strategies and digital twins, which overall shows promise to begin a new era of more efficient drug development.
Collapse
|
2
|
Chen Y, Wu X, Tsai C, Chang L, Yu J, Cao G, Guo B, Shi Y, Zhu D, Hu F, Yuan J, Liu Y, Zhao X, Zhang Y, Wu J, Zhang J. Integrative population pharmacokinetic/pharmacodynamic analysis of nemonoxacin capsule in Chinese patients with community-acquired pneumonia. Front Pharmacol 2023; 14:912962. [PMID: 36923351 PMCID: PMC10010492 DOI: 10.3389/fphar.2023.912962] [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: 04/21/2022] [Accepted: 02/06/2023] [Indexed: 02/28/2023] Open
Abstract
Introduction: Nemonoxacin is an innovative quinolone antibiotic for treatment of community-acquired pneumonia (CAP). As more data are available from clinical studies, it is necessary to perform an integrative pharmacokinetic/pharmacodynamic (PK/PD) analysis to support and justify the optimal dosing regimen of nemonoxacin in clinical practice. Methods and Results: We developed a population PK model using non-linear mixed effect model based on the data of 195 Chinese subjects receiving nemonoxacin in phase I to III clinical trials. The base model was a standard two-compartment PK model defined by clearance (12 L/h) and central volume of distribution (86 L). Covariates included creatinine clearance (CLcr), body weight (BW), sex, disease status and food. Compared to the subject with BW 60 kg, Cmax and A U C 0 - 24 , ss reduced by 24% and 19% in the subject with BW 80 kg, respectively. Compared to the subject with CLcr 150 ml/min, A U C 0 - 24 , ss and T1/2 increased by 28% and 24%, respectively in the subject with CLcr 30 ml/min. Compared to the fasted status, Tmax of nemonoxacin increased by 1.2 h in the subject with fed status. Effects of sex and disease status on PK parameters were small (change of PK parameters ≤19%). AUC0-24/MIC and %T > MIC were identified as the optimal PK/PD indices for predicting clinical efficacy. The AUC0-24/MIC target was 63.3, 97.8, and 115.7 against Streptococcus pneumoniae, Staphylococcus aureus, and Haemophilus influenzae, respectively. The %T > MIC target was 7.96% against Klebsiella pneumoniae. Monte Carlo simulation showed that treatment with nemonoxacin 500 mg q24 h could attain a PK/PD cutoff value higher than the MIC90 against S. pneumoniae and S. aureus. The corresponding cumulative fraction of response (CFR) was greater than 93%, while nemonoxacin 750 mg q24 h would provide higher PK/PD cutoff value against Haemophilus parainfluenzae, and higher CFR (83%) than 500 mg q24 h. Conclusion: Integrative PK/PD analysis justifies the reliable clinical and microbiological efficacy of nemonoxacin 500 mg q24 h in treating CAP caused by S. pneumoniae, S. aureus, and K. pneumoniae, irrespective of patient sex, mild renal impairment, empty stomach or not. However, nemonoxacin 750 mg q24 h would provide better efficacy than 500 mg q24 h for the CAP caused by H. parainfluenzae in terms of CFR.
Collapse
Affiliation(s)
- Yuancheng Chen
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaojie Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Chengyuan Tsai
- TaiGen Biopharmaceuticals Beijing Co., Ltd., Beijing, China
| | - Liwen Chang
- TaiGen Biopharmaceuticals Beijing Co., Ltd., Beijing, China
| | - Jicheng Yu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Guoying Cao
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Beining Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaoguo Shi
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Demei Zhu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinyi Yuan
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xu Zhao
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yingyuan Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jufang Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Phase I Unit, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Zhang Y, Sun L, Chen X, Zhao L, Wang X, Zhao Z, Mei S. A Systematic Review of Population Pharmacokinetic Models of Methotrexate. Eur J Drug Metab Pharmacokinet 2022; 47:143-164. [PMID: 34985725 DOI: 10.1007/s13318-021-00737-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND OBJECTIVES Methotrexate (MTX) is widely used for the treatment of a variety of neoplastic and autoimmune diseases. However, its toxicity and efficacy varied greatly among individuals, and they could be predicted by its pharmacokinetics. Many population pharmacokinetic models have been published to describe MTX pharmacokinetics. The objective of this systematic review was to summarize and discuss covariates with significant influence on MTX pharmacokinetics. METHODS We searched PubMed and EMBASE databases from their inception to April 2021 for population pharmacokinetic of MTX. The articles were screened by inclusion and exclusion criteria. The characteristics of studies and information for model construction and validation were extracted, summarized and discussed. RESULTS Thirty-five articles were included. The two-compartment model well described the pharmacokinetic behavior of MTX. For inter-individual variability, an exponential distribution error model was usually used for high-dose MTX population pharmacokinetic models, while a proportional distribution error model was used for low-dose MTX population pharmacokinetic models. Proportional and combined proportional and additive error models were used to describe residual error. Renal function was an independent indicator of MTX clearance. Body weight, age, gene polymorphisms (SLCO1B1, ABCC2, ABCB1, ABCG2 and MTHFR) and co-medications (proton pump inhibitors, non-steroidal anti-inflammatory drug, dexamethasone, vancomycin, penicillin and salicylic acid) could influence MTX clearance. Body weight, body surface area, age and dosage regimen have significant influence on MTX central compartment volume. Internal bootstrap test, external validation and visual predictive check were used to evaluate model predictive ability. CONCLUSIONS Various covariates could affect MTX pharmacokinetics, and their relationships have been summarized and discussed. This review will be helpful for researchers to develop their own population pharmacokinetic models and select appropriate models for individualized therapy of MTX.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, 119 Nansihuan West Road, Fengtai District, Beijing, 100070, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Liyu Sun
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, 119 Nansihuan West Road, Fengtai District, Beijing, 100070, People's Republic of China.,Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xinwei Chen
- Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, People's Republic of China
| | - Libo Zhao
- Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, People's Republic of China
| | - Xiaoling Wang
- Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.,Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, People's Republic of China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, 119 Nansihuan West Road, Fengtai District, Beijing, 100070, People's Republic of China. .,Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
| | - Shenghui Mei
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, 119 Nansihuan West Road, Fengtai District, Beijing, 100070, People's Republic of China. .,Department of Clinical Pharmacology, College of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
4
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [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] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
5
|
Kurosawa K, Rossenu S, Biewenga J, Ouwerkerk-Mahadevan S, Willems W, Ernault E, Kambili C. Population Pharmacokinetic Analysis of Bedaquiline-Clarithromycin for Dose Selection Against Pulmonary Nontuberculous Mycobacteria Based on a Phase 1, Randomized, Pharmacokinetic Study. J Clin Pharmacol 2021; 61:1344-1355. [PMID: 33991350 PMCID: PMC8518967 DOI: 10.1002/jcph.1887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/23/2021] [Indexed: 11/10/2022]
Abstract
Based on the in vitro profile of bedaquiline against mycobacterial species, it is being investigated for clinical efficacy against pulmonary nontuberculous mycobacteria (PNTM). Being a cytochrome P450 3A substrate, pharmacokinetic interactions of bedaquiline are anticipated with clarithromycin (a cytochrome P450 3A inhibitor), which is routinely used in pulmonary nontuberculous mycobacteria treatment. This phase 1, randomized, crossover study assessed the impact of steady-state clarithromycin (500 mg every 12 hours for 14 days) on the pharmacokinetics of bedaquiline and its metabolite (M2) after single-dose bedaquiline (100 mg; n = 16). Using these data, population pharmacokinetic modeling and simulation analyses were performed to determine the effect of clarithromycin on steady-state bedaquiline exposure. Although no effect was observed on maximum plasma concentration of bedaquiline and time to achieve maximum plasma concentration, its mean plasma exposure increased by 14% after 10 days of clarithromycin coadministration, with slower formation of M2. Simulations showed that bedaquiline plasma trough concentration at steady state was higher (up to 41% until week 48) with clarithromycin coadministration as compared to its monotherapy (400 mg once daily for 2 weeks, followed by 200 mg 3 times a week for 46 weeks; reference regimen). The overall exposure of a simulated bedaquiline regimen (400 mg once dialy for 2 weeks, followed by 200 mg twice a week for 46 weeks) with clarithromycin was comparable (<15% difference) to the monotherapy. Overall, combination of bedaquiline (400 mg once daily for 2 weeks, followed by 200 mg twice a week for 46 weeks) with clarithromycin seems a suitable regimen to be explored for efficacy and safety against pulmonary nontuberculous mycobacteria.
Collapse
Affiliation(s)
- Ken Kurosawa
- Department of Clinical Pharmacology, Janssen Pharmaceutical KK, Tokyo, Japan
| | | | | | | | | | | | - Chrispin Kambili
- Johnson and Johnson Services, Inc, New Brunswick, New Jersey, USA
| |
Collapse
|
6
|
Paccaly AJ, Migden MR, Papadopoulos KP, Yang F, Davis JD, Rippley RK, Lowy I, Fury MG, Stankevich E, Rischin D. Fixed Dose of Cemiplimab in Patients with Advanced Malignancies Based on Population Pharmacokinetic Analysis. Adv Ther 2021; 38:2365-2378. [PMID: 33768419 PMCID: PMC8107152 DOI: 10.1007/s12325-021-01638-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/27/2021] [Indexed: 11/28/2022]
Abstract
Introduction This study outlined cemiplimab intravenous (IV) dosing strategy to move from body weight (BW)-based 3 mg/kg every-2-week (Q2W) dosing in first-in-human study (study 1423; NCT02383212) to fixed 350 mg every-3-week (Q3W) dosing, utilizing population pharmacokinetics (PopPK) modeling and simulations, and supported by a limited dataset from a phase 2 study (study 1540; NCT02760498). Methods Cemiplimab concentration data from a total of 505 patients were pooled from study 1423 in advanced malignancies and study 1540 in advanced cutaneous squamous cell carcinoma (CSCC). All patients received weight-based cemiplimab dose (1, 3, 10 mg/kg Q2W or 3 mg/kg Q3W) except 4% who received 200 mg Q2W. A linear two-compartment PopPK model incorporating covariates that improved goodness-of-fit statistics was developed to compare cemiplimab exposure at 350 mg Q3W versus 3 mg/kg Q2W. Upon availability, observed cemiplimab concentration at 350 mg Q3W in study 1540 was then compared with the simulated values. Results Post hoc estimates of cemiplimab exposure and variability (505 patients; weight range 30.9–156 kg; median 76.1 kg) at steady state were found to be similar at 350 mg Q3W and 3 mg/kg Q2W. Effect of BW on cemiplimab exposure was described by exposure versus BW plots and at extreme BW. Overlay of individual observed cemiplimab concentrations in 51 patients with metastatic CSCC on simulated concentration–time profiles in 2000 patients at 350 mg Q3W confirmed cemiplimab exposure similarity and demonstrated the robustness of dose optimization based on PopPK modeling and simulations. Conclusions Cemiplimab 350 mg Q3W is being further investigated in multiple indications. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s12325-021-01638-5.
Collapse
Affiliation(s)
| | - Michael R Migden
- Departments of Dermatology and Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Feng Yang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Israel Lowy
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | - Danny Rischin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Guidi M, Csajka C, Buclin T. Parametric Approaches in Population Pharmacokinetics. J Clin Pharmacol 2020; 62:125-141. [DOI: 10.1002/jcph.1633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Monia Guidi
- Center for Research and Innovation in Clinical Pharmaceutical Sciences Lausanne University Hospital and University of Lausanne Lausanne Switzerland
- Service of Clinical Pharmacology Lausanne University Hospital and University of Lausanne Lausanne Switzerland
| | - Chantal Csajka
- Center for Research and Innovation in Clinical Pharmaceutical Sciences Lausanne University Hospital and University of Lausanne Lausanne Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland University of Geneva University of Lausanne Geneva Lausanne Switzerland
| | - Thierry Buclin
- Service of Clinical Pharmacology Lausanne University Hospital and University of Lausanne Lausanne Switzerland
| |
Collapse
|
8
|
Jin R, Hisaka A. Implementing PRED Subroutine of NONMEM for Versatile Pharmacokinetic Analysis Using Fast Inversion of Laplace Transform (FILT). Chem Pharm Bull (Tokyo) 2020; 68:891-894. [PMID: 32611991 DOI: 10.1248/cpb.c20-00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In pharmacokinetic (PK) analysis, conventional models are described by ordinary differential equations (ODE) that are generally solved in their Laplace transformed forms. The solution in the Laplace transformed forms is inverse Laplace transformed to derive an analytical solution. However, inverse Laplace transform is often mathematically difficult. Consequently, numerical inverse Laplace transform methods have been developed. In this study, we focus on extending the modeling functions of Nonlinear Mixed Effect Model (NONMEM), a standard software for PK and population pharmacokinetic (PPK) analyses, by adding the Fast Inversion of Laplace Transform (FILT) method, one of the representative numerical inverse Laplace transform methods. We implemented PREDFILT, a specialized PRED subroutine, which functions as an internal model unit in NONMEM to enable versatile FILT analysis with second-order precision. The calculation results of the compartment models and a dispersion model are in good agreement with the ordinary analytical solutions and theoretical values. Therefore, PREDFILT ensures enhanced flexibility in PK or PPK analyses under NONMEM environments.
Collapse
Affiliation(s)
- Ryota Jin
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
9
|
Alsultan A, Alghamdi WA, Alghamdi J, Alharbi AF, Aljutayli A, Albassam A, Almazroo O, Alqahtani S. Clinical pharmacology applications in clinical drug development and clinical care: A focus on Saudi Arabia. Saudi Pharm J 2020; 28:1217-1227. [PMID: 33132716 PMCID: PMC7584801 DOI: 10.1016/j.jsps.2020.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/14/2020] [Indexed: 01/10/2023] Open
Abstract
Drug development, from preclinical to clinical studies, is a lengthy and complex process. There is an increased interest in the Kingdom of Saudi Arabia (KSA) to promote innovation, research and local content including clinical trials (Phase I-IV). Currently, there are over 650 registered clinical trials in Saudi Arabia, and this number is expected to increase. An important part of drug development and clinical trials is to assure the safe and effective use of drugs. Clinical pharmacology plays a vital role in informed decision making during the drug development stage as it focuses on the effects of drugs in humans. Disciplines such as pharmacokinetics, pharmacodynamics and pharmacogenomics are components of clinical pharmacology. It is a growing discipline with a range of applications in all phases of drug development, including selecting optimal doses for Phase I, II and III studies, evaluating bioequivalence and biosimilar studies and designing clinical studies. Incorporating clinical pharmacology in research as well as in the requirements of regulatory agencies will improve the drug development process and accelerate the pipeline. Clinical pharmacology is also applied in direct patient care with the goal of personalizing treatment. Tools such as therapeutic drug monitoring, pharmacogenomics and model informed precision dosing are used to optimize dosing for patients at an individual level. In KSA, the science of clinical pharmacology is underutilized and we believe it is important to raise awareness and educate the scientific community and healthcare professionals in terms of its applications and potential. In this review paper, we provide an overview on the use and applications of clinical pharmacology in both drug development and clinical care.
Collapse
Affiliation(s)
- Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Wael A Alghamdi
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Jahad Alghamdi
- The Saudi Biobank, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abeer F Alharbi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | | | - Ahmed Albassam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Jin T, Liu Z, Chu Y, Ma X, Li S, Wang X, Wang G, Zhou S, Sun H, Yang J. UFLC-MS/MS Determination and Population Pharmacokinetic Study of Tanshinol, Ginsenoside Rb1 and Rg1 in Rat Plasma After Oral Administration of Compound Danshen Dripping Pills. Eur J Drug Metab Pharmacokinet 2020; 45:523-533. [PMID: 32304023 DOI: 10.1007/s13318-020-00618-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND AND OBJECTIVES As a traditional Chinese Materia Medica (CMM), the Compound Danshen Dripping Pill (CDDP) is widely used for the treatments of cardiovascular diseases. In view of its undefined applicable population and dosage, a population pharmacokinetic (PPK) study is required. The objective of this study was to explore the feasibility of multi-component CMM PPK in rat plasma after oral administration of CDDP based on sparse sampling. METHODS In this research, a simple, rapid and highly sensitive UFLC-MS/MS method for the simultaneous determination of tanshinol (TSL), ginsenoside Rb1 (GRb1) and ginsenoside Rg1 (GRg1) has been successfully developed in rat plasma. Moreover, the validated method has been applied to a PPK study of CDDP based on sparse data. We established the PPK models for these three main active constituents using a nonlinear mixed-effects model, taking into account of factors such as gender, age in weeks and weight. RESULTS The PPK models of TSL and GRb1 were best described by a one-compartment model with linear elimination and first-order absorption. The model of GRg1 was best described by a two-compartment model with first-order absorption. Bootstrap validation and a visual predictive check confirmed the predictive ability, the model stability and the precision of the parameter estimates from these models. CONCLUSION As a preliminary exploration toward the clinical population pharmacokinetic research, this study provides a reference for the population pharmacokinetic study of traditional CMM.
Collapse
Affiliation(s)
- Tianqian Jin
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China.,Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zuhui Liu
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China.,Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Chu
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Xiaohui Ma
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Shuming Li
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Xiangyang Wang
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Genbei Wang
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - Shuiping Zhou
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China
| | - He Sun
- Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, 300410, China.,State Key Laboratory of Critical Technology in Innovative Chinese Medicine, Tasly Pharmaceutical Group Co., Ltd., Tianjin, 300410, China.,Tasly Pharmaceuticals Inc, Rockville, MD, 20850, USA
| | - Jin Yang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Sassen SDT, Zwaan CM, van der Sluis IM, Mathôt RAA. Pharmacokinetics and population pharmacokinetics in pediatric oncology. Pediatr Blood Cancer 2020; 67:e28132. [PMID: 31876123 DOI: 10.1002/pbc.28132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 12/28/2022]
Abstract
Pharmacokinetic research has become increasingly important in pediatric oncology as it can have direct clinical implications and is a crucial component in individualized medicine. Population pharmacokinetics has become a popular method especially in children, due to the potential for sparse sampling, flexible sampling times, computing of heterogeneous data, and identification of variability sources. However, population pharmacokinetic reports can be complex and difficult to interpret. The aim of this article is to provide a basic explanation of population pharmacokinetics, using clinical examples from the field of pediatric oncology, to facilitate the translation of pharmacokinetic research into the daily clinic.
Collapse
Affiliation(s)
- Sebastiaan D T Sassen
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Ron A A Mathôt
- Department of Hospital Pharmacy, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Shi ZY, Liu YO, Gu HY, Xu XQ, Yan C, Yang XY, Yan D. Population pharmacokinetics of high-dose methotrexate in Chinese pediatric patients with medulloblastoma. Biopharm Drug Dispos 2020; 41:101-110. [PMID: 32017134 DOI: 10.1002/bdd.2221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 01/03/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022]
Abstract
Methotrexate (MTX) pharmacokinetics has substantial inter-individual variability and toxicity. In children with medulloblastoma treated with high-dose methotrexate (HD-MTX), the pharmacokinetic properties of methotrexate have not been established. A total of 660 serum samples from 105 pediatric patients with medulloblastoma were included in a population pharmacokinetic (PPK) analysis of methotrexate by using the nonlinear mixed-effects modeling method. The basic one-compartment population pharmacokinetic model was established by NONMEM software and the first-order conditional estimation (FOCE) method, and the final covariate model was obtained by the stepwise regression method. Weight (WT), creatinine clearance (CrCL), and whether the treatment was combined with dexamethasone (DEX) were covariates that had significant effects on the clearance rate (CL) of the model. The pharmacokinetic equation of CL in the final covariate model was as follows: CLi = 9.23× (1 + 0.0005× (θCrCL -105.78)) × (1 + 0.0017× (θWT -16)) × eηcl,i (L/h), IF (θDEX ) CLi = 1.19× CLi (L/h). The estimation accuracy of all pharmacokinetic parameters were acceptable (relative standard error < 14.74%). The goodness-of-fit diagram and bootstrap tests indicated that the final PPK model was stable with acceptable predictive ability. The PPK model may be useful for determining personalized medication levels in pediatric medulloblastoma patients undergoing HD-MTX therapy.
Collapse
Affiliation(s)
- Zheng-Yuan Shi
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China.,Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China
| | - Ya-Ou Liu
- Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China
| | - Hong-Yan Gu
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China.,Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China
| | - Xi-Qiao Xu
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China.,Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China
| | - Can Yan
- Research Centre of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xin-Yu Yang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China.,Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China
| | - Dan Yan
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China.,Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Clinical Rational Drug Use, Beijing, 100038, China
| |
Collapse
|
13
|
Liu YO, Wang ZN, Chen CY, Zhuang XH, Ruan CG, Zhou Y, Cui YM. Antiplatelet Effect of a Pulaimab [Anti-GPIIb/IIIa F(ab)2 Injection] Evaluated by a Population Pharmacokinetic-pharmacodynamic Model. Curr Drug Metab 2019; 20:1060-1072. [PMID: 31755383 DOI: 10.2174/1389200220666191122120238] [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: 08/30/2019] [Revised: 10/01/2019] [Accepted: 10/25/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cardiovascular disease has one of the highest mortality rates among all the diseases. Platelets play an important role in the pathogenesis of cardiovascular diseases. Platelet membrane glycoprotein GPIIb/IIIa antagonists are the most effective antiplatelet drugs, and pulaimab is one of these. The study aims to promote individual medication of pulaimab [anti-GPIIb/IIIa F(ab)2 injection] by discovering the pharmacological relationship among the dose, concentration, and effects. The goal of this study is to establish a population pharmacokineticpharmacodynamic model to evaluate the antiplatelet effect of intravenous pulaimab injection. METHODS Data were collected from 59 healthy subjects who participated in a Phase-I clinical trial. Plasma concentration was used as the pharmacokinetic index, and platelet aggregation inhibition rate was used as the pharmacodynamic index. The basic pharmacokinetics model was a two-compartment model, whereas the basic pharmacodynamics model was a sigmoid-EMAX model with a direct effect. The covariable model was established by a stepwise method. The final model was verified by a goodness-of-fit method, and predictive performance was assessed by a Bootstrap (BS) method. RESULTS In the final model, typical population values of the parameters were as follows: central distribution Volume (V1), 183 L; peripheral distribution Volume (V2), 349 L; Central Clearance (CL), 31 L/h; peripheral clearance(Q), 204 L/h; effect compartment concentration reaching half of the maximum effect (EC50), 0.252 mg/L; maximum effect value (EMAX), 54.0%; and shape factor (γ), 0.42. In the covariable model, thrombin time had significant effects on CL and EMAX. Verification by the goodness-of-fit and BS methods showed that the final model was stable and reliable. CONCLUSION A model was successfully established to evaluate the antiplatelet effect of intravenous pulaimab injection that could provide support for the clinical therapeutic regimen.
Collapse
Affiliation(s)
- Ya-Ou Liu
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Zi-Ning Wang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Chao-Yang Chen
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Xian-Han Zhuang
- Shanghai Asia United Antibody Medicine Limited Company, Shanghai, China
| | - Chang-Geng Ruan
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu, China
| | - Ying Zhou
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Yi-Min Cui
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| |
Collapse
|
14
|
Population pharmacokinetics of vactosertib, a new TGF-β receptor type Ι inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2019; 85:173-183. [DOI: 10.1007/s00280-019-03979-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/17/2019] [Indexed: 12/18/2022]
|
15
|
Huang L, Yang L, Huang J, Tan HY, Liu SK, Guo CX, Zuo XC, Yang GP, Pei Q. Effects of UGT1A1 Polymorphism, Gender and Triglyceride on the Pharmacokinetics of Telmisartan in Chinese Patients with Hypertension: A Population Pharmacokinetic Analysis. Eur J Drug Metab Pharmacokinet 2019; 44:797-806. [PMID: 31254178 DOI: 10.1007/s13318-019-00567-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Telmisartan is an angiotensin receptor blocker used for the treatment of hypertension. The effects of gender and uridine diphosphate-glycosytransferase 1A1 (UGT1A1) genetic polymorphisms (rs4124874, rs4148323, and rs6742078) on telmisartan plasma concentration and blood pressure in Chinese patients with hypertension have been reported previously. In this study, we aimed to develop a population pharmacokinetic (PopPK) model to quantify the effects of gender and UGT1A1 polymorphisms on the pharmacokinetics of telmisartan. METHODS Population pharmacokinetic analyses were performed using data collected prospectively from 58 Chinese patients with mild to moderate essential hypertension (aged 45-72 years; 36 men, 22 women) receiving 80 mg/day telmisartan orally for 4 weeks. Blood samples were collected in heparinized tubes at 0, 0.5, 1, and 6 h on day 28 after telmisartan administration. The plasma concentrations and UGT1A1 genetic variants were determined by high-performance liquid chromatography-mass spectrometry and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, respectively. RESULTS A two-compartment pharmacokinetic structural model with first-order elimination and absorption best described the pharmacokinetic characteristics of telmisartan. Gender and triglyceride influenced the apparent oral clearance (CL) of telmisartan. UGT1A1 (rs4124874) affected the bioavailability (F1) of telmisartan. Lower CL and bioavailability resulted in higher plasma concentrations being observed in female subjects with UGT1A1 CC or CA genotype and high triglyceride. CONCLUSION A PopPK model of telmisartan was established to confirm that UGT1A1 genotype, gender and triglyceride can affect the pharmacokinetics of telmisartan in Chinese patients with hypertension. Our findings can provide relevant pharmacokinetic parameters for further study of telmisartan.
Collapse
Affiliation(s)
- Lu Huang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Liu Yang
- Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, 410013, People's Republic of China.,Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China
| | - Jie Huang
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China
| | - Hong-Yi Tan
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China
| | - Shi-Kun Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Cheng-Xian Guo
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China
| | - Xiao-Cong Zuo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Guo-Ping Yang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China.
| | - Qi Pei
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Hunan, 410013, People's Republic of China.
| |
Collapse
|
16
|
Tylutki Z, Polak S, Wiśniowska B. Top-down, Bottom-up and Middle-out Strategies for Drug Cardiac Safety Assessment via Modeling and Simulations. CURRENT PHARMACOLOGY REPORTS 2016; 2:171-177. [PMID: 27429898 PMCID: PMC4929154 DOI: 10.1007/s40495-016-0060-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiac safety is an issue causing early terminations at various stages of drug development. Efforts are put into the elimination of false negatives as well as false positives resulting from the current testing paradigm. In silico approaches offer mathematical system and data description from the ion current, through cardiomyocytes level, up to incorporation of inter-individual variability at the population level. The article aims to review three main modelling and simulation approaches, i.e. "top-down" which refers to models built on the observed data, "bottom-up", which stands for a mechanistic description of human physiology, and "middle-out" which combines both strategies. Modelling and simulation is a well-established tool in the assessment of drug proarrhythmic potency with an impact on research and development as well as on regulatory decisions, and it is certainly here to stay. What is more, the shift to systems biology and physiology-based models makes the cardiac effect more predictable.
Collapse
Affiliation(s)
- Zofia Tylutki
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688 Cracow, Poland
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688 Cracow, Poland
- Simcyp Ltd. (part of Certara), Blades Enterprise Centre, S2 4SU Sheffield, UK
| | - Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688 Cracow, Poland
| |
Collapse
|
17
|
Clinical Pharmacokinetics of Once-Daily Tacrolimus in Solid-Organ Transplant Patients. Clin Pharmacokinet 2015; 54:993-1025. [DOI: 10.1007/s40262-015-0282-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Iversen P, Mouridsen K, Hansen MB, Jensen SB, Sørensen M, Bak LK, Waagepetersen HS, Schousboe A, Ott P, Vilstrup H, Keiding S, Gjedde A. Oxidative metabolism of astrocytes is not reduced in hepatic encephalopathy: a PET study with [(11)C]acetate in humans. Front Neurosci 2014; 8:353. [PMID: 25404890 PMCID: PMC4217371 DOI: 10.3389/fnins.2014.00353] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/14/2014] [Indexed: 12/31/2022] Open
Abstract
In patients with impaired liver function and hepatic encephalopathy (HE), consistent elevations of blood ammonia concentration suggest a crucial role in the pathogenesis of HE. Ammonia and acetate are metabolized in brain both primarily in astrocytes. Here, we used dynamic [(11)C]acetate PET of the brain to measure the contribution of astrocytes to the previously observed reduction of brain oxidative metabolism in patients with liver cirrhosis and HE, compared to patients with cirrhosis without HE, and to healthy subjects. We used a new kinetic model to estimate uptake from blood to astrocytes and astrocyte metabolism of [(11)C]acetate. No significant differences of the rate constant of oxidation of [(11)C]acetate (k 3) were found among the three groups of subjects. The net metabolic clearance of [(11)C]acetate from blood was lower in the group of patients with cirrhosis and HE than in the group of healthy subjects (P < 0.05), which we interpret to be an effect of reduced cerebral blood flow rather than a reflection of low [(11)C]acetate metabolism. We conclude that the characteristic decline of whole-brain oxidative metabolism in patients with cirrhosis with HE is not due to malfunction of oxidative metabolism in astrocytes. Thus, the observed decline of brain oxidative metabolism implicates changes of neurons and their energy turnover in patients with HE.
Collapse
Affiliation(s)
- Peter Iversen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital Aarhus, Denmark
| | - Kim Mouridsen
- Center of Functionally Integrative Neuroscience, Aarhus University Aarhus, Denmark
| | - Mikkel B Hansen
- Center of Functionally Integrative Neuroscience, Aarhus University Aarhus, Denmark
| | - Svend B Jensen
- Department of Nuclear Medicine, Aalborg University Hospital Aalborg, Denmark ; Department of Chemistry and Biochemistry, Aalborg University Aalborg, Denmark
| | - Michael Sørensen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital Aarhus, Denmark ; Department of Hepatology and Gastroenterology, Aarhus University Hospital Aarhus, Denmark
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Peter Ott
- Department of Hepatology and Gastroenterology, Aarhus University Hospital Aarhus, Denmark
| | - Hendrik Vilstrup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital Aarhus, Denmark
| | - Susanne Keiding
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital Aarhus, Denmark ; Department of Hepatology and Gastroenterology, Aarhus University Hospital Aarhus, Denmark ; Brain Research and Integrative Neuroscience Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Albert Gjedde
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital Aarhus, Denmark ; Center of Functionally Integrative Neuroscience, Aarhus University Aarhus, Denmark ; Brain Research and Integrative Neuroscience Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| |
Collapse
|
19
|
Population pharmacokinetics of imatinib in Iranian patients with chronic-phase chronic myeloid leukemia. Cancer Chemother Pharmacol 2014; 74:85-93. [PMID: 24817601 DOI: 10.1007/s00280-014-2473-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 04/23/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE We evaluated the population pharmacokinetics (PPK) and exposure-response relationship of imatinib mesylate in Iranian patients with chronic myeloid leukemia (CML).This study was designed to assess steady state (SS) imatinib trough concentrations (Cmin) and pharmacokinetics parameters of imatinib in patients with CML in chronic phase after at least 12-month treatment. METHODS Plasma concentrations from a randomized controlled trial consist of 61 patients who received oral imatinib at doses ranged between 300 and 800 mg in various dosing interval, which were quantified using a validated reversed-phase high-performance liquid chromatographic method with UV detection method on different occasions at SS and evaluated using PPK model. RESULTS A one-compartment model with zero-order absorption and a lag time was sufficient in describing the concentration-time profile. Inter-individual variability (IIV) was modeled for all parameters. Oral clearance (CL/F) and the volume of distribution (V/F) were estimated to 10.8 L/h with 30 % IIV and 265 L with 53 % IIV, respectively. Inter-occasion variability (IOV) was included in CL/F (17 %) and V/F (22 %).The proportional residual error of the model was 8 %. CONCLUSIONS Simulation analysis from individual parameters shows exposure to imatinib is highly variable among patients. Imatinib trough plasma levels <1,257 ng/mL were associated with lower rates of major molecular response. Because of the wide IIV compared with IOV with imatinib in our study, trough levels may play a role in investigating instances of suboptimal response.
Collapse
|
20
|
Influence of a priori Information, Designs, and Undetectable Data on Individual Parameters Estimation and Prediction of Hepatitis C Treatment Outcome. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2013; 2:e56. [PMID: 23863865 PMCID: PMC3731824 DOI: 10.1038/psp.2013.31] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/24/2013] [Indexed: 12/18/2022]
Abstract
Hepatitis C viral kinetic analysis based on nonlinear mixed effect models can be used to individualize treatment. For that purpose, it is necessary to obtain precise estimation of individual parameters. Here, we evaluated by simulation the influence on Bayesian individual parameter estimation and outcome prediction of a priori information on population parameters, viral load sampling designs, and methods for handling data below detection limit (BDL). We found that a precise estimation of both individual parameters and treatment outcome could be obtained using as few as six measurements in the first month of therapy. This result remained valid even when incorrect a priori information on population parameters was set as long as the parameters were identifiable and BDL data were properly handled. However, setting wrong values for a priori population parameters could lead to severe estimation/prediction errors if BDL data were ignored and not properly accounted in the likelihood function.
Collapse
|
21
|
Munekage M, Ichikawa K, Kitagawa H, Ishihara K, Uehara H, Watanabe J, Kono T, Hanazaki K. Population pharmacokinetic analysis of daikenchuto, a traditional Japanese medicine (Kampo) in Japanese and US health volunteers. Drug Metab Dispos 2013; 41:1256-63. [PMID: 23545807 DOI: 10.1124/dmd.112.050112] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We constructed population pharmacokinetic (PK) models for the five constituents of daikenchuto (DKT), a traditional Japanese herbal medicine. Data were collected from two randomized PK studies conducted in Japan and the United States. Participants received single oral doses of 2.5 g, 5 g, and 10 g of DKT. The plasma concentrations of five DKT constituents--hydroxy-α-sanshool (HAS), hydroxyl-β-sanshool (HBS), 6-shogaol (6S), 10-shogaol (10S), and ginsenoside Rb1 (GRB1)--were determined by liquid chromatography-tandem mass spectrometry. A total of 1859 samples from 55 participants (US, n = 36; Japanese, n = 19) were included in the analysis. Population PK models of HAS, HBS, 6S, and 10S were best described by a one or two-compartment model with a bolus input. On the other hand, the model of GRB1 was best described by a one-compartment model with nonlinear extravascular input. Among the covariates evaluated, body mass index (BMI) and age were found to influence oral clearance (CL/F) and volume of distribution (Vd/F) for HAS and HBS, respectively. The influence of body weight on CL/F and Vd/F for 6S was demonstrated. Marked differences were observed in mean plasma concentrations of HAS and HBS between Japanese and US participants. However, the simulation results indicated that the difference in plasma concentrations may be attributed to the difference in demographic factors such as BMI, body weight, and age, whereas ethnic difference between the Japanese and US participants was considered minimal.
Collapse
|
22
|
Zhou H. Population-Based Assessments of Clinical Drug-Drug Interactions: Qualitative Indices or Quantitative Measures? J Clin Pharmacol 2013; 46:1268-89. [PMID: 17050792 DOI: 10.1177/0091270006294278] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Population-based assessments of drug-drug interactions have become more common since the introduction and acceptance of the population pharmacokinetic approach. Unlike traditional methods, population-based studies provide clinically relevant results that can be applied directly to a target patient population. Furthermore, population-based studies do not demand the traditional requirements of intensive pharmacokinetic sampling, rigorous inpatient stays, or stringent assessment schedules. As such, the population-based approach can effectively be used to confirm known drug-drug interactions and further characterize anticipated interactions. A prospectively designed analysis can also reveal drug-drug interactions that might otherwise have gone undetected with traditional methods. Ultimately, these results could help to alleviate clinicians' concerns about using widely marketed drugs in combination therapies and also reduce patients' risk of experiencing unacceptable side effects. This article intends to provide a balanced overview of the population-based approach and its merits, drawbacks, and potential utility in the assessment of drug-drug interactions during clinical drug development.
Collapse
Affiliation(s)
- Honghui Zhou
- Pharmacokinetics, Modeling & Simulation, Clinical Pharmacology & Experimental Medicine, Centocor Research & Development, Malvern, PA 19087, USA
| |
Collapse
|
23
|
Sherwin CMT, Kiang TKL, Spigarelli MG, Ensom MHH. Fundamentals of population pharmacokinetic modelling: validation methods. Clin Pharmacokinet 2012; 51:573-90. [PMID: 22799590 DOI: 10.1007/bf03261932] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Population pharmacokinetic modelling is widely used within the field of clinical pharmacology as it helps to define the sources and correlates of pharmacokinetic variability in target patient populations and their impact upon drug disposition; and population pharmacokinetic modelling provides an estimation of drug pharmacokinetic parameters. This method's defined outcome aims to understand how participants in population pharmacokinetic studies are representative of the population as opposed to the healthy volunteers or highly selected patients in traditional pharmacokinetic studies. This review focuses on the fundamentals of population pharmacokinetic modelling and how the results are evaluated and validated. This review defines the common aspects of population pharmacokinetic modelling through a discussion of the literature describing the techniques and placing them in the appropriate context. The concept of validation, as applied to population pharmacokinetic models, is explored focusing on the lack of consensus regarding both terminology and the concept of validation itself. Population pharmacokinetic modelling is a powerful approach where pharmacokinetic variability can be identified in a target patient population receiving a pharmacological agent. Given the lack of consensus on the best approaches in model building and validation, sound fundamentals are required to ensure the selected methodology is suitable for the particular data type and/or patient population. There is a need to further standardize and establish the best approaches in modelling so that any model created can be systematically evaluated and the results relied upon.
Collapse
Affiliation(s)
- Catherine M T Sherwin
- Division of Clinical Pharmacology Clinical Trials Office, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | |
Collapse
|
24
|
Laffont CM, Concordet D. A new exact test for the evaluation of population pharmacokinetic and/or pharmacodynamic models using random projections. Pharm Res 2011; 28:1948-62. [PMID: 21491150 DOI: 10.1007/s11095-011-0422-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 03/04/2011] [Indexed: 11/24/2022]
Abstract
PURPOSE Within-subject dependency of observations has a strong impact on the evaluation of population pharmacokinetic (PK) and/or pharmacodynamic (PD) models. To our knowledge, none of the current model evaluation tools correctly address this issue. We present a new method with a global test and easy diagnostic plot which relies on the use of a random projection technique that allows the analysis of dependent data. METHODS For each subject, the vector of standardised residuals is calculated and projected onto many random directions drawn uniformly from the unit sphere. Our test compares the empirical distribution of projections with their distribution under the model. Simulation studies assess the level of the test and compare its performance with common metrics including normalised prediction distribution errors and different types of weighted residuals. An application to real data is performed. RESULTS In contrast to other evaluated methods, our test shows adequate level for all models and designs investigated, which confirms its good theoretical properties. The weakness of other methods is demonstrated and discussed. CONCLUSIONS This new test appears promising and could be used in combination with other tools to drive model evaluation in population PK/PD analyses.
Collapse
|
25
|
Jakovljević MB, Janković SM, Todorović N, Milovanović JR, Janković S. [Pharmacokinetic modelling of valproate in epileptic patients]. MEDICINSKI PREGLED 2010; 63:349-55. [PMID: 21186545 DOI: 10.2298/mpns1006349j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The aim of our study was to develop and use a population pharmacokinetic model for assessment of individual valproate clearance in children and young adults suffering from epilepsy. MATERIAL AND METHODS The analysis was performed using 52 steady-state concentrations of valproate collected from 26 epileptic patients during the routine clinical practice in our hospital. The mean values of age and total body weight were 19.92 years and 57.12 kg, respectively. NONMEM software with ADVAN 1 subroutine was used for model building and assessing the influence of different covariates. A validation set of 20 epileptic patients (one blood sample per a patient) was used to estimate predicted performances of the pharmacokinetic model. RESULTS The typical mean value of the clearance of valproate estimated by the base model in our population was 0.3 77 I/h. Out of five considered covariates (total body weight, age, total daily dose, gender and polytherapy) only the age of the patients was a significant determinant of the clearance of valproate. The final regression model for the clearance of valproate was as following: CL (l/h) = 0.223 + 0.00819 * AGE CONCLUSION: The derived pharmacokinetical model describes the clearance of valproate in relation to patient's age in the observed population. it will help to improve the seizure control in young patients with epilepsy in Serbian population.
Collapse
|
26
|
Kloft C, Poggesi I. Current and future directions of pharmacokinetic and pharmacokinetic-pharmacodynamic modelling and simulation: population approach group in Europe 19th annual meeting. Expert Opin Drug Metab Toxicol 2010; 6:1599-604. [PMID: 20969486 DOI: 10.1517/17425255.2010.529899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Population Approach Group in Europe meeting is the annual meeting of a group of scientists interested in quantitative pharmacology approaches. This meeting provides insights on new modelling approaches and applications thereof in different therapeutic areas and in different phases of drug development, in which modelling and simulation is set 'at work'. These meeting highlights mainly focus on the contributions pertinent to this journal, such as physiology-based pharmacokinetic modelling, modelling of pharmacokinetic-pharmacodynamic relationships (including binary/categorical end points) in non-clinical studies and also include presentations of junior scientists in the annual Lewis Sheiner Student Session.
Collapse
Affiliation(s)
- Charlotte Kloft
- Institute of Pharmacy, Department of Clinical Pharmacy, Martin-Luther-Universitaet Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle, Germany
| | | |
Collapse
|
27
|
Jin Y, Pollock BG, Coley K, Miller D, Marder SR, Florian J, Schneider L, Lieberman J, Kirshner M, Bies RR. Population pharmacokinetics of perphenazine in schizophrenia patients from CATIE: impact of race and smoking. J Clin Pharmacol 2010; 50:73-80. [PMID: 19843655 PMCID: PMC3648660 DOI: 10.1177/0091270009343694] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The goal of the study was to characterize population pharmacokinetics (PPK) for perphenazine in patients with schizophrenia from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE). Patients (n = 156) received 8 to 32 mg of perphenazine daily for 14 to 600 days for a total of 421 plasma concentrations measurements. Nonlinear mixed-effects modeling was used to determine PPK characteristics of perphenazine. One- and 2-compartment models with various random effect implementations and mixture distributions were evaluated. Objective function values and goodness-of-fit plots were used as model selection criteria. Age, weight, sex, race, smoking, and concomitant medications were evaluated as covariates. A 1-compartment linear model with proportional error best described the data. The population mean clearance and volume of distribution for perphenazine were 483 L/h and 18 200 L, respectively. Race and smoking status had significant impacts on perphenazine clearance estimates. In addition, the estimated population mean clearance was 48% higher in nonsmoking African Americans than in nonsmoking other races (512 L/h vs 346 L/h). Active smokers eliminated perphenazine 159 L/h faster than nonsmokers in each race. Clearances for smoking African Americans versus smokers in other races were 671 L/h versus 505 L/h, respectively.
Collapse
Affiliation(s)
- Yuyan Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jin Y, Pollock BG, Frank E, Cassano GB, Rucci P, Müller DJ, Kennedy JL, Forgione RN, Kirshner M, Kepple G, Fagiolini A, Kupfer DJ, Bies RR. Effect of age, weight, and CYP2C19 genotype on escitalopram exposure. J Clin Pharmacol 2010; 50:62-72. [PMID: 19841156 PMCID: PMC3571021 DOI: 10.1177/0091270009337946] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to characterize escitalopram population pharmacokinetics (PK) in patients treated for major depression in a cross-national, US-Italian clinical trial. Data from the 2 sites participating in this trial, conducted at Pittsburgh (United States) and Pisa (Italy), were used. Patients received 5, 10, 15, or 20 mg of escitalopram daily for a minimum of 32 weeks. Nonlinear mixed effects modeling was used to model the PK characteristics of escitalopram. One- and 2-compartment models with various random effect implementations were evaluated during model development. Objective function values and goodness-of-fit plots were used as model selection criteria. CYP2C19 genotype, age, weight, body mass index, sex, race, and clinical site were evaluated as possible covariates. In total, 320 plasma concentrations from 105 Pittsburgh patients and 153 plasma concentrations from 67 Pisa patients were available for the PK model development. A 1-compartmental model with linear elimination and proportional error best described the data. Apparent clearance (CL/F) and volume of distribution (V/F) for escitalopram without including any covariates in the patient population were 23.5 L/h and 884 L, respectively. CYP2C19 genotype, weight, and age had a significant effect on CL/F, and patient body mass index affected estimated V/F. Patients from Pisa, Italy, had significantly lower clearances than patients from Pittsburgh that disappeared after controlling for patient CYP2C19 genotype, age, and weight. Postprocessed individual empirical Bayes estimates on clearance for the 172 patients show that patients without allele CYP2C19(*)2 or (*)3 (n = 82) cleared escitalopram 33.7% faster than patients with heterogeneous or homogeneous (*)2 or (*)3 ((*)17/(*)2, (*)17/(*)3, (*)1/(*)2, (*)1/(*)3, (*)2/(*)2, (*)2/(*)3, and (*)3/(*)3, n = 46). CL/F significantly decreased with increasing patient age. Patients younger than 30 years (n = 45) cleared escitalopram 20.7% and 42.7% faster than patients aged 30 to 50 years (n = 84) and older than 50 years of age (n = 43), respectively. CYP2C19 genotype, age, and weight strongly influenced the CL/F of escitalopram. These variables may affect patient tolerance of this antidepressant and may provide important information in the effort to tailor treatments to patients' individual needs.
Collapse
Affiliation(s)
- Yuyan Jin
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Minami H, Kawada K, Sasaki Y, Tahara M, Igarashi T, Itoh K, Fujii H, Saeki T, Ozawa K, Sato H. Population pharmacokinetics of docetaxel in patients with hepatic dysfunction treated in an oncology practice. Cancer Sci 2009; 100:144-9. [PMID: 19018756 PMCID: PMC11158642 DOI: 10.1111/j.1349-7006.2009.00992.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 08/22/2008] [Accepted: 09/01/2008] [Indexed: 11/30/2022] Open
Abstract
To investigate the relationship between the degree of liver dysfunction and the pharmacokinetics of docetaxel, a population pharmacokinetic model was developed in an oncology practice without excluding patients with moderate to severe liver dysfunction. Two hundred patients were treated with docetaxel as a single agent or in combination chemotherapy. The plasma concentration-time course data were analyzed using a three-compartment open model with zero-order administration and first-order elimination on the NONMEM program. Sixty-one had elevated transaminase levels, and alkaline phosphatase was elevated in 40. Body surface area, albumin, alpha1-acid glycoprotein, and liver function were found to be significant covariates for the systemic clearance of docetaxel. Compared to patients with normal or minimal impairment of liver function, patients with grade 2 and 3 elevations of transaminases at baseline in conjunction with elevation of alkaline phosphatase had 22 and 38% lower clearances, respectively. Goodness-of-fit plots indicated that the model was fitted well with the observed data, and the bootstrap method guaranteed robustness of the model. We developed a population pharmacokinetic model for docetaxel, which can be used in the setting of an oncology practice. Based on the model, dose reduction by approximately 20 and 40% should be considered for patients with grade 2 and 3 elevations of transaminases at baseline in conjunction with elevation of alkaline phosphatase, respectively.
Collapse
Affiliation(s)
- Hironobu Minami
- Medical Oncology, Department of Medicine, Kobe University Hospital and Graduate School of Medicine, Hyogo 650-0017, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Guo F, Letrent SP, Sharma A. Population pharmacokinetics of a HER2 tyrosine kinase inhibitor CP-724,714 in patients with advanced malignant HER2 positive solid tumors. Cancer Chemother Pharmacol 2007; 60:799-809. [PMID: 17285315 DOI: 10.1007/s00280-007-0427-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 01/15/2007] [Indexed: 01/11/2023]
Abstract
PURPOSE To develop a population pharmacokinetic (popPK) model for CP-724,714, a novel HER2 tyrosine kinase inhibitor is under development for the treatment of advanced HER2 positive cancers. METHODS Concentration-time data (n = 484) from 30 cancer patients receiving daily oral CP-724,714 at doses of 250 mg QD, 250 mg BID, 250 mg TID, and 400 mg BID in 21-day cycles in an open label First-in-Human dose-escalation study were evaluated. Serum concentrations of CP-724,714 were obtained after single dose and at steady state. Nonlinear mixed effect analysis in NONMEM using first order conditional estimation with interaction (FOCEI) was performed. The effect of covariates was assessed. Diagnostic plots, decrease of objective function value (>7.8), bootstrapping, and predictive check were used as model selection criteria. RESULTS A 2-compartment first-order absorption pharmacokinetic (PK) model with inter-subject variability (ISV) on the apparent oral elimination clearance (CL/F), apparent central volume of distribution (V1/F), apparent peripheral volume of distribution (V2/F), and first-order oral absorption rate constant (ka), interoccasion variability (IOV) on CL/F, and body weight (WT) as covariate on CL/F was developed. There was no evidence of dose-dependent and/or time-dependent PK. CL/F increased with increasing WT. The ISV of CL/F was reduced by approximately 20% with WT as a covariate. Age, race, and liver metastasis did not influence CP-724,714 disposition. CONCLUSIONS A popPK model was developed that adequately described the pharmacokinetics of CP-724,714. WT was identified as a covariate on CL/F that reduced ISV and improved model performance. Future studies will further assess the importance of WT as a pharmacokinetic covariate. The proposed popPK model can be used to simulate CP-724,714 Phase 2/3 trials.
Collapse
Affiliation(s)
- Feng Guo
- Department of Clinical Pharmacology, Pfizer Global Research and Development, 50 Pequot Avenue MS6025-A3238, New London, CT 06320, USA.
| | | | | |
Collapse
|
31
|
Valenzuela B, López-Pintor E, Pérez-Ruixo JJ, Nácher A, Martín-Villodre A, Casabó VG. Modelling intestinal absorption of salbutamol sulphate in rats. Int J Pharm 2006; 314:21-30. [PMID: 16574353 DOI: 10.1016/j.ijpharm.2006.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 01/13/2006] [Indexed: 11/26/2022]
Abstract
The objective was to develop a semiphysiological population pharmacokinetic model that describes the complex salbutamol sulphate absorption in rat small intestine. In situ techniques were used to characterize the salbutamol sulphate absorption at different concentrations (range: 0.15-18 mM). Salbutamol sulphate at concentration of 0.29 mM was administered in presence of verapamil (10 and 20 mM), grapefruit juice and sodium azide (NaN3) (0.3, 3 and 6 mM). Different pharmacokinetic models were fitted to the dataset using NONMEM. Parametric and non-parametric bootstrap analyses were employed as internal model evaluation techniques. The validated model suggested instantaneous equilibrium between salbutamol sulphate concentrations in lumen and enterocyte, and the salbutamol sulphate absorption was best described by a simultaneous passive diffusion (ka = 0.636 h(-1)) and active absorption (VMax = 0.726 mM/h, Km = 0.540 mM) processes from intestinal lumen to enterocyte, together with an active capacity-limited P-gp efflux (V'max = 0.678 mM/h, K'm = 0.357 mM) from enterocyte to intestinal lumen. The extent of salbutamol sulphate absorption in rat small intestine can be improved by NaN3, grapefruit juice and verapamil.
Collapse
Affiliation(s)
- B Valenzuela
- Pharmacy and Pharmaceutics Division, Department of Engineering, Faculty of Pharmacy, Miguel Hernández University, Crta. Alicante-Valencia km. 87, 03550 San Juan de Alicante, Alicante, Spain.
| | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Jolling K, Perez Ruixo JJ, Hemeryck A, Vermeulen A, Greway T. Mixed-effects modelling of the interspecies pharmacokinetic scaling of pegylated human erythropoietin. Eur J Pharm Sci 2005; 24:465-75. [PMID: 15784336 DOI: 10.1016/j.ejps.2005.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Revised: 12/16/2004] [Accepted: 01/04/2005] [Indexed: 11/25/2022]
Abstract
The aim of this study was to develop a population pharmacokinetic model for interspecies allometric scaling of pegylated r-HuEPO (PEG-EPO) pharmacokinetics to man. A total of 927 serum concentrations from 193 rats, 6 rabbits, 34 monkeys, and 9 dogs obtained after a single dose of PEG-EPO, administered by the i.v. (dose range: 12.5-550 microg/kg) and s.c. (dose range: 12.5-500 microg/kg) routes, were pooled in this analysis. An open two-compartment model with first-order absorption and lag time (Tlag) and linear elimination from the central compartment was fitted to the data using the NONMEM V software. Body weight (WT) was used as a scaling factor and the effect of brain weight (BW), sex, and pregnancy status on the pharmacokinetic parameters was investigated. The final model was evaluated by means of a non-parametric bootstrap analysis and used to predict the PEG-EPO pharmacokinetic parameters in healthy male subjects. The systemic clearance (CL) in males was estimated to be 4.08WT1.030xBW-0.345 ml/h. In females, the CL was 90.7% of the CL in males. The volumes of the central (Vc) and the peripheral (Vp) compartment were characterized as 57.8WT0.959 ml, and 48.1WT1.150 ml, respectively. Intercompartmental flow was estimated at 2.32WT0.930 ml/h. Absorption rate constant (Ka) was estimated at 0.0538WT-0.149. The absolute s.c. bioavailability F was calculated at 52.5, 80.2, and 49.4% in rat, monkey, and dog, respectively. The interindividual variability in the population pharmacokinetic parameters was fairly low (<35%). Non-parametric bootstrap confirmed the accuracy of the NONMEM estimates. The mean model predicted pharmacokinetic parameters in healthy male subjects of 70 kg were estimated at: CL: 26.2 ml/h; Vc: 3.6l; Q: 286 l/h; Vp: 6.9l, and Ka: 0.031 h-1. The population pharmacokinetic model developed was appropriate to describe the time course of PEG-EPO serum concentrations and their variability in different species. The model predicted pharmacokinetics of PEG-EPO in humans suggest a less frequent dosing regimen relative to erythropoietin and darbepoetin, potentially leading to a simplification of anemia management.
Collapse
Affiliation(s)
- Koen Jolling
- Advanced PKPD Modeling and Simulation Department, Global Clinical Pharmacokinetics and Clinical Pharmacology, 2340 Beerse, Belgium
| | | | | | | | | |
Collapse
|
34
|
Abstract
Relaxation of the upper age limits for solid organ transplantation coupled with improvements in post-transplant survival have resulted in greater numbers of elderly patients receiving immunosuppressant drugs such as tacrolimus. Tacrolimus is a potent agent with a narrow therapeutic window and large inter- and intraindividual pharmacokinetic variability. Numerous physiological changes occur with aging that could potentially affect the pharmacokinetics of tacrolimus and, hence, patient dosage requirements. Tacrolimus is primarily metabolised by cytochrome P450 (CYP) 3A enzymes in the gut wall and liver. It is also a substrate for P-glycoprotein, which counter-transports diffused tacrolimus out of intestinal cells and back into the gut lumen. Age-associated alterations in CYP 3A and P-glycoprotein expression and/or activity, along with liver mass and body composition changes, would be expected to affect the pharmacokinetics of tacrolimus in the elderly. However, interindividual variation in these processes may mask any changes caused by aging. More investigation is needed into the impact aging has on CYP and P-glycoprotein activity and expression. No single-dose, intense blood-sampling study has specifically compared the pharmacokinetics of tacrolimus across different patient age groups. However, five population pharmacokinetic studies, one in kidney, one in bone marrow and three in liver transplant recipients, have investigated age as a co-variate. None found a significant influence for age on tacrolimus bioavailability, volume of distribution or clearance. The number of elderly patients included in each study, however, was not documented and may have been only small. It is likely that inter- and intraindividual pharmacokinetic variability associated with tacrolimus increase in elderly populations. In addition to pharmacokinetic differences, donor organ viability, multiple co-morbidity, polypharmacy and immunological changes need to be considered when using tacrolimus in the elderly. Aging is associated with decreased immunoresponsiveness, a slower body repair process and increased drug adverse effects. Elderly liver and kidney transplant recipients are more likely to develop new-onset diabetes mellitus than younger patients. Elderly transplant recipients exhibit higher mortality from infectious and cardiovascular causes than younger patients but may be less likely to develop acute rejection. Elderly kidney recipients have a higher potential for chronic allograft nephropathy, and a single rejection episode can be more devastating. There is a paucity of information on optimal tacrolimus dosage and target trough concentration in the elderly. The therapeutic window for tacrolimus concentrations may be narrower. Further integrated pharmacokinetic-pharmacodynamic studies of tacrolimus are required. It would appear reasonable, based on current knowledge, to commence tacrolimus at similar doses as those used in younger patients. Maintenance dose requirements over the longer term may be lower in the elderly, but the increased variability in kinetics and the variety of factors that impact on dosage suggest that patient care needs to be based around more frequent monitoring in this age group.
Collapse
Affiliation(s)
- Christine E Staatz
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia.
| | | |
Collapse
|
35
|
Jolling K, Ruixo JJP, Hemeryck A, Piotrovskij V, Greway T. Population pharmacokinetic analysis of pegylated human erythropoietin in rats. J Pharm Sci 2004; 93:3027-38. [PMID: 15503315 DOI: 10.1002/jps.20200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The purpose of this study was to model the pharmacokinetics of the pegylated human erythropoietin (PEG-EPO) after single-dose administration in rats, and to evaluate the influence of weight, sex, and pregnancy status on the pharmacokinetic parameters. A total of 436 serum concentrations from 193 Sprague-Dawley rats were obtained from four pharmacokinetic/toxicokinetic studies, in which a single dose of PEG-EPO was administered by the intravenous (i.v.; dose range: 2.5 to 500 microg/kg) and subcutaneous (s.c.; dose range: 12.5 to 500 microg/kg) route. Pharmacokinetic analysis was performed using nonlinear mixed effect modeling (NONMEM V software) to determine the population mean of pharmacokinetic parameters and the variances of the interindividual random effects. The effect of weight, sex, and pregnancy status on the pharmacokinetic parameters was evaluated by forward inclusion and backward elimination process, using the likelihood ratio test. Nonparametric bootstrap analysis was employed as an internal model evaluation technique to qualify the model developed. An open two-compartment model with linear elimination from the central compartment, a first-order absorption with lag time characterized the serum concentration-time profiles of PEG-EPO after i.v. and s.c. administration. For a male rat of 0.24 kg, the average CL, Vc, Q, Vp, Ka, Tlag, and F was estimated to be 0.728 mL/h, 15.8 mL, 0.373 mL/h, 6.99 mL, 0.0618 h(-1), 3.13 h, and 48.8%, respectively. A twofold increase in weight corresponded with a 170 and 238% increase in CL and Vc, respectively. In female rats, Vp was reduced by 11%, whereas F was increased by 15%. No effect of pregnancy status on any of the parameters could be identified. The interindividual variability in CL, Vc, Vp, Ka, and F was estimated at 10.7, 14.7, 16.6, 11.0, and 13.6%, respectively. Nonparametric bootstrap analysis confirmed the accuracy and the precision of the NONMEM parameter estimates. A population pharmacokinetic approach was used to integrate the knowledge gathered from several pharmacokinetic/toxicokinetic studies in rats. The pharmacokinetics of PEG-EPO in the rat was successfully modeled using a two-compartmental model with a linear elimination from the central compartment and a first-order absorption process with lag time. Weight and sex, but not pregnancy status, were identified as covariates of interest during preclinical development. The population pharmacokinetic model developed will be further used for the purpose of interspecies scaling and PK/PD modeling.
Collapse
Affiliation(s)
- Koen Jolling
- Advanced PK/PD Modeling and Simulation department, Global Clinical Pharmacokinetics and Clinical Pharmacology, Johnson & Johnson Pharmaceutical Research & Development (a division of Janssen Pharmaceutica NV), 2340 Beerse, Belgium
| | | | | | | | | |
Collapse
|
36
|
Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clin Pharmacokinet 2004; 43:623-53. [PMID: 15244495 DOI: 10.2165/00003088-200443100-00001] [Citation(s) in RCA: 629] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aim of this review is to analyse critically the recent literature on the clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplant recipients. Dosage and target concentration recommendations for tacrolimus vary from centre to centre, and large pharmacokinetic variability makes it difficult to predict what concentration will be achieved with a particular dose or dosage change. Therapeutic ranges have not been based on statistical approaches. The majority of pharmacokinetic studies have involved intense blood sampling in small homogeneous groups in the immediate post-transplant period. Most have used nonspecific immunoassays and provide little information on pharmacokinetic variability. Demographic investigations seeking correlations between pharmacokinetic parameters and patient factors have generally looked at one covariate at a time and have involved small patient numbers. Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Population analyses are adding to our understanding of the pharmacokinetics of tacrolimus, but such investigations are still in their infancy. A significant proportion of model variability remains unexplained. Population modelling and Bayesian forecasting may be improved if CYP isoenzymes and/or P-glycoprotein expression could be considered as covariates. Reports have been conflicting as to whether low tacrolimus trough concentrations are related to rejection. Several studies have demonstrated a correlation between high trough concentrations and toxicity, particularly nephrotoxicity. The best predictor of pharmacological effect may be drug concentrations in the transplanted organ itself. Researchers have started to question current reliance on trough measurement during therapeutic drug monitoring, with instances of toxicity and rejection occurring when trough concentrations are within 'acceptable' ranges. The correlation between blood concentration and drug exposure can be improved by use of non-trough timepoints. However, controversy exists as to whether this will provide any great benefit, given the added complexity in monitoring. Investigators are now attempting to quantify the pharmacological effects of tacrolimus on immune cells through assays that measure in vivo calcineurin inhibition and markers of immunosuppression such as cytokine concentration. To date, no studies have correlated pharmacodynamic marker assay results with immunosuppressive efficacy, as determined by allograft outcome, or investigated the relationship between calcineurin inhibition and drug adverse effects. Little is known about the magnitude of the pharmacodynamic variability of tacrolimus.
Collapse
Affiliation(s)
- Christine E Staatz
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | | |
Collapse
|
37
|
Chen X, Bies RR, Ramanathan RK, Zuhowski EG, Trump DL, Egorin MJ. Population pharmacokinetic analysis of 17-(allylamino)-17-demethoxygeldanamycin (17AAG) in adult patients with advanced malignancies. Cancer Chemother Pharmacol 2004; 55:237-43. [PMID: 15503027 DOI: 10.1007/s00280-004-0836-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2003] [Accepted: 04/07/2004] [Indexed: 01/11/2023]
Abstract
PURPOSE 17-(Allylamino)-17-demethoxygeldanamycin (17AAG) is a novel anticancer agent in clinical development. The objectives of this study were to develop a population pharmacokinetic model for 17AAG and its major metabolite, 17AG, and to investigate influences of patient characteristics and biochemical markers on pharmacokinetic parameters estimated for 17AAG and 17AG. EXPERIMENTAL DESIGN In a phase I clinical study, 17AAG was administered by intravenous infusion to 43 patients with refractory, advanced malignancies. Plasma concentrations of 17AAG and 17AG were determined by high-performance liquid chromatography. Plasma concentration vs time data were modeled using NONMEM. Nine covariates (age, sex, performance status, weight, height, body surface area, AST, bilirubin and serum creatinine) were investigated for their influences on individual pharmacokinetic parameters. RESULTS Plasma concentration vs time data were best described by a two-compartment model for 17AAG and a one-compartment model for 17AG. Volumes of distribution were 24.2 and 89.6 l for 17AAG. Total elimination clearances were 26.7 and 21.3 l/h for 17AAG and 17AG, respectively. Both fixed and random effects pharmacokinetic parameters were well estimated. None of the covariates explained the interindividual variability in 17AAG and 17AG pharmacokinetic parameters or improved the fit of the model based on objective function changes. CONCLUSIONS A population pharmacokinetic model was developed to describe 17AAG and 17AG population pharmacokinetic parameters and interindividual variabilities. There were substantial interindividual variabilities in 17AAG and 17AG pharmacokinetic parameters despite BSA-normalized dosing.
Collapse
Affiliation(s)
- Xueyu Chen
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Room 5-221A, 610 University Ave., Toronto, ON, Canada, M5G 2M9.
| | | | | | | | | | | |
Collapse
|
38
|
Tatami S, Yamamura N, Sarashina A, Yong CL, Igarashi T, Tanigawara Y. Pharmacokinetic Comparison of an Angiotensin II Receptor Antagonist, Telmisartan, in Japanese and Western Hypertensive Patients Using Population Pharmacokinetic Method. Drug Metab Pharmacokinet 2004; 19:15-23. [PMID: 15499165 DOI: 10.2133/dmpk.19.15] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objectives of this study were to develop a population pharmacokinetic (PPK) model for telmisartan based on the pooled data obtained from the different racial populations and then to identify the factors that affect the pharmacokinetics of telmisartan for the comparison between the regions. A PPK model was established based on the data of 1343 subjects in 12 clinical trials. The PK profiles of telmisartan were described with a 2-compartment model with first-order absorption. The obtained model could predict the observed plasma concentrations well. This PPK model suggested that CL/F was a function of age, dose, gender, race, alcohol consumption and liver function. A marked difference was observed in the plasma concentration profiles between Japanese and other countries' subjects. However, the effect of the factor "race" on CL/F was not large. In the present PPK model, "trial condition" affected all PK parameters except for V(2)/F. The condition differences were in food condition and formulation (Japanese: fed, capsule, US and EU: fasted, tablet). The extent of difference in the plasma concentration profiles simulated for Japanese and Caucasian using the PPK model under the same demographic condition was comparable with the results of the food effect study performed previously in Japan. The findings suggest that the difference in the plasma concentration profiles between Japanese and other countries' subjects was mainly due to the difference of food intake conditions under which the clinical trials were performed.
Collapse
Affiliation(s)
- Shinji Tatami
- Department of Drug Metabolism and Pharmacokinetics, Kawanishi Pharma Research Institute, Nippon Boehringer Ingelheim Co., Ltd., Kawanishi, Japan.
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
The aim of the current review is to summarise the present status of physiologically based pharmacokinetic (PBPK) modelling and its applications in drug research, and thus serve as a reference point to people interested in the methodology. The review is structured into three major sections. The first discusses the existing methodologies and techniques of PBPK model development. The second describes some of the most interesting PBPK model implementations published. The final section is devoted to a discussion of the current limitations and the possible future developments of the PBPK modelling approach. The current review is focused on papers dealing with the pharmacokinetics and/or toxicokinetics of medicinal compounds; references discussing PBPK models of environmental compounds are mentioned only if they represent considerable methodological developments or reveal interesting interpretations and/or applications.The major conclusion of the review is that, despite its significant potential, PBPK modelling has not seen the development and implementation it deserves, especially in the drug discovery, research and development processes. The main reason for this is that the successful development and implementation of a PBPK model is seen to require the investment of significant experience, effort, time and resources. Yet, a substantial body of PBPK-related research has been accumulated that can facilitate the PBPK modelling and implementation process. What is probably lagging behind is the expertise component, where the demand for appropriately qualified staff far outreaches availability.
Collapse
Affiliation(s)
- Ivan Nestorov
- Pharmacokinetics and Drug Metabolism, Amgen Inc., 30-O-B, One Amgen Center Drive, Thousand Oaks, CA 91320-1789, USA.
| |
Collapse
|
40
|
Aronson JK. Features of this month's Journal. Br J Clin Pharmacol 2003. [DOI: 10.1111/j.1365-2125.2003.1931a.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
41
|
van Kesteren C, Mathôt RAA, Beijnen JH, Schellens JHM. Pharmacokinetic-pharmacodynamic guided trial design in oncology. Invest New Drugs 2003; 21:225-41. [PMID: 12889741 DOI: 10.1023/a:1023577514605] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The application of pharmacokinetic (PK) and pharmacodynamic (PD) modeling in drug development has emerged during the past decades and it is has been suggested that the investigation of PK-PD relationships during drug development may facilitate and optimize the design of subsequent clinical development. Especially in oncology, well designed PK-PD modeling could be extremely useful as anticancer agents usually have a very narrow therapeutic index. This paper describes the application of the current insights in the use of PK-PD modeling to the design of clinical trials in oncology. The application of PK-PD modeling in each separate stage of (pre)clinical drug development of anticancer agents is discussed. The implementation of this approach is illustrated with the clinical development of docetaxel.
Collapse
Affiliation(s)
- Ch van Kesteren
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervnaart Hospital, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Tatami S, Sarashina A, Yamamura N, Igarashi T, Tanigawara Y. Population Pharmacokinetics of an Angiotensin II Receptor Antagonist, Telmisartan, in Healthy Volunteers and Hypertensive Patients. Drug Metab Pharmacokinet 2003; 18:203-11. [PMID: 15618736 DOI: 10.2133/dmpk.18.203] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To describe the factors affecting pharmacokinetics of telmisartan, an angiotensin II receptor antagonist, a population pharmacokinetic (PPK) model has been developed based upon the data collected from healthy volunteers and hypertensive patients. METHODS A total of 1566 plasma samples were collected from 20 healthy volunteers and 129 hypertensive patients, together with the demographic background. The data were analyzed by the NONMEM program using two-compartment model with first-order absorption. The robustness of the obtained PPK model was validated by the bootstrapping resampling method. RESULTS The oral clearance (CL/F) was found to be associated with age, dose and alcohol consumption, but neither related to serum creatinine nor smoking history. The volume of distribution for the central compartment was related to age and dose, and the volume of distribution for the peripheral compartment was related to body weight and gender. The absorption rate constant (Ka) and the absorption lag time were described as function of dose. The CL/F decreased with advanced age. The CL/F decreased and Ka increased with higher dose, reflecting the super-proportional increase in the plasma levels of telmisartan. The AUC and C(max) values predicted by the present PPK model were well consistent with the observed values. The means of parameter estimates obtained with 200 bootstrap replicates were within 95-111% of the final parameter estimates from the original data set. CONCLUSION A PPK model for telmisartan developed here well described the individual variability and exposure, and robustness of the model has been validated by the bootstrapping method.
Collapse
Affiliation(s)
- Shinji Tatami
- Department of Drug Metabolism and Pharmacokinetics, Kawanishi Pharma Research Institute, Nippon Boehringer Ingelheim Co., Ltd., Japan.
| | | | | | | | | |
Collapse
|
43
|
Van Kesteren C, Mathôt RAA, Raymond E, Armand JP, Dittrich C, Dumez H, Roché H, Droz JP, Punt C, Ravic M, Wanders J, Beijnen JH, Fumoleau P, Schellens JHM. Population pharmacokinetics of the novel anticancer agent E7070 during four phase I studies: model building and validation. J Clin Oncol 2002; 20:4065-73. [PMID: 12351604 DOI: 10.1200/jco.2002.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE N-(3-Chloro-7-indolyl)-1,4-benzenedisulfonamide (E7070) is a novel sulfonamide anticancer agent currently in phase II clinical development for the treatment of solid tumors. Four phase I studies have been finalized, with E7070 administered at four different treatment schedules to identify the maximum-tolerated dose and the dose-limiting toxicities. Pharmacokinetic analyses of all studies revealed E7070 to have nonlinear pharmacokinetics. A population pharmacokinetic model was designed and validated to describe the pharmacokinetics of E7070 at all four treatment schedules and to identify the possible influences of patient characteristics on the pharmacokinetic parameters. PATIENTS AND METHODS Plasma concentration-time data of all patients (n = 143) were fitted to several pharmacokinetic models using NONMEM. Seventeen covariables were investigated for their relation with individual pharmacokinetic parameters. A bootstrap procedure was performed to check the validity of the model. RESULTS The data were best described using a three-compartment model with nonlinear distribution to a peripheral compartment and two parallel pathways of elimination from the central compartment: a linear and a saturable pathway. Body-surface area (BSA) was significantly correlated to both the volume of distribution of the central compartment and to the maximal elimination capacity. The fits of 500 bootstrap replicates of the data set demonstrated the robustness of the developed population pharmacokinetic model. CONCLUSION A population pharmacokinetic model has been designed and validated that accurately describes the data of four phase I studies with E7070. Furthermore, it has been demonstrated that BSA-guided dosing for E7070 is important.
Collapse
Affiliation(s)
- Ch Van Kesteren
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|