1
|
Chen J, Wang Q, Li S, Han R, Wang C, Cheng S, Yang B, Diao L, Yang T, Sun D, Zhang D, Dong Y, Wang T. Does Two-Step Infusion Improve the Pharmacokinetics/Pharmacodynamics Target Attainment of Meropenem in Critically Ill Patients? J Pharm Sci 2024; 113:2904-2914. [PMID: 38996917 DOI: 10.1016/j.xphs.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
The optimal method for administering meropenem remains controversial. This study was conducted to explore the optimal two-step infusion strategy (TIT), and to investigate whether TIT is superior to intermittent infusion therapy (IIT) and prolonged infusion therapy (PIT). A physiologically based pharmacokinetics model for critically ill patients was established and evaluated. The validated model was utilized to evaluate the pharmacokinetics/pharmacodynamics (PK/PD) target attainment of meropenem. The PK/PD target attainment of different TITs varied greatly, and the total infusion duration and the first-step dose greatly affected these values. The optimal TIT was 0.25 g (30 min) + 0.75 g (150 min) at MICs of ≤2 mg/L, and 0.25 g (45 min) + 0.75 g (255 min) at MICs of 4-8 mg/L. The PK/PD target attainment of optimal TIT, PIT, and IIT were 100 % at MICs of ≤1 mg/L. When MIC increased to 2-8 mg/L, the PK/PD target attainment of optimal TIT was similar to that of PIT and higher than IIT. In conclusion, TIT did not significantly improve the PK/PD target attainment of meropenem compared with PIT. IIT is adequate at MICs of ≤1 mg/L, and PIT may be the optimal meropenem infusion method in critically ill patients with MICs of 2-8 mg/L.
Collapse
Affiliation(s)
- Jiaojiao Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Quanfang Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Sihan Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ruiying Han
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, Xi'an 710021, China
| | - Chuhui Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shiqi Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Baogui Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Lizhuo Diao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Tingting Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Dan Sun
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, Xi'an 710021, China
| | - Di Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Taotao Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
2
|
Martischang R, Nikolaou A, Daali Y, Samer CF, Terrier J. Guidance on Selecting Optimal Steady-State Tacrolimus Concentrations for Continuous IV Perfusion: Insights from Physiologically Based Pharmacokinetic Modeling. Pharmaceuticals (Basel) 2024; 17:1047. [PMID: 39204152 PMCID: PMC11357179 DOI: 10.3390/ph17081047] [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: 07/01/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Introduction: The dose-response relationships of tacrolimus have been primarily assessed through trough concentrations during intermittent administrations. In scenarios where oral administration (PO) is unfeasible, continuous intravenous (IV) administration is advised. Under these circumstances, only steady-state (Css) plasma or blood concentrations are measured, with the absence of distinct trough levels (Cmin). Consequently, the measured concentrations are frequently misinterpreted as trough concentrations, potentially resulting in sub-therapeutic true tacrolimus blood levels. This study employs physiologically based pharmacokinetic modeling (PBPK) to establish the Css/Cmin ratio for tacrolimus across various clinical scenarios. Method: Using a validated PBPK model, the tacrolimus dose (both PO and IV) and the Css/Cmin ratios corresponding to matching area under the blood concentration-time curve during a dosage interval (AUCτ) values were estimated under different conditions, including healthy subjects and individuals exhibiting cytochrome P450 3A (CYP3A) interactions or CYP3A5 polymorphisms, along with a demonstration of a real-life clinical application. Result: In healthy volunteers, the oral/intravenous (PO/IV) dose ratio was found to be 4.25, and the Css/Cmin ratio was 1.40. A specific clinical case substantiated the practical applicability of the Css/Cmin ratio as simulated by PBPK, demonstrating no immediate clinical complications related to the transplant. When considering liver donors versus recipients expressing CYP3A5, the tacrolimus AUCτ was notably affected, yielding a PO/IV dose ratio of 4.00 and a Css/Cmin ratio of 1.75. Furthermore, the concomitant administration of the CYP3A inhibitor itraconazole given PO resulted in a PO/IV ratio of 1.75 with and a Css/Cmin ratio of 1.28. Notably, the inhibitory effect of itraconazole was diminished when administered IV. Conclusions: Through the application of PBPK methodologies, this study estimates the PO/IV dose ratios and Css/Cmin ratios that can enhance dose adjustment and therapeutic drug monitoring during the switch between IV and PO administration of tacrolimus in transplant patients, ultimately guiding clinicians in real-time decision-making. Further validation with in vivo data is recommended to support these findings.
Collapse
Affiliation(s)
- Romain Martischang
- Division of General Internal Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Argyro Nikolaou
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
- Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Caroline Flora Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
- Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Jean Terrier
- Division of General Internal Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
3
|
Zang YN, Wan Z, Jia F, Yang Q, Liu CG, Wang Q, Liu SS, Dong F, Li AN, de Leon J, Wang G, Ruan CJ. Population pharmacokinetics of olanzapine in pediatric patients with psychiatric disorders. Expert Opin Drug Metab Toxicol 2024; 20:827-840. [PMID: 39010781 DOI: 10.1080/17425255.2024.2380472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE To develop and validate a population pharmacokinetic (PPK) model of oral olanzapine in pediatric Chinese patients in order to individualize therapy in this population. METHODS A total of 897 serum concentrations from 269 pediatric patients taking oral olanzapine (ages 8-17 years) were collected. Demographic parameters, biological characteristics and concomitant medications were investigated as covariates. The data were analyzed using a nonlinear mixed-effects modeling approach. Bootstrapping (1000 runs), normalized prediction distribution error (NPDE), and external validation of 62 patients were employed. Simulations were performed to explore the individualized dosing regimens in various situations. RESULTS The one-compartment model with first-order absorption and elimination had an apparent clearance (CL/F) of 10.38 L/h, a distribution volume (V/F) of 9.41 L/kg and an absorption rate constant (Ka) fixed at 0.3 h-1. The equation was CL∕F (L∕h) = 10.38 × (body weight∕60)0.25 ×1.33 (if male) × 0.71 (if co-occurrence of infection) × 0.51 (if co-therapy with fluvoxamine) × 1.27 (if co-therapy with sertraline) × 1.43 (if co-therapy with valproate). The final model had satisfactory stability, robustness, and predictive ability. The results from a simulation suggested the oral olanzapine doses required for male and female pediatric patients weighing between 40 and 60 kg without co-medication were 10-15 mg/day and 7.5-10 mg/day, respectively, and dosage adjustments should be based on sex and body weight; and co-administrated with valproate, sertraline, or fluvoxamine. CONCLUSION This model may help individualize optimum dosing of oral olanzapine for pediatric patients.
Collapse
Affiliation(s)
- Yan-Nan Zang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhou Wan
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fei Jia
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qi Yang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Chen-Geng Liu
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qian Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Shan-Shan Liu
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fang Dong
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - An-Ning Li
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jose de Leon
- Mental Health Research Center at Eastern State Hospital, Lexington, KY, USA
- Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apóstol Hospital, University of the Basque Country, Vitoria, Spain
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Can-Jun Ruan
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Mostafa S, Rafizadeh R, Polasek TM, Bousman CA, Rostami‐Hodjegan A, Stowe R, Carrion P, Sheffield LJ, Kirkpatrick CMJ. Virtual twins for model-informed precision dosing of clozapine in patients with treatment-resistant schizophrenia. CPT Pharmacometrics Syst Pharmacol 2024; 13:424-436. [PMID: 38243630 PMCID: PMC10941576 DOI: 10.1002/psp4.13093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/14/2023] [Accepted: 11/02/2023] [Indexed: 01/21/2024] Open
Abstract
Model-informed precision dosing using virtual twins (MIPD-VTs) is an emerging strategy to predict target drug concentrations in clinical practice. Using a high virtualization MIPD-VT approach (Simcyp version 21), we predicted the steady-state clozapine concentration and clozapine dosage range to achieve a target concentration of 350 to 600 ng/mL in hospitalized patients with treatment-resistant schizophrenia (N = 11). We confirmed that high virtualization MIPD-VT can reasonably predict clozapine concentrations in individual patients with a coefficient of determination (R2 ) ranging between 0.29 and 0.60. Importantly, our approach predicted the final dosage range to achieve the desired target clozapine concentrations in 73% of patients. In two thirds of patients treated with fluvoxamine augmentation, steady-state clozapine concentrations were overpredicted two to four-fold. This work supports the application of a high virtualization MIPD-VT approach to inform the titration of clozapine doses in clinical practice. However, refinement is required to improve the prediction of pharmacokinetic drug-drug interactions, particularly with fluvoxamine augmentation.
Collapse
Affiliation(s)
- Sam Mostafa
- Centre for Medicine Use and SafetyMonash UniversityParkvilleVictoriaAustralia
- MyDNA Life Australia LimitedVictoriaAustralia
| | - Reza Rafizadeh
- BC Mental Health and Substance Use Services, BC Psychosis ProgramLower Mainland Pharmacy ServicesVancouverBritish ColumbiaCanada
| | - Thomas M. Polasek
- Centre for Medicine Use and SafetyMonash UniversityParkvilleVictoriaAustralia
- CertaraPrincetonNew JerseyUSA
- Department of Clinical PharmacologyRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Chad A. Bousman
- Department of Psychiatry, Melbourne Neuropsychiatry CentreUniversity of Melbourne and Melbourne HealthMelbourneVictoriaAustralia
- Alberta Children's Hospital Research Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Departments of Medical Genetics, Psychiatry, Physiology and Pharmacology, and Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Health SciencesUniversity of ManchesterManchesterUK
- Simcyp DivisionCertara UK LimitedSheffieldUK
| | - Robert Stowe
- Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Djavid Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Neurology (Medicine)University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Prescilla Carrion
- Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | | |
Collapse
|
5
|
Terrier J, Gaspar F, Gosselin P, Raboud O, Lenoir C, Rollason V, Csajka C, Samer C, Fontana P, Daali Y, Reny J. Apixaban and rivaroxaban's physiologically-based pharmacokinetic model validation in hospitalized patients: A first step for larger use of a priori modeling approach at bed side. CPT Pharmacometrics Syst Pharmacol 2023; 12:1872-1883. [PMID: 37794718 PMCID: PMC10725260 DOI: 10.1002/psp4.13036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/21/2023] [Accepted: 08/14/2023] [Indexed: 10/06/2023] Open
Abstract
When used in real-world conditions, substantial interindividual variations in direct oral anticoagulant (DOAC) plasma concentrations are observed for a given dose, leading to a risk of over- or under-exposure and clinically significant adverse events. Physiologically-based pharmacokinetic (PBPK) models could help physicians to tailor DOAC prescriptions in vulnerable patient populations, such as those in the hospital setting. The present study aims to validate prospectively PBPK models for rivaroxaban and apixaban in a large cohort of elderly, polymorbid, and hospitalized patients. In using a model of geriatric population integrating appropriate physiological parameters into models first optimized with healthy volunteer data, observed plasma concentration collected in hospitalized patients on apixaban (n = 100) and rivaroxaban (n = 100) were adequately predicted (ratio predicted/observed area under the concentration curve for a dosing interval [AUCtau ] = 0.97 [0.96-0.99] geometric mean, 90% confidence interval, ratio predicted/observed AUCtau = 1.03 [1.02-1.05]) for apixaban and rivaroxaban, respectively. Validation of the present PBPK models for rivaroxaban and apixaban in in-patients represent an additional step toward the feasibility of bedside use.
Collapse
Affiliation(s)
- Jean Terrier
- Division of General Internal MedicineGeneva University HospitalsGenevaSwitzerland
- Geneva Platelet Group, Faculty of MedicineUniversity of GenevaGenevaSwitzerland
- Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care DepartmentGeneva University HospitalsGenevaSwitzerland
| | - Frédéric Gaspar
- Center for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
- Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of Geneva, University of LausanneGeneva, LausanneSwitzerland
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Pauline Gosselin
- Division of General Internal MedicineGeneva University HospitalsGenevaSwitzerland
| | - Olivier Raboud
- Center for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
- Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of Geneva, University of LausanneGeneva, LausanneSwitzerland
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Camille Lenoir
- Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care DepartmentGeneva University HospitalsGenevaSwitzerland
| | - Victoria Rollason
- Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care DepartmentGeneva University HospitalsGenevaSwitzerland
| | - Chantal Csajka
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
- Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of Geneva, University of LausanneGeneva, LausanneSwitzerland
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Caroline Samer
- Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care DepartmentGeneva University HospitalsGenevaSwitzerland
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of MedicineUniversity of GenevaGenevaSwitzerland
- Division of Angiology and HaemostasisGeneva University HospitalsGenevaSwitzerland
| | - Youssef Daali
- Geneva Platelet Group, Faculty of MedicineUniversity of GenevaGenevaSwitzerland
- Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care DepartmentGeneva University HospitalsGenevaSwitzerland
| | - Jean‐Luc Reny
- Division of General Internal MedicineGeneva University HospitalsGenevaSwitzerland
- Geneva Platelet Group, Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | | |
Collapse
|
6
|
Wyszogrodzka-Gaweł G, Shuklinova O, Lisowski B, Wiśniowska B, Polak S. 3D printing combined with biopredictive dissolution and PBPK/PD modeling optimization and personalization of pharmacotherapy: Are we there yet? Drug Discov Today 2023; 28:103731. [PMID: 37541422 DOI: 10.1016/j.drudis.2023.103731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Precision medicine requires selecting the appropriate dosage regimen for a patient using the right drug, at the right time. Model-Informed Precision Dosing (MIPD) is a concept suggesting utilization of model-based prediction methods for optimizing the treatment benefit-harm balance, based on individual characteristics of the patient, disease, treatment method, and other factors. Here, we discuss a theoretical workflow comprising several elements, beginning from the physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) models, through 3D printed tablets with the model proposed dose, information range and flow, and the patient themselves. We also describe each of these elements, and the connection between them, highlighting challenges and potential obstacles.
Collapse
Affiliation(s)
- Gabriela Wyszogrodzka-Gaweł
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Olha Shuklinova
- Chair of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy. Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Bartek Lisowski
- Chair of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy. Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Barbara Wiśniowska
- Chair of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy. Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Sebastian Polak
- Chair of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy. Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
7
|
Polasek TM. Virtual twin for healthcare management. Front Digit Health 2023; 5:1246659. [PMID: 37781454 PMCID: PMC10540783 DOI: 10.3389/fdgth.2023.1246659] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Healthcare is increasingly fragmented, resulting in escalating costs, patient dissatisfaction, and sometimes adverse clinical outcomes. Strategies to decrease healthcare fragmentation are therefore attractive from payer and patient perspectives. In this commentary, a patient-centered smart phone application called Virtual Twin for Healthcare Management (VTHM) is proposed, including its organizational layout, basic functionality, and potential clinical applications. The platform features a virtual twin hub that displays the body and its health data. This is a physiologically based human model that is "virtualized" for the patient based on their unique genetic, molecular, physiological, and disease characteristics. The spokes of the system are a full service and interoperable electronic-health record, accessible to healthcare providers with permission on any device with internet access. Theoretical case studies based on real scenarios are presented to show how VTHM could potentially improve patient care and clinical efficiency. Challenges that must be overcome to turn VTHM into reality are also briefly outlined. Notably, the VTHM platform is designed to operationalize current and future precision medicine initiatives, such as access to molecular diagnostic results, pharmacogenomics-guided prescribing, and model-informed precision dosing.
Collapse
Affiliation(s)
- Thomas M. Polasek
- Certara, Princeton, NJ, United States
- Centre for Medicines Use and Safety, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Ansaar R, Meech R, Rowland A. A Physiologically Based Pharmacokinetic Model to Predict Determinants of Variability in Epirubicin Exposure and Tissue Distribution. Pharmaceutics 2023; 15:pharmaceutics15041222. [PMID: 37111707 PMCID: PMC10143085 DOI: 10.3390/pharmaceutics15041222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Epirubicin is an anthracycline antineoplastic drug that is primarily used in combination therapies for the treatment of breast, gastric, lung and ovarian cancers and lymphomas. Epirubicin is administered intravenously (IV) over 3 to 5 min once every 21 days with dosing based on body surface area (BSA; mg/m2). Despite accounting for BSA, marked inter-subject variability in circulating epirubicin plasma concentration has been reported. METHODS In vitro experiments were conducted to determine the kinetics of epirubicin glucuronidation by human liver microsomes in the presence and absence of validated UGT2B7 inhibitors. A full physiologically based pharmacokinetic model was built and validated using Simcyp® (version 19.1, Certara, Princeton, NJ, USA). The model was used to simulate epirubicin exposure in 2000 Sim-Cancer subjects over 158 h following a single intravenous dose of epirubicin. A multivariable linear regression model was built using simulated demographic and enzyme abundance data to determine the key drivers of variability in systemic epirubicin exposure. RESULTS Multivariable linear regression modelling demonstrated that variability in simulated systemic epirubicin exposure following intravenous injection was primarily driven by differences in hepatic and renal UGT2B7 expression, plasma albumin concentration, age, BSA, GFR, haematocrit and sex. By accounting for these factors, it was possible to explain 87% of the variability in epirubicin in a simulated cohort of 2000 oncology patients. CONCLUSIONS The present study describes the development and evaluation of a full-body PBPK model to assess systemic and individual organ exposure to epirubicin. Variability in epirubicin exposure was primarily driven by hepatic and renal UGT2B7 expression, plasma albumin concentration, age, BSA, GFR, haematocrit and sex.
Collapse
Affiliation(s)
- Radwan Ansaar
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Robyn Meech
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| |
Collapse
|
9
|
Moreira FDL, Benzi JRDL, Pinto L, Thomaz MDL, Duarte G, Lanchote VL. Optimizing Therapeutic Drug Monitoring in Pregnant Women: A Critical Literature Review. Ther Drug Monit 2023; 45:159-172. [PMID: 36127797 DOI: 10.1097/ftd.0000000000001039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/18/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND More than 90% of pregnant women take at least one drug during pregnancy. Drug dose adjustments during pregnancy are sometimes necessary due to various pregnancy-induced physiological alterations frequently associated with lower plasma concentrations. However, the clinical relevance or benefits of therapeutic drug monitoring (TDM) in pregnant women have not been specifically studied. Clinical pharmacokinetic studies in pregnant women are incredibly challenging for many reasons. Despite this, regulatory agencies have made efforts to encourage the inclusion of this population in clinical trials to achieve more information on the pharmacotherapy of pregnant women. This review aims to provide support for TDM recommendations and dose adjustments in pregnant women. METHODS The search was conducted after a predetermined strategy on PubMed and Scopus databases using the MeSH term "pregnancy" alongside other terms such as "Pregnancy and dose adjustment," "Pregnancy and therapeutic drug monitoring," "Pregnancy and PBPK," "Pregnancy and pharmacokinetics," and "Pregnancy and physiological changes." RESULTS The main information on TDM in pregnant women is available for antiepileptics, antipsychotics, antidepressants, antibiotics, antimalarials, and oncologic and immunosuppressive drugs. CONCLUSIONS More data are needed to support informed benefit-risk decision making for the administration of drugs to pregnant women. TDM and/or pharmacokinetic studies could ensure that pregnant women receive an adequate dosage of an active drug. Mechanistic modeling approaches potentially could increase our knowledge about the pharmacotherapy of this special population, and they could be used to better design dosage regimens.
Collapse
Affiliation(s)
- Fernanda de Lima Moreira
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; and
| | - Jhohann Richard de Lima Benzi
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; and
| | - Leonardo Pinto
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; and
| | - Matheus de Lucca Thomaz
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; and
| | - Geraldo Duarte
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo; and
| |
Collapse
|
10
|
Falkenhagen U, Knöchel J, Kloft C, Huisinga W. Deriving mechanism-based pharmacodynamic models by reducing quantitative systems pharmacology models: An application to warfarin. CPT Pharmacometrics Syst Pharmacol 2023; 12:432-443. [PMID: 36866520 PMCID: PMC10088086 DOI: 10.1002/psp4.12903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 03/04/2023] Open
Abstract
Quantitative systems pharmacology (QSP) models integrate comprehensive qualitative and quantitative knowledge about pharmacologically relevant processes. We previously proposed a first approach to leverage the knowledge in QSP models to derive simpler, mechanism-based pharmacodynamic (PD) models. Their complexity, however, is typically still too large to be used in the population analysis of clinical data. Here, we extend the approach beyond state reduction to also include the simplification of reaction rates, elimination of reactions, and analytic solutions. We additionally ensure that the reduced model maintains a prespecified approximation quality not only for a reference individual but also for a diverse virtual population. We illustrate the extended approach for the warfarin effect on blood coagulation. Using the model-reduction approach, we derive a novel small-scale warfarin/international normalized ratio model and demonstrate its suitability for biomarker identification. Due to the systematic nature of the approach in comparison with empirical model building, the proposed model-reduction algorithm provides an improved rationale to build PD models also from QSP models in other applications.
Collapse
Affiliation(s)
- Undine Falkenhagen
- Institute of Mathematics, University of Potsdam, Potsdam, Germany.,Graduate Research Training Program PharMetrX: Pharmacometrics & Computational Disease Modelling, Freie Universität Berlin and University of Potsdam, Potsdam, Germany
| | - Jane Knöchel
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Wilhelm Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| |
Collapse
|
11
|
Mostafa S, Polasek TM, Bousman C, Rostami‐Hodjegan A, Sheffield LJ, Everall I, Pantelis C, Kirkpatrick CMJ. Delineating gene-environment effects using virtual twins of patients treated with clozapine. CPT Pharmacometrics Syst Pharmacol 2022; 12:168-179. [PMID: 36424701 PMCID: PMC9931435 DOI: 10.1002/psp4.12886] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/27/2022] Open
Abstract
Studies that focus on individual covariates, while ignoring their interactions, may not be adequate for model-informed precision dosing (MIPD) in any given patient. Genetic variations that influence protein synthesis should be studied in conjunction with environmental covariates, such as cigarette smoking. The aim of this study was to build virtual twins (VTs) of real patients receiving clozapine with interacting covariates related to genetics and environment and to delineate the impact of interacting covariates on predicted clozapine plasma concentrations. Clozapine-treated patients with schizophrenia (N = 42) with observed clozapine plasma concentrations, demographic, environmental, and genotype data were used to construct VTs in Simcyp. The effect of increased covariate virtualization was assessed by performing simulations under three conditions: "low" (demographic), "medium" (demographic and environmental interaction), and "high" (demographic and environmental/genotype interaction) covariate virtualization. Increasing covariate virtualization with interaction improved the coefficient of variation (R2 ) from 0.07 in the low model to 0.391 and 0.368 in the medium and high models, respectively. Whereas R2 was similar between the medium and high models, the high covariate virtualization model had improved accuracy, with systematic bias of predicted clozapine plasma concentration improving from -138.48 ng/ml to -74.65 ng/ml. A high level of covariate virtualization (demographic, environmental, and genotype) may be required for MIPD using VTs.
Collapse
Affiliation(s)
- Sam Mostafa
- Centre for Medicine Use and SafetyMonash UniversityVictoriaParkvilleAustralia,MyDNA LifeAustralia LimitedVictoriaSouth YarraAustralia
| | - Thomas M. Polasek
- Centre for Medicine Use and SafetyMonash UniversityVictoriaParkvilleAustralia,CertaraNew JerseyPrincetonUSA,Department of Clinical PharmacologyRoyal Adelaide HospitalSouth AustraliaAdelaideAustralia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of PsychiatryUniversity of Melbourne & Melbourne HealthVictoriaMelbourneAustralia,The Cooperative Research Centre (CRC) for Mental HealthVictoriaMelbourneAustralia,Alberta Children's Hospital Research Institute, Cumming School of MedicineUniversity of CalgaryAlbertaCalgaryCanada,Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryAlbertaCalgaryCanada,Departments of Medical Genetics, Psychiatry, and Physiology and PharmacologyUniversity of CalgaryAlbertaCalgaryCanada
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Health SciencesUniversity of ManchesterManchesterUK,Simcyp DivisionCertara UK LimitedSheffieldUK
| | | | - Ian Everall
- Melbourne Neuropsychiatry Centre, Department of PsychiatryUniversity of Melbourne & Melbourne HealthVictoriaMelbourneAustralia,The Cooperative Research Centre (CRC) for Mental HealthVictoriaMelbourneAustralia,Western Australian Health Translation NetworkNedlandsWestern AustraliaAustralia,Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneVictoriaMelbourneAustralia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of PsychiatryUniversity of Melbourne & Melbourne HealthVictoriaMelbourneAustralia,The Cooperative Research Centre (CRC) for Mental HealthVictoriaMelbourneAustralia,Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneVictoriaMelbourneAustralia
| | | |
Collapse
|
12
|
Jelliffe R, Liu J, Drusano GL, Martinez MN. Individualized Patient Care Through Model-Informed Precision Dosing: Reflections on Training Future Practitioners. AAPS J 2022; 24:117. [PMID: 36380020 DOI: 10.1208/s12248-022-00769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Prior to his passing, Dr. Roger Jelliffe, expressed the need for educating future physicians and clinical pharmacists on the availability of computer-based tools to support dose optimization in patients in stable or unstable physiological states. His perspectives were to be captured in a commentary for the AAPS J with a focus on incorporating population pharmacokinetic (PK)/pharmacodynamic (PD) models that are designed to hit the therapeutic target with maximal precision. Unfortunately, knowing that he would be unable to complete this project, Dr. Jelliffe requested that a manuscript conveying his concerns be completed upon his passing. With this in mind, this final installment of the AAPS J theme issue titled "Alternative Perspectives for Evaluating Drug Exposure Characteristics in a Population - Avoiding Analysis Pitfalls and Pigeonholes" is an effort to honor Dr. Jelliffe's request, conveying his concerns and the need to incorporate modeling and simulation into the training of physicians and clinical pharmacists. Accordingly, Dr. Jelliffe's perspectives have been integrated with those of the other three co-authors on the following topics: the clinical utility of population PK models; the role of multiple model (MM) dosage regimens to identify an optimal dose for an individual; tools for determining dosing regimens in renal dialysis patients (or undergoing other therapies that modulate renal clearance); methods to analyze and track drug PK in acutely ill patients presenting with high inter-occasion variability; implementation of a 2-cycle approach to minimize the duration between blood samples taken to estimate the changing PK in an acutely ill patient and for the generation of therapeutic decisions in advance for each dosing cycle based on an analysis of the previous cycle; and the importance of expressing therapeutic drug monitoring results as 1/variance rather than as the coefficient of variation. Examples showcase why, irrespective of the overall approach, the combination of therapeutic drug monitoring and computer-informed precision dosing is indispensable for maximizing the likelihood of achieving the target drug concentrations in the individual patient.
Collapse
Affiliation(s)
- Roger Jelliffe
- Laboratory of Applied Pharmacokinetics and Bioinformatics, University of Southern California School of Medicine, Children's Hospital of Los Angeles, 4650 Sunset Boulevard, #51, Los Angeles, California, 90027, USA
| | - Jiang Liu
- Division of Pharmacometrics, Office of Clinical Pharmacology, Center for Drug Evaluation and Research (CDER), FDA, Silver Spring, Maryland, 20993, USA
| | - George L Drusano
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Lake Nona, Florida, 32827, USA
| | - Marilyn N Martinez
- Office of New Animal Drugs, Center for Veterinary Medicine (CVM), US Food and Drug Administration (FDA), Rockville, Maryland, 20855, USA.
| |
Collapse
|
13
|
Mostafa S, Polasek TM, Bousman CA, Müeller DJ, Sheffield LJ, Rembach J, Kirkpatrick CM. Pharmacogenomics in psychiatry - the challenge of cytochrome P450 enzyme phenoconversion and solutions to assist precision dosing. Pharmacogenomics 2022; 23:857-867. [PMID: 36169629 DOI: 10.2217/pgs-2022-0104] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pharmacogenomic (PGx) testing of cytochrome P450 (CYP) enzymes may improve the efficacy and/or safety of some medications. This is facilitated by increased availability and affordability of genotyping, the development of clinical practice PGx guidelines and regulatory support. However, the common occurrence of CYP phenoconversion, a mismatch between genotype-predicted CYP phenotype and the actual CYP phenotype, currently limits the application of PGx testing for precision dosing in psychiatry. This review proposes a stepwise approach to assist precision dosing in psychiatry via the introduction of PGx stewardship programs and innovative PGx education strategies. A future perspective on delivering precision dosing for psychiatrists is discussed that involves innovative clinical decision support systems powered by model-informed precision dosing.
Collapse
Affiliation(s)
- Sam Mostafa
- Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia.,MyDNA Life, Australia Limited, South Yarra, Victoria, Australia
| | - Thomas M Polasek
- Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia.,Certara, Princeton, NJ 08540, USA.,Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, South Australia, 5000, Australia
| | - Chad A Bousman
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, University of Melbourne & Melbourne Health, Melbourne, Victoria, 3010, Australia.,The Cooperative Research Centre (CRC) for Mental Health, Carlton, Victoria, 3053, Australia.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 1N4, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 1N4, Canada.,Departments of Medical Genetics, Psychiatry, & Physiology & Pharmacology, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Daniel J Müeller
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8, Canada
| | | | - Joel Rembach
- MyDNA Life, Australia Limited, South Yarra, Victoria, Australia
| | - Carl Mj Kirkpatrick
- Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia
| |
Collapse
|
14
|
Xiao T, Hu J, Liu S, Lu H, Li X, Kong W, Huang S, Zhu X, Zhang M, Lu H, Ni X, Yang H, Shang D, Wen Y. Population pharmacokinetics and dosing optimization of olanzapine in Chinese paediatric patients: Based on the impact of sex and concomitant valproate on clearance. J Clin Pharm Ther 2022; 47:1811-1819. [DOI: 10.1111/jcpt.13770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Tao Xiao
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Department of Clinical Research Guangdong Second Provincial General Hospital Guangzhou China
| | - Jin‐Qing Hu
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Shu‐Jing Liu
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Hui‐Qin Lu
- Department of Clinical Research Guangdong Second Provincial General Hospital Guangzhou China
| | - Xiao‐Lin Li
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Wan Kong
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Shan‐Qing Huang
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Xiu‐Qing Zhu
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Ming Zhang
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Hao‐Yang Lu
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Xiao‐Jia Ni
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Han‐Lun Yang
- School of Pharmaceutical Sciences Sun Yat‐sen University Shenzhen China
| | - De‐Wei Shang
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| | - Yu‐Guan Wen
- Department of Pharmacy The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders Guangzhou China
| |
Collapse
|
15
|
Emoto C, Johnson TN. Cytochrome P450 enzymes in the pediatric population: Connecting knowledge on P450 expression with pediatric pharmacokinetics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:365-391. [PMID: 35953161 DOI: 10.1016/bs.apha.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytochrome P450 enzymes play an important role in the pharmacokinetics, efficacy, and toxicity of drugs. Age-dependent changes in P450 enzyme expression have been studied based on several detection systems, as well as by deconvolution of in vivo pharmacokinetic data observed in pediatric populations. The age-dependent changes in P450 enzyme expression can be important determinants of drug disposition in childhood, in addition to the changes in body size and the other physiological parameters, and effects of pharmacogenetics and disease on organ functions. As a tool incorporating drug-specific and body-specific factors, physiologically-based pharmacokinetic (PBPK) models have become increasingly used to characterize and explore mechanistic insights into drug disposition. Thus, PBPK models can be a bridge between findings from basic science and utilization in predictive science. Pediatric PBPK models incorporate additional system specific information on developmental physiology and ontogeny and have been used to predict pharmacokinetic parameters from preterm neonates onwards. These models have been advocated by regulatory authorities in order to support pediatric clinical trials. The purpose of this chapter is to highlight accumulated knowledge and findings from basic research focusing on P450 enzymes, as well as the current status and future challenges of expanding the utilization of pediatric PBPK modeling.
Collapse
Affiliation(s)
- Chie Emoto
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan; Translational Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan.
| | | |
Collapse
|
16
|
van Hoogdalem MW, Johnson TN, McPhail BT, Kamatkar S, Wexelblatt SL, Ward LP, Christians U, Akinbi HT, Vinks AA, Mizuno T. Physiologically-Based Pharmacokinetic Modeling to Investigate the Effect of Maturation on Buprenorphine Pharmacokinetics in Newborns with Neonatal Opioid Withdrawal Syndrome. Clin Pharmacol Ther 2022; 111:496-508. [PMID: 34679189 PMCID: PMC8748288 DOI: 10.1002/cpt.2458] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/15/2021] [Indexed: 02/03/2023]
Abstract
Neonatal opioid withdrawal syndrome (NOWS) is a major public health concern whose incidence has paralleled the opioid epidemic in the United States. Sublingual buprenorphine is an emerging treatment for NOWS, but given concerns about long-term adverse effects of perinatal opioid exposure, precision dosing of buprenorphine is needed. Buprenorphine pharmacokinetics (PK) in newborns, however, is highly variable. To evaluate underlying sources of PK variability, a neonatal physiologically-based pharmacokinetic (PBPK) model of sublingual buprenorphine was developed using Simcyp (version 19.1). The PBPK model included metabolism by cytochrome P450 (CYP) 3A4, CYP2C8, UDP-glucuronosyltransferase (UGT) 1A1, UGT1A3, UGT2B7, and UGT2B17, with additional biliary excretion. Maturation of metabolizing enzymes was incorporated, and default CYP2C8 and UGT2B7 ontogeny profiles were updated according to recent literature. A biliary clearance developmental profile was outlined using clinical data from neonates receiving sublingual buprenorphine as NOWS treatment. Extensive PBPK model validation in adults demonstrated good predictability, with geometric mean (95% confidence interval (CI)) predicted/observed ratios (P/O ratios) of area under the curve from zero to infinity (AUC0-∞ ), peak concentration (Cmax ), and time to reach peak concentration (Tmax ) equaling 1.00 (0.74-1.33), 1.04 (0.84-1.29), and 0.95 (0.72-1.26), respectively. In neonates, the geometric mean (95% CI) P/O ratio of whole blood concentrations was 0.75 (95% CI 0.64-0.87). PBPK modeling and simulation demonstrated that variability in biliary clearance, sublingual absorption, and CYP3A4 abundance are likely important drivers of buprenorphine PK variability in neonates. The PBPK model could be used to guide development of improved buprenorphine starting dose regimens for the treatment of NOWS.
Collapse
Affiliation(s)
- Matthijs W. van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | | | - Brooks T. McPhail
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- School of Medicine Greenville, University of South Carolina, Greenville, SC, USA
| | - Suyog Kamatkar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Community Hospital East, Indianapolis, IN, USA
| | - Scott L. Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Laura P. Ward
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Uwe Christians
- iC42 Clinical Research and Development, University of Colorado, Aurora, CO, USA
| | - Henry T. Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alexander A. Vinks
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
17
|
Collin CB, Gebhardt T, Golebiewski M, Karaderi T, Hillemanns M, Khan FM, Salehzadeh-Yazdi A, Kirschner M, Krobitsch S, Kuepfer L. Computational Models for Clinical Applications in Personalized Medicine—Guidelines and Recommendations for Data Integration and Model Validation. J Pers Med 2022; 12:jpm12020166. [PMID: 35207655 PMCID: PMC8879572 DOI: 10.3390/jpm12020166] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
The future development of personalized medicine depends on a vast exchange of data from different sources, as well as harmonized integrative analysis of large-scale clinical health and sample data. Computational-modelling approaches play a key role in the analysis of the underlying molecular processes and pathways that characterize human biology, but they also lead to a more profound understanding of the mechanisms and factors that drive diseases; hence, they allow personalized treatment strategies that are guided by central clinical questions. However, despite the growing popularity of computational-modelling approaches in different stakeholder communities, there are still many hurdles to overcome for their clinical routine implementation in the future. Especially the integration of heterogeneous data from multiple sources and types are challenging tasks that require clear guidelines that also have to comply with high ethical and legal standards. Here, we discuss the most relevant computational models for personalized medicine in detail that can be considered as best-practice guidelines for application in clinical care. We define specific challenges and provide applicable guidelines and recommendations for study design, data acquisition, and operation as well as for model validation and clinical translation and other research areas.
Collapse
Affiliation(s)
- Catherine Bjerre Collin
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 N Copenhagen, Denmark; (C.B.C.); (T.K.)
| | - Tom Gebhardt
- Department of Systems Biology and Bioinformatics, University of Rostock, 18057 Rostock, Germany; (T.G.); (M.H.); (F.M.K.)
| | - Martin Golebiewski
- Heidelberg Institute for Theoretical Studies gGmbH, 69118 Heidelberg, Germany;
| | - Tugce Karaderi
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 N Copenhagen, Denmark; (C.B.C.); (T.K.)
- Center for Health Data Science, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 N Copenhagen, Denmark
| | - Maximilian Hillemanns
- Department of Systems Biology and Bioinformatics, University of Rostock, 18057 Rostock, Germany; (T.G.); (M.H.); (F.M.K.)
| | - Faiz Muhammad Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, 18057 Rostock, Germany; (T.G.); (M.H.); (F.M.K.)
| | | | - Marc Kirschner
- Forschungszentrum Jülich GmbH, Project Management Jülich, 52425 Jülich, Germany; (M.K.); (S.K.)
| | - Sylvia Krobitsch
- Forschungszentrum Jülich GmbH, Project Management Jülich, 52425 Jülich, Germany; (M.K.); (S.K.)
| | | | - Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Correspondence: ; Tel.: +49-241-8085900
| |
Collapse
|
18
|
Zheng L, Yang H, Dallmann A, Jiang X, Wang L, Hu W. Physiologically Based Pharmacokinetic Modeling in Pregnant Women Suggests Minor Decrease in Maternal Exposure to Olanzapine. Front Pharmacol 2022; 12:793346. [PMID: 35126130 PMCID: PMC8807508 DOI: 10.3389/fphar.2021.793346] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023] Open
Abstract
Pregnancy is accompanied by significant physiological changes that might affect the in vivo drug disposition. Olanzapine is prescribed to pregnant women with schizophrenia, while its pharmacokinetics during pregnancy remains unclear. This study aimed to develop a physiologically based pharmacokinetic (PBPK) model of olanzapine in the pregnant population. With the contributions of each clearance pathway determined beforehand, a full PBPK model was developed and validated in the non-pregnant population. This model was then extrapolated to predict steady-state pharmacokinetics in the three trimesters of pregnancy by introducing gestation-related alterations. The model adequately simulated the reported time-concentration curves. The geometric mean fold error of Cmax and AUC was 1.14 and 1.09, respectively. The model predicted that under 10 mg daily dose, the systematic exposure of olanzapine had minor changes (less than 28%) throughout pregnancy. We proposed that the reduction in cytochrome P4501A2 activity is counteracted by the induction of other enzymes, especially glucuronyltransferase1A4. In conclusion, the PBPK model simulations suggest that, at least at the tested stages of pregnancy, dose adjustment of olanzapine can hardly be recommended for pregnant women if effective treatment was achieved before the onset of pregnancy and if fetal toxicity can be ruled out.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hongyi Yang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals Bayer AG, Leverkusen, Germany
| | - Xuehua Jiang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Ling Wang, ; Wei Hu,
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ling Wang, ; Wei Hu,
| |
Collapse
|
19
|
Combining Therapeutic Drug Monitoring and Pharmacokinetic Modelling Deconvolutes Physiological and Environmental Sources of Variability in Clozapine Exposure. Pharmaceutics 2021; 14:pharmaceutics14010047. [PMID: 35056943 PMCID: PMC8779032 DOI: 10.3390/pharmaceutics14010047] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Clozapine is a key antipsychotic drug for treatment-resistant schizophrenia but exhibits highly variable pharmacokinetics and a propensity for serious adverse effects. Currently, these challenges are addressed using therapeutic drug monitoring (TDM). This study primarily sought to (i) verify the importance of covariates identified in a prior clozapine population pharmacokinetic (popPK) model in the absence of environmental covariates using physiologically based pharmacokinetic (PBPK) modelling, and then to (ii) evaluate the performance of the popPK model as an adjunct or alternative to TDM-guided dosing in an active TDM population. Methods: A popPK model incorporating age, metabolic activity, sex, smoking status and weight was applied to predict clozapine trough concentrations (Cmin) in a PBPK-simulated population and an active TDM population comprising 142 patients dosed to steady state at Flinders Medical Centre in Adelaide, South Australia. Post hoc analyses were performed to deconvolute the impact of physiological and environmental covariates in the TDM population. Results: Analysis of PBPK simulations confirmed age, cytochrome P450 1A2 activity, sex and weight as physiological covariates associated with variability in clozapine Cmin (R2 = 0.7698; p = 0.0002). Prediction of clozapine Cmin using a popPK model based on these covariates accounted for <5% of inter-individual variability in the TDM population. Post hoc analyses confirmed that environmental covariates accounted for a greater proportion of the variability in clozapine Cmin in the TDM population. Conclusions: Variability in clozapine exposure was primarily driven by environmental covariates in an active TDM population. Pharmacokinetic modelling can be used as an adjunct to TDM to deconvolute sources of variability in clozapine exposure.
Collapse
|
20
|
Abouir K, Samer CF, Gloor Y, Desmeules JA, Daali Y. Reviewing Data Integrated for PBPK Model Development to Predict Metabolic Drug-Drug Interactions: Shifting Perspectives and Emerging Trends. Front Pharmacol 2021; 12:708299. [PMID: 34776945 PMCID: PMC8582169 DOI: 10.3389/fphar.2021.708299] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023] Open
Abstract
Physiologically-based pharmacokinetics (PBPK) modeling is a robust tool that supports drug development and the pharmaceutical industry and regulatory authorities. Implementation of predictive systems in the clinics is more than ever a reality, resulting in a surge of interest for PBPK models by clinicians. We aimed to establish a repository of available PBPK models developed to date to predict drug-drug interactions (DDIs) in the different therapeutic areas by integrating intrinsic and extrinsic factors such as genetic polymorphisms of the cytochromes or environmental clues. This work includes peer-reviewed publications and models developed in the literature from October 2017 to January 2021. Information about the software, type of model, size, and population model was extracted for each article. In general, modeling was mainly done for DDI prediction via Simcyp® software and Full PBPK. Overall, the necessary physiological and physio-pathological parameters, such as weight, BMI, liver or kidney function, relative to the drug absorption, distribution, metabolism, and elimination and to the population studied for model construction was publicly available. Of the 46 articles, 32 sensibly predicted DDI potentials, but only 23% integrated the genetic aspect to the developed models. Marked differences in concentration time profiles and maximum plasma concentration could be explained by the significant precision of the input parameters such as Tissue: plasma partition coefficients, protein abundance, or Ki values. In conclusion, the models show a good correlation between the predicted and observed plasma concentration values. These correlations are all the more pronounced as the model is rich in data representative of the population and the molecule in question. PBPK for DDI prediction is a promising approach in clinical, and harmonization of clearance prediction may be helped by a consensus on selecting the best data to use for PBPK model development.
Collapse
Affiliation(s)
- Kenza Abouir
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Geneva, Switzerland
| | - Caroline F Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yvonne Gloor
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Jules A Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
21
|
Zubiaur P, Soria-Chacartegui P, Villapalos-García G, Gordillo-Perdomo JJ, Abad-Santos F. The pharmacogenetics of treatment with olanzapine. Pharmacogenomics 2021; 22:939-958. [PMID: 34528455 DOI: 10.2217/pgs-2021-0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Genetic polymorphism in olanzapine-metabolizing enzymes, transporters and drug targets is associated with alterations in safety and efficacy. The aim of this systematic review is to describe all clinically relevant pharmacogenetic information on olanzapine and to propose clinically actionable variants. Two hundred and eighty-four studies were screened; 76 complied with the inclusion criteria and presented significant associations. DRD2 Taq1A (rs1800497) *A1, LEP -2548 (rs7799039) G and CYP1A2*1F alleles were related to olanzapine effectiveness and safety variability in several studies, with a high level of evidence. DRD2 -141 (rs1799732) Ins, A-241G (rs1799978) G, DRD3 Ser9Gly (rs6280) Gly, HTR2A rs7997012 A, ABCB1 C3435T (rs1045642) T and G2677T/A (rs2032582) T and UGT1A4*3 alleles were related to safety, effectiveness and/or pharmacokinetic variability with moderated level of evidence.
Collapse
Affiliation(s)
- Pablo Zubiaur
- Department of Clinical Pharmacology, Hospital Universitario de La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain.,UICEC Hospital Universitario de La Princesa, Plataforma SCReN (Spanish Clinical Research Network), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain
| | - Paula Soria-Chacartegui
- Department of Clinical Pharmacology, Hospital Universitario de La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain
| | - Gonzalo Villapalos-García
- Department of Clinical Pharmacology, Hospital Universitario de La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain
| | - Juan J Gordillo-Perdomo
- Department of Clinical Analysis, Hospital Universitario de La Princesa, Madrid, 28006, Spain
| | - Francisco Abad-Santos
- Department of Clinical Pharmacology, Hospital Universitario de La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain.,UICEC Hospital Universitario de La Princesa, Plataforma SCReN (Spanish Clinical Research Network), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, 28006, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28006, Spain
| |
Collapse
|
22
|
Fendt R, Hofmann U, Schneider ARP, Schaeffeler E, Burghaus R, Yilmaz A, Blank LM, Kerb R, Lippert J, Schlender JF, Schwab M, Kuepfer L. Data-driven personalization of a physiologically based pharmacokinetic model for caffeine: A systematic assessment. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:782-793. [PMID: 34053199 PMCID: PMC8302243 DOI: 10.1002/psp4.12646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/17/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) models have been proposed as a tool for more accurate individual pharmacokinetic (PK) predictions and model‐informed precision dosing, but their application in clinical practice is still rare. This study systematically assesses the benefit of using individual patient information to improve PK predictions. A PBPK model of caffeine was stepwise personalized by using individual data on (1) demography, (2) physiology, and (3) cytochrome P450 (CYP) 1A2 phenotype of 48 healthy volunteers participating in a single‐dose clinical study. Model performance was benchmarked against a caffeine base model simulated with parameters of an average individual. In the first step, virtual twins were generated based on the study subjects' demography (height, weight, age, sex), which implicated the rescaling of average organ volumes and blood flows. The accuracy of PK simulations improved compared with the base model. The percentage of predictions within 0.8‐fold to 1.25‐fold of the observed values increased from 45.8% (base model) to 57.8% (Step 1). However, setting physiological parameters (liver blood flow determined by magnetic resonance imaging, glomerular filtration rate, hematocrit) to measured values in the second step did not further improve the simulation result (59.1% in the 1.25‐fold range). In the third step, virtual twins matching individual demography, physiology, and CYP1A2 activity considerably improved the simulation results. The percentage of data within the 1.25‐fold range was 66.15%. This case study shows that individual PK profiles can be predicted more accurately by considering individual attributes and that personalized PBPK models could be a valuable tool for model‐informed precision dosing approaches in the future.
Collapse
Affiliation(s)
- Rebekka Fendt
- Systems Pharmacology & Medicine, Bayer AG, Leverkusen, Germany.,Institute of Applied Microbiology, Aachen Biology and Biotechnology, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Annika R P Schneider
- Systems Pharmacology & Medicine, Bayer AG, Leverkusen, Germany.,Institute of Applied Microbiology, Aachen Biology and Biotechnology, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Rolf Burghaus
- Systems Pharmacology & Medicine, Bayer AG, Leverkusen, Germany
| | - Ali Yilmaz
- Department of Cardiology I, University Hospital Muenster, Münster, Germany
| | - Lars Mathias Blank
- Institute of Applied Microbiology, Aachen Biology and Biotechnology, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Reinhold Kerb
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Jörg Lippert
- Systems Pharmacology & Medicine, Bayer AG, Leverkusen, Germany
| | | | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Departments of Clinical Pharmacology and Biochemistry and Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Lars Kuepfer
- Systems Pharmacology & Medicine, Bayer AG, Leverkusen, Germany.,Institute for Systems Medicine With Focus on Organ Interactions, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
23
|
Zapke SE, Willmann S, Grebe SO, Menke K, Thürmann PA, Schmiedl S. Comparing Predictions of a PBPK Model for Cyclosporine With Drug Levels From Therapeutic Drug Monitoring. Front Pharmacol 2021; 12:630904. [PMID: 34054518 PMCID: PMC8161189 DOI: 10.3389/fphar.2021.630904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/27/2021] [Indexed: 01/05/2023] Open
Abstract
This study compared simulations of a physiologically based pharmacokinetic (PBPK) model implemented for cyclosporine with drug levels from therapeutic drug monitoring to evaluate the predictive performance of a PBPK model in a clinical population. Based on a literature search model parameters were determined. After calibrating the model using the pharmacokinetic profiles of healthy volunteers, 356 cyclosporine trough levels of 32 renal transplant outpatients were predicted based on their biometric parameters. Model performance was assessed by calculating absolute and relative deviations of predicted and observed trough levels. The median absolute deviation was 6 ng/ml (interquartile range: 30 to 31 ng/ml, minimum = -379 ng/ml, maximum = 139 ng/ml). 86% of predicted cyclosporine trough levels deviated less than twofold from observed values. The high intra-individual variability of observed cyclosporine levels was not fully covered by the PBPK model. Perspectively, consideration of clinical and additional patient-related factors may improve the model's performance. In summary, the current study has shown that PBPK modeling may offer valuable contributions for pharmacokinetic research in clinical drug therapy.
Collapse
Affiliation(s)
- Sonja E Zapke
- Department of Clinical Pharmacology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Stefan Willmann
- Bayer AG, Research and Development, Clinical Pharmacometrics, Wuppertal, Germany
| | - Scott-Oliver Grebe
- Medical Clinic 1, Division of Nephrology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Kristin Menke
- Bayer AG, Research and Development, Systems Pharmacology and Medicine I, Leverkusen, Germany
| | - Petra A Thürmann
- Department of Clinical Pharmacology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany.,Philipp Klee-Institute for Clinical Pharmacology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Sven Schmiedl
- Department of Clinical Pharmacology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany.,Philipp Klee-Institute for Clinical Pharmacology, Helios University Hospital Wuppertal, Wuppertal, Germany
| |
Collapse
|
24
|
Mechanistic Modelling Identifies and Addresses the Risks of Empiric Concentration-Guided Sorafenib Dosing. Pharmaceuticals (Basel) 2021; 14:ph14050389. [PMID: 33919091 PMCID: PMC8143107 DOI: 10.3390/ph14050389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
The primary objective of this study is to evaluate the capacity of concentration-guided sorafenib dosing protocols to increase the proportion of patients that achieve a sorafenib maximal concentration (Cmax) within the range 4.78 to 5.78 μg/mL. A full physiologically based pharmacokinetic model was built and validated using Simcyp® (version 19.1). The model was used to simulate sorafenib exposure in 1000 Sim-Cancer subjects over 14 days. The capacity of concentration-guided sorafenib dose adjustment, with/without model-informed dose selection (MIDS), to achieve a sorafenib Cmax within the range 4.78 to 5.78 μg/mL was evaluated in 500 Sim-Cancer subjects. A multivariable linear regression model incorporating hepatic cytochrome P450 (CYP) 3A4 abundance, albumin concentration, body mass index, body surface area, sex and weight provided robust prediction of steady-state sorafenib Cmax (R2 = 0.883; p < 0.001). These covariates identified subjects at risk of failing to achieve a sorafenib Cmax ≥ 4.78 μg/mL with 95.0% specificity and 95.2% sensitivity. Concentration-guided sorafenib dosing with MIDS achieved a sorafenib Cmax within the range 4.78 to 5.78 μg/mL for 38 of 52 patients who failed to achieve a Cmax ≥ 4.78 μg/mL with standard dosing. In a simulation setting, concentration-guided dosing with MIDS was the quickest and most effective approach to achieve a sorafenib Cmax within a designated range.
Collapse
|
25
|
Lee J, Kim MG, Jeong HC, Shin KH. Physiologically-based pharmacokinetic model for clozapine in Korean patients with schizophrenia. Transl Clin Pharmacol 2021; 29:33-44. [PMID: 33854999 PMCID: PMC8020364 DOI: 10.12793/tcp.2021.29.e3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Clozapine has been used as a treatment of schizophrenia. Despite its large interindividual variability, few reports addressed the physiologically-based pharmacokinetic modeling and simulation (PBPK M&S) of clozapine in patients. This study aimed to develop a PBPK M&S of clozapine in Korean patients with schizophrenia. PBPK modeling for clozapine was constructed using a population-based PBPK platform, the SimCYP® Simulator (V19; Certara, Sheffield, UK). The PBPK model was developed by optimizing the physiological parameters of the built-in population and compound libraries in the SimCYP® Simulator. The model verification was performed with the predicted/observed ratio for pharmacokinetic parameters and visual predictive checks (VPCs) plot. Simulations were performed to predict toxicities according to dosing regimens. From published data, 230 virtual trials were simulated for each dosing regimen. The predicted/observed ratio for the area under the curve and peak plasma concentration was calculated to be from 0.78 to 1.34. The observation profiles were within the 5th and 95th percentile range with no serious model misspecification through the VPC plot. A significant impact on age and gender was found for clozapine clearance. The simulation results suggested that 150 mg twice a day and 150 mg three times a day of clozapine have toxicity concerns. In conclusion, a PBPK model was developed and reasonable parameters were made from the data of Korean patients with schizophrenia. The provided model might be used to predict the pharmacokinetics of clozapine and assist dose adjustment in clinical settings.
Collapse
Affiliation(s)
- Joomi Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Min-Gul Kim
- Center for Clinical Pharmacology and Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Korea.,Department of Pharmacology, School of Medicine, Jeonbuk National University, Jeonju 54907, Korea
| | - Hyeon-Cheol Jeong
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Kwang-Hee Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
26
|
Arnaiz JA, Rodrigues-Silva C, Mezquida G, Amoretti S, Cuesta MJ, Fraguas D, Lobo A, González-Pinto A, Díaz-Caneja MC, Corripio I, Vieta E, Baeza I, Mané A, García-Rizo C, Bioque M, Saiz J, Bernardo M, Mas S. The usefulness of Olanzapine plasma concentrations in monitoring treatment efficacy and metabolic disturbances in first-episode psychosis. Psychopharmacology (Berl) 2021; 238:665-676. [PMID: 33230696 DOI: 10.1007/s00213-020-05715-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The role of Olanzapine therapeutic drug monitoring is controversial. The present study explores the associations of Olanzapine plasma concentrations with clinical response and metabolic side effects in first episode psychosis (FEP) after 2 months of treatment. METHODS Forty-seven patients were included. Improvement in clinical symptomatology was assessed using the PANSS. Metabolic assessment included weight, blood pressure, waist circumference, blood glucose, total cholesterol, high-density lipoprotein, low-density lipoprotein, and triglycerides. RESULTS The Olanzapine plasma concentrations after 2 months of treatment were positively correlated with weight gain (r = 0.49, p = 0.003), and a concentration > 23.28 ng/mL was identified as a positive predictor of weight gain (≥ 7%). The Olanzapine concentration to dose (C/D) ratio was positively correlated with the percentage of improvement in the total PANSS (r = 0.46, p = 0.004), and a C/D ratio > 2.12 was identified as a positive predictor of a good response (percentage of improvement > 30%) after 2 months of treatment. We also identified several factors that could alter Olanzapine pharmacokinetics: gender (p = 0.03), diagnosis (p = 0.05), smoking habit (p = 0.05), and co-medications such as valproic acid (p = 0.05) and anxiolytics (p = 0.01). DISCUSSION In conclusion, our results suggest that therapeutic drug monitoring of Olanzapine could be helpful to evaluate therapeutic efficacy and metabolic dysfunction in FEP patients treated with Olanzapine.
Collapse
Affiliation(s)
- J A Arnaiz
- Phase I Unit, Clinical Pharmacology Department, Hospital Clinic de Barcelona (HCB), Barcelona, Spain.,Department of Basic Clinical Practice, University of Barcelona (UB), Casanova 143, E-08036, Barcelona, Spain
| | - C Rodrigues-Silva
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - G Mezquida
- Department of Basic Clinical Practice, University of Barcelona (UB), Casanova 143, E-08036, Barcelona, Spain.,Barcelona Clínic Schizophrenia Unit, Neuroscience Institute, HCB, Barcelona, Catalunya, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain
| | - S Amoretti
- Barcelona Clínic Schizophrenia Unit, Neuroscience Institute, HCB, Barcelona, Catalunya, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain
| | - M J Cuesta
- Departmentof Psychiatry, Complejo Hospitalario de Navarra, Instituto de Investigaciones Sanitarias de Navarra (IdiSNa), Pamplona, Spain
| | - D Fraguas
- Institute of Psychiatry and Mental Health, Hospital Clínico San Carlos, IdISSC, CIBERSAM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - A Lobo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Medicine and Psychiatry, Zaragoza University, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - A González-Pinto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Hospital Universitario Araba, Servicio de Psiquiatria, UPV/EHU, Bioaraba, Spain
| | - M C Díaz-Caneja
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañon, CIBERSAM, IiSGM, School of Medicine, Universidad Complutense, Madrid, Spain
| | - I Corripio
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Psychiatry Department, Institut d'Investigació Biomèdica-Sant Pau (IIB-SANT PAU), Hospital de la Santa Creu i Sant Pau; Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - E Vieta
- Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - I Baeza
- Department of Child and Adolescent Psychiatry and Psychology, Clínic Institute of Neurosciences, Hospital Clínic de Barcelona, 2017SGR881, University of Barcelona, CIBERSAM, IDIBAPS, Barcelona, Spain
| | - A Mané
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Autonomous university of Barcelona (UAB), Barcelona, Spain
| | - C García-Rizo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain.,Barcelona Clínic Schizophrenia Unit, Neuroscience Institute, Hospital Clínic of Barcelona, Barcelona, Spain.,Department of Medicine, Barcelona, UB, Spain
| | - M Bioque
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain.,Barcelona Clínic Schizophrenia Unit, Neuroscience Institute, Hospital Clínic of Barcelona, Barcelona, Spain.,Department of Medicine, Barcelona, UB, Spain
| | - J Saiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Psychiatry, Hospital Universitario Ramón y Cajal, IRYCIS, Universidad de Alcalá, Madrid, Spain
| | - M Bernardo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain.,Barcelona Clínic Schizophrenia Unit, Neuroscience Institute, Hospital Clínic of Barcelona, Barcelona, Spain.,Department of Medicine, Barcelona, UB, Spain
| | - S Mas
- Department of Basic Clinical Practice, University of Barcelona (UB), Casanova 143, E-08036, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain. .,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain.
| | | |
Collapse
|
27
|
Franchetti Y, Nolin TD. Application of Individualized PBPK Modeling of Rate Data to Evaluate the Effect of Hemodialysis on Nonrenal Clearance Pathways. J Clin Pharmacol 2021; 61:769-781. [PMID: 33459400 DOI: 10.1002/jcph.1818] [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: 11/07/2020] [Accepted: 01/11/2021] [Indexed: 11/06/2022]
Abstract
The aim of this study was to apply individualized, physiologically based pharmacokinetic modeling of 14 CO2 production rates (iPBPK-R) measured by the erythromycin breath test to characterize the effect of hemodialysis on the function of nonrenal clearance pathways in patients with end-stage renal disease. Twelve patients previously received 14 C-erythromycin intravenously pre- and post-hemodialysis. Serial breath samples were collected after each dose over 2 hours. Eight PBPK parameters were co-estimated across periods, whereas activity of cytochrome P450 (CYP) 3A4 clearance was independently estimated for each period. Inhibition coefficients for organic anion transporting polypeptide (OATP), P-glycoprotein, and multidrug resistance-associated protein 2 activities were also estimated. Nonrenal clearance parameter estimates were explored regarding sex differences and correlations with uremic toxins and were used in hierarchical cluster analysis (HCA). Relationships between the function of nonrenal clearance pathways and uremic toxin concentrations were explored. Mean CYP 3A4 clearance increased by 2.2% post-hemodialysis. Uptake transporter activity was highly intervariable across hemodialysis. Females had 22% and 30% higher median CYP3A4 activity than males pre- and post-hemodialysis, respectively. Exploratory HCA indicated that using both CYP3A4 and OATP activity parameters at pre- and post-hemodialysis best identifies heterogeneous patients. This is the first study to use the iPBPK-R approach to simultaneously estimate multiple in vivo nonrenal elimination pathways in individual patients with kidney disease and to assess the effect of hemodialysis.
Collapse
Affiliation(s)
- Yoko Franchetti
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Badaoui S, Hopkins AM, Rodrigues AD, Miners JO, Sorich MJ, Rowland A. Application of Model Informed Precision Dosing to Address the Impact of Pregnancy Stage and CYP2D6 Phenotype on Foetal Morphine Exposure. AAPS JOURNAL 2021; 23:15. [PMID: 33404848 DOI: 10.1208/s12248-020-00541-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Guidance regarding the effect of codeine and its metabolites on foetal development is limited by small studies and inconsistent findings. The primary objective was to use physiologically based pharmacokinetic modelling to investigate the impact of gestational stage and maternal CYP2D6 phenotype on foetal morphine exposure following codeine administration. Full body physiologically based pharmacokinetic models were developed and verified for codeine and morphine using Simcyp (version 19.1). The impact of gestational age and maternal CYP2D6 phenotype on foetal and maternal morphine and codeine exposure following oral codeine administration was modelled in a cohort of 250 pregnant females and foetuses at gestational weeks 0 (mothers only), 6, 12, 24 and 36. Consistent with the known effect on codeine metabolism, a clinically meaningful (> 1.65-fold) increase in foetal morphine AUC was observed in the CYP2D6 UM phenotype cohort compared to the CYP2D6 EM and PM phenotype cohorts. The mean (95% CI) foetal morphine AUC in the CYP2D6 UM cohort of 0.988 (0.902 to 1.073) ng/mL.h was 1.8-fold higher than the CYP2D6 EM cohort of 0.546 (0.492 to 0.600) ng/mL.h (p < 0.001). Despite a 2.8-fold increase in maternal CYP2D6 protein abundance between gestational weeks 6 and 36, the mean foetal morphine AUC in the CYP2D6 EM and UM phenotype cohorts reduced by 1.55- and 1.75-fold, respectively, over this period. Maternal CYP2D6 phenotype is a significant determinant of foetal morphine AUC. Simulations suggest that the greatest risk with respect to foetal morphine exposure is during the first trimester of pregnancy, particularly in CYP2D6 UM phenotype mothers.
Collapse
Affiliation(s)
- Sarah Badaoui
- College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia
| | - A David Rodrigues
- ADME Sciences, Medicine Design, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - John O Miners
- College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia.
| |
Collapse
|
29
|
Mostafa S, Polasek TM, Sheffield LJ, Huppert D, Kirkpatrick CMJ. Quantifying the Impact of Phenoconversion on Medications With Actionable Pharmacogenomic Guideline Recommendations in an Acute Aged Persons Mental Health Setting. Front Psychiatry 2021; 12:724170. [PMID: 34489765 PMCID: PMC8416898 DOI: 10.3389/fpsyt.2021.724170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Polypharmacy and genetic variants that strongly influence medication response (pharmacogenomics, PGx) are two well-described risk factors for adverse drug reactions. Complexities arise in interpreting PGx results in the presence of co-administered medications that can cause cytochrome P450 enzyme phenoconversion. Aim: To quantify phenoconversion in a cohort of acute aged persons mental health patients and evaluate its impact on the reporting of medications with actionable PGx guideline recommendations (APRs). Methods: Acute aged persons mental health patients (N = 137) with PGx and medication data at admission and discharge were selected to describe phenoconversion frequencies for CYP2D6, CYP2C19 and CYP2C9 enzymes. The expected impact of phenoconversion was then assessed on the reporting of medications with APRs. Results: Post-phenoconversion, the predicted frequency at admission and discharge increased for CYP2D6 intermediate metabolisers (IMs) by 11.7 and 16.1%, respectively. Similarly, for CYP2C19 IMs, the predicted frequency at admission and discharge increased by 13.1 and 11.7%, respectively. Nineteen medications with APRs were prescribed 120 times at admission, of which 50 (42%) had APRs pre-phenoconversion, increasing to 60 prescriptions (50%) post-phenoconversion. At discharge, 18 medications with APRs were prescribed 122 times, of which 48 (39%) had APRs pre-phenoconversion, increasing to 57 prescriptions (47%) post-phenoconversion. Discussion: Aged persons mental health patients are commonly prescribed medications with APRs, but interpretation of these recommendations must consider the effects of phenoconversion. Adopting a collaborative care model between prescribers and clinical pharmacists should be considered to address phenoconversion and ensure the potential benefits of PGx are maximised.
Collapse
Affiliation(s)
- Sam Mostafa
- Centre for Medicine Use and Safety, Monash University, Parkville, VIC, Australia.,MyDNA Life, Australia Limited, South Yarra, VIC, Australia
| | - Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Parkville, VIC, Australia.,Certara, Princeton, NJ, United States.,Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Leslie J Sheffield
- MyDNA Life, Australia Limited, South Yarra, VIC, Australia.,Department of Genetic Medicine, Melbourne Health, Parkville, VIC, Australia
| | - David Huppert
- Department of Aged & Liaison Psychiatry, Alfred Health, Melbourne, VIC, Australia.,Northwestern Mental Health, Melbourne Health, Melbourne, VIC, Australia
| | - Carl M J Kirkpatrick
- Centre for Medicine Use and Safety, Monash University, Parkville, VIC, Australia
| |
Collapse
|
30
|
Franchetti Y, Nolin TD. Dose Optimization in Kidney Disease: Opportunities for PBPK Modeling and Simulation. J Clin Pharmacol 2020; 60 Suppl 1:S36-S51. [PMID: 33205428 DOI: 10.1002/jcph.1741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022]
Abstract
Kidney disease affects pharmacokinetic (PK) profiles of not only renally cleared drugs but also nonrenally cleared drugs. The impact of kidney disease on drug disposition has not been fully elucidated, but describing the extent of such impact is essential for conducting dose optimization in kidney disease. Accurate evaluation of kidney function has been a clinical interest for dose optimization, and more scientists pay attention and conduct research for clarifying the role of drug transporters, metabolic enzymes, and their interplay in drug disposition as kidney disease progresses. Physiologically based pharmacokinetic (PBPK) modeling and simulation can provide valuable insights for dose optimization in kidney disease. It is a powerful tool to integrate discrete knowledge from preclinical and clinical research and mechanistically investigate system- and drug-dependent factors that may contribute to the changes in PK profiles. PBPK-based prediction of drug exposures may be used a priori to adjust dosing regimens and thereby minimize the likelihood of drug-related toxicity. With real-time clinical studies, parameter estimation may be performed with PBPK approaches that can facilitate identification of sources of interindividual variability. PBPK modeling may also facilitate biomarker research that aids dose optimization in kidney disease. U.S. Food and Drug Administration guidances related to conduction of PK studies in kidney impairment and PBPK documentation provide the foundation for facilitating model-based dose-finding research in kidney disease.
Collapse
Affiliation(s)
- Yoko Franchetti
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Geerts H, van der Graaf P. A modeling informed quantitative approach to salvage clinical trials interrupted due to COVID-19. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12053. [PMID: 33163611 PMCID: PMC7606183 DOI: 10.1002/trc2.12053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/01/2020] [Indexed: 11/29/2022]
Abstract
Many ongoing Alzheimer's disease central nervous system clinical trials are being disrupted and halted due to the COVID-19 pandemic. They are often of a long duration' are very complex; and involve many stakeholders, not only the scientists and regulators but also the patients and their family members. It is mandatory for us as a community to explore all possibilities to avoid losing all the knowledge we have gained from these ongoing trials. Some of these trials will need to completely restart, but a substantial number can restart after a hiatus with the proper protocol amendments. To salvage the information gathered so far, we need out-of-the-box thinking for addressing these missingness problems and to combine information from the completers with those subjects undergoing complex protocols deviations and amendments after restart in a rational, scientific way. Physiology-based pharmacokinetic (PBPK) modeling has been a cornerstone of model-informed drug development with regard to drug exposure at the site of action, taking into account individual patient characteristics. Quantitative systems pharmacology (QSP), based on biology-informed and mechanistic modeling of the interaction between a drug and neuronal circuits, is an emerging technology to simulate the pharmacodynamic effects of a drug in combination with patient-specific comedications, genotypes, and disease states on functional clinical scales. We propose to combine these two approaches into the concept of computer modeling-based virtual twin patients as a possible solution to harmonize the readouts from these complex clinical datasets in a biologically and therapeutically relevant way.
Collapse
|
32
|
Darwich AS, Polasek TM, Aronson JK, Ogungbenro K, Wright DFB, Achour B, Reny JL, Daali Y, Eiermann B, Cook J, Lesko L, McLachlan AJ, Rostami-Hodjegan A. Model-Informed Precision Dosing: Background, Requirements, Validation, Implementation, and Forward Trajectory of Individualizing Drug Therapy. Annu Rev Pharmacol Toxicol 2020; 61:225-245. [PMID: 33035445 DOI: 10.1146/annurev-pharmtox-033020-113257] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Model-informed precision dosing (MIPD) has become synonymous with modern approaches for individualizing drug therapy, in which the characteristics of each patient are considered as opposed to applying a one-size-fits-all alternative. This review provides a brief account of the current knowledge, practices, and opinions on MIPD while defining an achievable vision for MIPD in clinical care based on available evidence. We begin with a historical perspective on variability in dose requirements and then discuss technical aspects of MIPD, including the need for clinical decision support tools, practical validation, and implementation of MIPD in health care. We also discuss novel ways to characterize patient variability beyond the common perceptions of genetic control. Finally, we address current debates on MIPD from the perspectives of the new drug development, health economics, and drug regulations.
Collapse
Affiliation(s)
- Adam S Darwich
- Logistics and Informatics in Health Care, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, SE-141 57 Huddinge, Sweden
| | - Thomas M Polasek
- Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, South Australia 5000, Australia.,Centre for Medicine Use and Safety, Monash University, Melbourne, Victoria 3052, Australia.,Certara, Princeton, New Jersey 08540, USA
| | - Jeffrey K Aronson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| | | | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland.,Division of General Internal Medicine, Geneva University Hospitals, CH-1211 Geneva, Switzerland
| | - Youssef Daali
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Birgit Eiermann
- Inera AB, Swedish Association of Local Authorities and Regions, SE-118 93 Stockholm, Sweden
| | - Jack Cook
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut 06340, USA
| | - Lawrence Lesko
- Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida 32827, USA
| | - Andrew J McLachlan
- School of Pharmacy, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Amin Rostami-Hodjegan
- Certara, Princeton, New Jersey 08540, USA.,Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
33
|
Terrier J, Daali Y, Fontana P, Csajka C, Reny JL. Towards Personalized Antithrombotic Treatments: Focus on P2Y 12 Inhibitors and Direct Oral Anticoagulants. Clin Pharmacokinet 2020; 58:1517-1532. [PMID: 31250210 DOI: 10.1007/s40262-019-00792-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oral anticoagulants and antiplatelet drugs are commonly prescribed to lower the risk of cardiovascular diseases, such as venous and arterial thrombosis, which represent the leading causes of mortality worldwide. A significant percentage of patients taking antithrombotics will nevertheless experience bleeding or recurrent ischemic events, and this represents a major public health issue. Cardiovascular medicine is now questioning the one-size-fits-all policy, and more personalized approaches are increasingly being considered. However, the available tools are currently limited and they are only moderately able to predict clinical events or have a significant impact on clinical outcomes. Predicting concentrations of antithrombotics in blood could be an effective means of personalization as they have been associated with bleeding and recurrent ischemia. Target concentration interventions could take advantage of physiologically based pharmacokinetic (PBPK) and population-based pharmacokinetic (POPPK) models, which are increasingly used in clinical settings and have attracted the interest of governmental regulatory agencies, to propose dosages adapted to specific population characteristics. These models have the benefit of combining parameters from different sources, such as experimental in vitro data and patients' demographic, genetic, and physiological in vivo data, to characterize the dose-concentration relationships of compounds of interest. As such, they can be used to predict individual drug exposure. In the near future, these models could therefore be a valuable means of predicting personalized antithrombotic blood concentrations and, hopefully, of preventing clinical non-response or bleeding in a given patient. Existing approaches for personalization of antithrombotic prescriptions will be reviewed using practical examples for P2Y12 inhibitors and direct oral anticoagulants. The review will additionally focus on the existing PBPK and POPPK models for these two categories of drugs. Lastly, we address potential scenarios for their implementation in clinics, along with the main limitations and challenges.
Collapse
Affiliation(s)
- Jean Terrier
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Youssef Daali
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care Department, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Chantal Csajka
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Jean-Luc Reny
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland. .,Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Division of Internal Medicine and Rehabilitation, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205, Geneva, Switzerland.
| |
Collapse
|
34
|
Opinion: Imagine 5P3A Global Health. Ther Innov Regul Sci 2020; 54:988-990. [DOI: 10.1007/s43441-020-00118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/14/2019] [Indexed: 10/25/2022]
|
35
|
Martins FS, Zhu P, Heinrichs MT, Sy SKB. Physiologically based pharmacokinetic-pharmacodynamic evaluation of meropenem plus fosfomycin in paediatrics. Br J Clin Pharmacol 2020; 87:1012-1023. [PMID: 32638408 DOI: 10.1111/bcp.14456] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/26/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS The objective of the current study was to evaluate paediatric dosing regimens for meropenem plus fosfomycin that generate sufficient coverage against multidrug-resistant bacteria. METHODS The physiologically based pharmacokinetic (PBPK) models of meropenem and fosfomycin were developed from previously published pharmacokinetic studies in five populations: healthy subjects of Japanese origin, and healthy adults, geriatric, paediatric and renally impaired of primarily Caucasian origins. Pharmacodynamic (PD) analyses were carried out by evaluating dosing regimens that achieved a ≥90% joint probability of target attainment (PTA), which was defined as the minimum of the marginal probabilities to achieve the target PD index of each antibiotic. For meropenem, the percentage of time over a 24-hour period wherein the free drug concentration was above the minimum inhibitory concentration (fT > MIC) of at least 40% was its PD target. The fosfomycin PD index was described by fAUC/MIC of at least 40.8. RESULTS For coadministration consisting of 20 mg/kg meropenem q8h as a 3-hour infusion and 35 mg/kg fosfomycin q8h also as a 3-hour infusion in a virtual paediatric population between 1 month and 12 years of age with normal renal function and a corresponding body weight between 3 and 50 kg, a joint PTA ≥ 90% is achieved at MICs of 16 and 64 mg/L for meropenem and fosfomycin coadministration, respectively, against Klebsiella pneumoniae and Pseudomonas aeruginosa. CONCLUSION The current study identified potentially effective paediatric dosing regimens for meropenem plus fosfomycin coadministration against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Frederico S Martins
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Peijuan Zhu
- Clinical Pharmacology & Pharmacometrics, Janssen Research & Development LLC, Raritan, NJ, USA
| | - M Tobias Heinrichs
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Sherwin K B Sy
- Department of Statistics, State University of Maringá, Paraná, Brazil
| |
Collapse
|
36
|
Geerts H, van der Graaf PH. Salvaging CNS Clinical Trials Halted Due to COVID-19. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:367-370. [PMID: 32468710 PMCID: PMC7283764 DOI: 10.1002/psp4.12535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/22/2020] [Indexed: 01/06/2023]
|
37
|
Wang K, Parrott N, Olivares-Morales A, Dudal S, Singer T, Lavé T, Ribba B. Real-World Data and Physiologically-Based Mechanistic Models for Precision Medicine. Clin Pharmacol Ther 2020; 107:694-696. [PMID: 32090313 DOI: 10.1002/cpt.1780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/20/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Ken Wang
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Andres Olivares-Morales
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Sherri Dudal
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Singer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Thierry Lavé
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Benjamin Ribba
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
38
|
Polasek TM, Rostami-Hodjegan A. Virtual Twins: Understanding the Data Required for Model-Informed Precision Dosing. Clin Pharmacol Ther 2020; 107:742-745. [PMID: 32056199 DOI: 10.1002/cpt.1778] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Thomas M Polasek
- Certara, Princeton, New Jersey, USA
- Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, Australia
- Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Amin Rostami-Hodjegan
- Certara, Princeton, New Jersey, USA
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| |
Collapse
|
39
|
Tylutki Z, Szlęk J, Polak S. CardiacPBPK: A tool for the prediction and visualization of time-concentration profiles of drugs in heart tissue. Comput Biol Med 2019; 115:103484. [PMID: 31606584 DOI: 10.1016/j.compbiomed.2019.103484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND OBJECTIVE Prediction of drug concentration in heart tissue is important in terms of drug safety and efficacy. This work presents the Open-Source CardiacPBPK platform for the prediction of the time-concentration profile of drugs, which could potentially reduce the risk of drug development failure due to cardiotoxicity. The objective of the CardiacPBPK development is to accelerate and simplify the in-silico toxicological assessment of new drugs, and to provide supportive material for the research community to use. METHODS The CardiacPBPK software provides a modular implementation of the PBPK model of heart tissue. It can be easily accessed via the Internet or installed locally. The graphical user interface and tabular design are easy to configure and use. RESULTS CardiacPBPK is a tool designed to predict and visualize the time-concentration profiles of a parent compound, and one metabolite, in venous plasma and heart tissue after oral or intravenous drug administration. CardiacPBPK is built on the R-environment framework and supports shiny application features such as interactive visualization of the results, and web applications interface by default. A shiny application refers to a computer program created with the use of shiny package in R. The application is freely available at https://github.com/jszlek/CardiacPBPK and https://sourceforge.net/projects/cardiacpbpk/. This open-source application runs on all platforms supporting R-environment (Linux, Windows, Mac OS X, Solaris). CONCLUSIONS We demonstrate the application of CardiacPBPK by simulating the study of amitriptyline intoxication in the case of CYP2D6 genetic polymorphism.
Collapse
Affiliation(s)
- Zofia Tylutki
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland; Certara UK - Simcyp Division, Sheffield, UK
| | - Jakub Szlęk
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, Krakow, Poland.
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland; Certara UK - Simcyp Division, Sheffield, UK
| |
Collapse
|
40
|
Metabolic and endocrinal effects of N-desmethyl-olanzapine in mice with obesity: Implication for olanzapine-associated metabolic changes. Psychoneuroendocrinology 2019; 108:163-171. [PMID: 31302499 DOI: 10.1016/j.psyneuen.2019.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 01/01/2023]
Abstract
Clinical use of the antipsychotic drug olanzapine (OLA) is associated with metabolic side effects to variable degrees. N-desmethyl-olanzapine (DMO) is one major metabolite of OLA, but its potential involvement in the metabolic responses remains unclear. Here we examined whether DMO can directly impact the metabolic, endocrinal and inflammatory parameters under conditions of metabolic disturbance. DMO administration (2 mg/kg, i.g.) to high-fat diet induced obesity mice for 4 weeks induced a remarkable loss of body weight and fat mass. DMO improved insulin resistance and energy expenditure in mice, but had no significant effects on dyslipidemia or hepatic steatosis. Moreover, DMO induced morphological changes in the white adipose tissue, accompanied by reduced interleukin-1β (IL-1β) production and increased UCP1 expression. These findings demonstrate that DMO is devoid of the metabolic side effects commonly observed for OLA during obesity, which suggests that the N-desmethyl metabolism may function to regulate the metabolic responses to OLA.
Collapse
|
41
|
Emoto C, Hahn D, Euteneuer JC, Mizuno T, Vinks AA, Fukuda T. Next Challenge From the Variance in Individual Physiologically-Based Pharmacokinetic Model-Predicted to Observed Morphine Concentration in Critically Ill Neonates. Clin Pharmacol Ther 2019; 107:319-320. [PMID: 31513716 DOI: 10.1002/cpt.1607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Chie Emoto
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David Hahn
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joshua C Euteneuer
- Division of Neonatology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Neonatology, Children's Hospital & Medical Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tsuyoshi Fukuda
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Biesdorf C, Martins FS, Sy SKB, Diniz A. Physiologically-based pharmacokinetics of ziprasidone in pregnant women. Br J Clin Pharmacol 2019; 85:914-923. [PMID: 30669177 DOI: 10.1111/bcp.13872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/29/2018] [Accepted: 01/06/2019] [Indexed: 01/19/2023] Open
Abstract
AIMS Pregnancy is associated with physiological changes that alter the pharmacokinetics (PK) of drugs. The aim of this study was to predict the PK of ziprasidone in pregnant women. METHODS A full physiologically-based pharmacokinetic (PBPK) model of ziprasidone was developed and validated for the non-pregnant population (healthy adults, paediatrics, geriatrics), and this was extended to the pregnant state to assess the change in PK profile of ziprasidone throughout pregnancy. RESULTS The PBPK model successfully predicted the ziprasidone disposition in healthy adult volunteers, wherein the predicted and observed AUC, Cmax and tmax were within the fold-difference of 0.94-1.09, 0.89-1.40 and 0.80-1.08, respectively. The paediatric and geriatric population, also showed predicted AUC, Cmax and tmax within a two-fold range of the observed values. The simulated exposure in pregnant women using a p-PBPK model showed no significant difference when compared to non-pregnant women. CONCLUSIONS The PBPK model predicted the impact of physiological changes during pregnancy on PK and exposure of ziprasidone, suggesting that dose adjustment is not necessary in this special population.
Collapse
Affiliation(s)
- Carla Biesdorf
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| | | | - Sherwin K B Sy
- Department of Statistics, State University of Maringá, Maringá, Brazil
| | - Andrea Diniz
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| |
Collapse
|
43
|
Polasek TM, Rostami-Hodjegan A, Yim DS, Jamei M, Lee H, Kimko H, Kim JK, Nguyen PTT, Darwich AS, Shin JG. What Does it Take to Make Model-Informed Precision Dosing Common Practice? Report from the 1st Asian Symposium on Precision Dosing. AAPS JOURNAL 2019; 21:17. [PMID: 30627939 DOI: 10.1208/s12248-018-0286-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022]
Abstract
Model-informed precision dosing (MIPD) is modeling and simulation in healthcare to predict the drug dose for a given patient based on their individual characteristics that is most likely to improve efficacy and/or lower toxicity in comparison to traditional dosing. This paper describes the background and status of MIPD and the activities at the 1st Asian Symposium of Precision Dosing. The theme of the meeting was the question, "What does it take to make MIPD common practice?" Formal presentations highlighted the distinction between genetic and non-genetic sources of variability in drug exposure and response, the use of modeling and simulation as decision support tools, and the facilitators to MIPD implementation. A panel discussion addressed the types of models used for MIPD, how the pharmaceutical industry views MIPD, ways to upscale MIPD beyond academic hospital centers, and the essential role of healthcare professional education as a way to progress. The meeting concluded with an ongoing commitment to use MIPD to improve patient care.
Collapse
Affiliation(s)
- Thomas M Polasek
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA. .,Centre for Medicines Use and Safety, Monash University, Melbourne, Australia.
| | - Amin Rostami-Hodjegan
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA.,Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Dong-Seok Yim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Masoud Jamei
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Holly Kimko
- Janssen Research and Development, Lower Gwynedd Township, Pennsylvania, USA
| | - Jae Kyoung Kim
- Korea Advanced Institute of Advanced Technology, Daedoek Innopolis, Daejeon, South Korea
| | - Phuong Thi Thu Nguyen
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Faculty of Pharmacy, Haiphong University of Medicine and Pharmacy, Haiphong, Vietnam
| | - Adam S Darwich
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Jae-Gook Shin
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
44
|
Polasek TM, Rayner CR, Peck RW, Rowland A, Kimko H, Rostami‐Hodjegan A. Toward Dynamic Prescribing Information: Codevelopment of Companion Model‐Informed Precision Dosing Tools in Drug Development. Clin Pharmacol Drug Dev 2018; 8:418-425. [DOI: 10.1002/cpdd.638] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Thomas M. Polasek
- Certara Princeton NJ USA
- Centre for Medicines Use and SafetyMonash University Melbourne Australia
| | - Craig R. Rayner
- Certara Princeton NJ USA
- Centre for Medicines Use and SafetyMonash University Melbourne Australia
| | - Richard W. Peck
- Pharma Research and Exploratory DevelopmentRoche Innovation Centre Basel Basel Switzerland
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders University Adelaide Australia
| | - Holly Kimko
- Janssen Research and Development Exton PA USA
| | - Amin Rostami‐Hodjegan
- Certara Princeton NJ USA
- Centre for Applied Pharmacokinetic ResearchUniversity of Manchester Manchester UK
| |
Collapse
|
45
|
Tylutki Z, Mendyk A, Polak S. Physiologically based pharmacokinetic-quantitative systems toxicology and safety (PBPK-QSTS) modeling approach applied to predict the variability of amitriptyline pharmacokinetics and cardiac safety in populations and in individuals. J Pharmacokinet Pharmacodyn 2018; 45:663-677. [PMID: 29943290 PMCID: PMC6182726 DOI: 10.1007/s10928-018-9597-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022]
Abstract
The physiologically based pharmacokinetic (PBPK) models allow for predictive assessment of variability in population of interest. One of the future application of PBPK modeling is in the field of precision dosing and personalized medicine. The aim of the study was to develop PBPK model for amitriptyline given orally, predict the variability of cardiac concentrations of amitriptyline and its main metabolite-nortriptyline in populations as well as individuals, and simulate the influence of those xenobiotics in therapeutic and supratherapeutic concentrations on human electrophysiology. The cardiac effect with regard to QT and RR interval lengths was assessed. The Emax model to describe the relationship between amitriptyline concentration and heart rate (RR) length was proposed. The developed PBPK model was used to mimic 29 clinical trials and 19 cases of amitriptyline intoxication. Three clinical trials and 18 cases were simulated with the use of PBPK-QSTS approach, confirming lack of cardiotoxic effect of amitriptyline in therapeutic doses and the increase in heart rate along with potential for arrhythmia development in case of amitriptyline overdose. The results of our study support the validity and feasibility of the PBPK-QSTS modeling development for personalized medicine.
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, Krakow, Poland.
| | - Aleksander Mendyk
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, Medyczna 9 St, 30-688, Krakow, Poland
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688, Krakow, Poland
- Certara-Simcyp, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| |
Collapse
|
46
|
Polasek TM, Shakib S, Rostami-Hodjegan A. Precision dosing in clinical medicine: present and future. Expert Rev Clin Pharmacol 2018; 11:743-746. [PMID: 30010447 DOI: 10.1080/17512433.2018.1501271] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Thomas M Polasek
- a Certara , Princeton , NJ , USA.,b Centre for Medicines Use and Safety , Monash University , Melbourne , Australia
| | - Sepehr Shakib
- c Department of Clinical Pharmacology , University of Adelaide , Adelaide , Australia
| | - Amin Rostami-Hodjegan
- a Certara , Princeton , NJ , USA.,d Centre for Applied Pharmacokinetic Research , University of Manchester , Manchester , UK
| |
Collapse
|
47
|
Eugene AR, Eugene B. An opportunity for clinical pharmacology trained physicians to improve patient drug safety: A retrospective analysis of adverse drug reactions in teenagers. F1000Res 2018; 7:677. [PMID: 30271581 PMCID: PMC6143933 DOI: 10.12688/f1000research.14970.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Adverse drug reactions (ADRs) are a major cause of hospital admissions, prolonged hospital stays, morbidity, and drug-related mortality. In this study, we sought to identify the most frequently reported medications and associated side effects in adolescent-aged patients in an effort to prioritize clinical pharmacology consultation efforts for hospitals seeking to improve patient safety. Methods: Quarterly reported data were obtained from the United States Food and Drug Administration Adverse Events Reporting System (FAERS) from the third quarter of 2014 and ending in the third quarter of 2017. We then used the GeneCards database to map the pharmacogenomic biomarkers associated with the most reported FAERS drugs. Data homogenization and statistics analysis were all conducted in R for statistical programming. Results: We identified risperidone (10.64%) as the compound with the most reported ADRs from all reported cases. Males represented 90.1% of reported risperidone cases with gynecomastia being the most reported ADR. Ibuprofen OR=188 (95% CI, 105.00 – 335.00) and quetiapine fumarate OR=116 (95% CI, 48.40 – 278.00) were associated with the highest odds of completed suicide in teenagers. Ondansetron hydrochloride OR=7.12 (95% CI, 1.59 – 31.9) resulted in the highest odds of pneumothorax. Lastly, olanzapine (8.96%) represented the compound with the most reported drug-drug interactions cases, while valproic acid OR=221 (95% CI, 93.900 – 522.00) was associated with the highest odds of drug-drug interactions. Conclusion: Despite any data limitations, physicians prescribing risperidone in males should be aware of the high rates of adverse drug events and an alternative psychotropic should be considered in male patients. Further, patients with a history of pneumothorax or genetically predisposed to pneumothorax should be considered for an alternative antiemetic to ondansetron hydrochloride, due to increased odds associated with the drug and adverse event.
Collapse
Affiliation(s)
- Andy R Eugene
- Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Inova Center for Personalized Health, Shenandoah University, Fairfax, VA, 22031, USA.,Neurophysiology Unit, Department of Psychiatry, Medical University of Lublin, Aleje Racławickie 1, 20-059 Lublin, Poland
| | - Beata Eugene
- Marie-Curie Sklodowska University, Lublin, Poland
| |
Collapse
|
48
|
Rowland A, van Dyk M, Hopkins AM, Mounzer R, Polasek TM, Rostami-Hodjegan A, Sorich MJ. Physiologically Based Pharmacokinetic Modeling to Identify Physiological and Molecular Characteristics Driving Variability in Drug Exposure. Clin Pharmacol Ther 2018; 104:1219-1228. [DOI: 10.1002/cpt.1076] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Andrew Rowland
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
| | - Madelé van Dyk
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
| | - Ashley M. Hopkins
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
| | - Reham Mounzer
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
| | - Thomas M. Polasek
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
- d3 Medicine, A Certara Company; Melbourne Australia
| | - Amin Rostami-Hodjegan
- Simcyp, A Certara Company; Sheffield UK
- Centre of Applied Pharmacokinetic Research; University of Manchester; Manchester UK
| | - Michael J. Sorich
- Clinical Pharmacology, College of Medicine and Public Health; Flinders University; Adelaide Australia
| |
Collapse
|
49
|
Polasek TM, Tucker GT, Sorich MJ, Wiese MD, Mohan T, Rostami‐Hodjegan A, Korprasertthaworn P, Perera V, Rowland A. Prediction of olanzapine exposure in individual patients using physiologically based pharmacokinetic modelling and simulation. Br J Clin Pharmacol 2018; 84:462-476. [PMID: 29194718 PMCID: PMC5809347 DOI: 10.1111/bcp.13480] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/15/2022] Open
Abstract
AIM The aim of the present study was to predict olanzapine (OLZ) exposure in individual patients using physiologically based pharmacokinetic modelling and simulation (PBPK M&S). METHODS A 'bottom-up' PBPK model for OLZ was constructed in Simcyp® (V14.1) and validated against pharmacokinetic studies and data from therapeutic drug monitoring (TDM). The physiological, demographic and genetic attributes of the 'healthy volunteer population' file in Simcyp® were then individualized to create 'virtual twins' of 14 patients. The predicted systemic exposure of OLZ in virtual twins was compared with measured concentration in corresponding patients. Predicted exposures were used to calculate a hypothetical decrease in exposure variability after OLZ dose adjustment. RESULTS The pharmacokinetic parameters of OLZ from single-dose studies were accurately predicted in healthy Caucasians [mean-fold errors (MFEs) ranged from 0.68 to 1.14], healthy Chinese (MFEs 0.82 to 1.18) and geriatric Caucasians (MFEs 0.55 to 1.30). Cumulative frequency plots of trough OLZ concentration were comparable between the virtual population and patients in a TDM database. After creating virtual twins in Simcyp®, the R2 values for predicted vs. observed trough OLZ concentrations were 0.833 for the full cohort of 14 patients and 0.884 for the 7 patients who had additional cytochrome P450 2C8 genotyping. The variability in OLZ exposure following hypothetical dose adjustment guided by PBPK M&S was twofold lower compared with a fixed-dose regimen - coefficient of variation values were 0.18 and 0.37, respectively. CONCLUSIONS Olanzapine exposure in individual patients was predicted using PBPK M&S. Repurposing of available PBPK M&S platforms is an option for model-informed precision dosing and requires further study to examine clinical potential.
Collapse
Affiliation(s)
- Thomas M. Polasek
- Department of Clinical PharmacologyFlinders UniversityAdelaideSAAustralia
- d3 MedicineA Certara CompanyMelbourneVICAustralia
| | - Geoffrey T. Tucker
- Medicine and Biomedical Sciences (Emeritus)University of SheffieldSheffieldUK
| | - Michael J. Sorich
- Department of Clinical PharmacologyFlinders UniversityAdelaideSAAustralia
- Flinders Centre for Innovation in CancerFlinders UniversityAdelaideSAAustralia
| | - Michael D. Wiese
- School of Pharmacy and Medical SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Titus Mohan
- Department of PsychiatryFlinders Medical CentreAdelaideSAAustralia
| | - Amin Rostami‐Hodjegan
- Certara, Blades Enterprise CentreSheffieldUK
- Centre for Applied Pharmacokinetic ResearchUniversity of ManchesterManchesterUK
| | | | - Vidya Perera
- Clinical Pharmacology and Pharmacometrics, Early Clinical and Translational ResearchBristol Myers SquibbPrincetonNJUSA
| | - Andrew Rowland
- Department of Clinical PharmacologyFlinders UniversityAdelaideSAAustralia
- Flinders Centre for Innovation in CancerFlinders UniversityAdelaideSAAustralia
| |
Collapse
|