151
|
Predictive Performance of Physiologically-Based Pharmacokinetic Models in Predicting Drug–Drug Interactions Involving Enzyme Modulation. Clin Pharmacokinet 2018; 57:1337-1346. [DOI: 10.1007/s40262-018-0635-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
152
|
Shebley M, Sandhu P, Emami Riedmaier A, Jamei M, Narayanan R, Patel A, Peters SA, Reddy VP, Zheng M, de Zwart L, Beneton M, Bouzom F, Chen J, Chen Y, Cleary Y, Collins C, Dickinson GL, Djebli N, Einolf HJ, Gardner I, Huth F, Kazmi F, Khalil F, Lin J, Odinecs A, Patel C, Rong H, Schuck E, Sharma P, Wu SP, Xu Y, Yamazaki S, Yoshida K, Rowland M. Physiologically Based Pharmacokinetic Model Qualification and Reporting Procedures for Regulatory Submissions: A Consortium Perspective. Clin Pharmacol Ther 2018; 104:88-110. [PMID: 29315504 PMCID: PMC6032820 DOI: 10.1002/cpt.1013] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/05/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022]
Abstract
This work provides a perspective on the qualification and verification of physiologically based pharmacokinetic (PBPK) platforms/models intended for regulatory submission based on the collective experience of the Simcyp Consortium members. Examples of regulatory submission of PBPK analyses across various intended applications are presented and discussed. European Medicines Agency (EMA) and US Food and Drug Administration (FDA) recent draft guidelines regarding PBPK analyses and reporting are encouraging, and to advance the use and acceptability of PBPK analyses, more clarity and flexibility are warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ming Zheng
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | | | | | | - Jun Chen
- Sanofi, Région de Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | - Jing Lin
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | | | - Chirag Patel
- Takeda Pharmaceuticals International Co., Cambridge, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Jin Y, Borell H, Gardin A, Ufer M, Huth F, Camenisch G. In vitro studies and in silico predictions of fluconazole and CYP2C9 genetic polymorphism impact on siponimod metabolism and pharmacokinetics. Eur J Clin Pharmacol 2017; 74:455-464. [PMID: 29273968 PMCID: PMC5849655 DOI: 10.1007/s00228-017-2404-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/12/2017] [Indexed: 11/29/2022]
Abstract
Purpose The purpose of the study is to investigate the enzyme(s) responsible for siponimod metabolism and to predict the inhibitory effects of fluconazole as well as the impact of cytochrome P450 (CYP) 2C9 genetic polymorphism on siponimod pharmacokinetics (PK) and metabolism. Methods In vitro metabolism studies were conducted using human liver microsomes (HLM), and enzyme phenotyping was assessed using a correlation analysis method. SimCYP, a physiologically based PK model, was developed and used to predict the effects of fluconazole and CYP2C9 genetic polymorphism on siponimod metabolism. Primary PK parameters were generated using the SimCYP and WinNonlin software. Results Correlation analysis suggested that CYP2C9 is the main enzyme responsible for siponimod metabolism in humans. Compared with the CYP2C9*1/*1 genotype, HLM incubations from CYP2C9*3/*3 and CYP2C9*2/*2 donors showed ~ 10- and 3-fold decrease in siponimod metabolism, respectively. Simulations of enzyme contribution predicted that in the CYP2C9*1/*1 genotype, CYP2C9 is predominantly responsible for siponimod metabolism (~ 81%), whereas in the CYP2C9*3/*3 genotype, its contribution is reduced to 11%. The predicted exposure increase of siponimod with fluconazole 200 mg was 2.0–2.4-fold for CYP2C9*1/*1 genotype. In context of single dosing, the predicted mean area under the curve (AUC) is 2.7-, 3.0- and 4.5-fold higher in the CYP2C9*2/*2, CYP2C9*2/*3 and CYP2C9*3/*3 genotypes, respectively, compared with the CYP2C9*1/*1 genotype. Conclusion .Enzyme phenotyping with correlation analysis confirmed the predominant role of CYP2C9 in the biotransformation of siponimod and demonstrated the functional consequence of CYP2C9 genetic polymorphism on siponimod metabolism. Simulation of fluconazole inhibition closely predicted a 2-fold AUC change (ratio within ~ 20% deviation) to the observed value. In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes. Electronic supplementary material The online version of this article (10.1007/s00228-017-2404-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yi Jin
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland.
| | - Hubert Borell
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland
| | - Anne Gardin
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland
| | - Mike Ufer
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland
| | - Felix Huth
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland
| | - Gian Camenisch
- PK Sciences, Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 14, 202.3, CH-4002, Basel, Switzerland
| |
Collapse
|
154
|
Samant TS, Dhuria S, Lu Y, Laisney M, Yang S, Grandeury A, Mueller‐Zsigmondy M, Umehara K, Huth F, Miller M, Germa C, Elmeliegy M. Ribociclib Bioavailability Is Not Affected by Gastric pH Changes or Food Intake: In Silico and Clinical Evaluations. Clin Pharmacol Ther 2017; 104:374-383. [PMID: 29134635 PMCID: PMC6099197 DOI: 10.1002/cpt.940] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/30/2017] [Accepted: 11/05/2017] [Indexed: 02/06/2023]
Abstract
Ribociclib (KISQALI), a cyclin‐dependent kinase 4/6 inhibitor approved for the first‐line treatment of HR+/HER2– advanced breast cancer with an aromatase inhibitor, is administered with no restrictions on concomitant gastric pH‐elevating agents or food intake. The influence of proton pump inhibitors (PPIs) on ribociclib bioavailability was assessed using 1) biorelevant media solubility, 2) physiologically based pharmacokinetic (PBPK) modeling, 3) noncompartmental analysis (NCA) of clinical trial data, and 4) population PK (PopPK) analysis. This multipronged approach indicated no effect of gastric pH changes on ribociclib PK and served as a platform for supporting ribociclib labeling language, stating no impact of gastric pH‐altering agents on the absorption of ribociclib, without a dedicated drug–drug interaction trial. The bioequivalence of ribociclib exposure with or without a high‐fat meal was demonstrated in a clinical trial. Lack of restrictions on ribociclib dosing may facilitate better patient compliance and therefore clinical benefit.
Collapse
Affiliation(s)
| | | | - Yasong Lu
- Novartis PharmaceuticalsEast HanoverNew JerseyUSA
| | | | - Shu Yang
- Novartis PharmaceuticalsEast HanoverNew JerseyUSA
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Bell SM, Chang X, Wambaugh JF, Allen DG, Bartels M, Brouwer KLR, Casey WM, Choksi N, Ferguson SS, Fraczkiewicz G, Jarabek AM, Ke A, Lumen A, Lynn SG, Paini A, Price PS, Ring C, Simon TW, Sipes NS, Sprankle CS, Strickland J, Troutman J, Wetmore BA, Kleinstreuer NC. In vitro to in vivo extrapolation for high throughput prioritization and decision making. Toxicol In Vitro 2017; 47:213-227. [PMID: 29203341 DOI: 10.1016/j.tiv.2017.11.016] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/10/2023]
Abstract
In vitro chemical safety testing methods offer the potential for efficient and economical tools to provide relevant assessments of human health risk. To realize this potential, methods are needed to relate in vitro effects to in vivo responses, i.e., in vitro to in vivo extrapolation (IVIVE). Currently available IVIVE approaches need to be refined before they can be utilized for regulatory decision-making. To explore the capabilities and limitations of IVIVE within this context, the U.S. Environmental Protection Agency Office of Research and Development and the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods co-organized a workshop and webinar series. Here, we integrate content from the webinars and workshop to discuss activities and resources that would promote inclusion of IVIVE in regulatory decision-making. We discuss properties of models that successfully generate predictions of in vivo doses from effective in vitro concentration, including the experimental systems that provide input parameters for these models, areas of success, and areas for improvement to reduce model uncertainty. Finally, we provide case studies on the uses of IVIVE in safety assessments, which highlight the respective differences, information requirements, and outcomes across various approaches when applied for decision-making.
Collapse
Affiliation(s)
- Shannon M Bell
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | - Xiaoqing Chang
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | - John F Wambaugh
- U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27709, USA.
| | - David G Allen
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | | | - Kim L R Brouwer
- UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Campus Box 7569, Chapel Hill, NC 27599, USA.
| | - Warren M Casey
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, NC 27709, USA.
| | - Neepa Choksi
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | - Stephen S Ferguson
- National Toxicology Program, National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, NC 27709, USA.
| | | | - Annie M Jarabek
- U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27709, USA.
| | - Alice Ke
- Simcyp Limited (a Certara company), John Street, Sheffield, S2 4SU, United Kingdom.
| | - Annie Lumen
- National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA.
| | - Scott G Lynn
- U.S. Environmental Protection Agency, William Jefferson Clinton Building, 1200 Pennsylvania Ave. NW, Washington, DC 20460, USA.
| | - Alicia Paini
- European Commission, Joint Research Centre, Directorate Health, Consumers and Reference Materials, Chemical Safety and Alternative Methods Unit incorporating EURL ECVAM, Via E. Fermi 2749, Ispra, Varese 20127, Italy.
| | - Paul S Price
- U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27709, USA.
| | - Caroline Ring
- Oak Ridge Institute for Science and Education, P.O. Box 2008, Oak Ridge, TN 37831, USA.
| | - Ted W Simon
- Ted Simon LLC, 4184 Johnston Road, Winston, GA 30187, USA.
| | - Nisha S Sipes
- National Toxicology Program, National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, NC 27709, USA.
| | - Catherine S Sprankle
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | - Judy Strickland
- Integrated Laboratory Systems, Inc., P.O. Box 13501, Research Triangle Park, NC 27709, USA.
| | - John Troutman
- Central Product Safety, The Procter & Gamble Company, Cincinnati, OH 45202, USA.
| | - Barbara A Wetmore
- ScitoVation LLC, 6 Davis Drive, Research Triangle Park, NC 27709, USA.
| | - Nicole C Kleinstreuer
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
156
|
Li M, Zhao P, Pan Y, Wagner C. Predictive Performance of Physiologically Based Pharmacokinetic Models for the Effect of Food on Oral Drug Absorption: Current Status. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 7:82-89. [PMID: 29168611 PMCID: PMC5824104 DOI: 10.1002/psp4.12260] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Abstract
A comprehensive search in literature and published US Food and Drug Administration reviews was conducted to assess whether physiologically based pharmacokinetic (PBPK) modeling could be prospectively used to predict clinical food effect on oral drug absorption. Among the 48 resulted food effect predictions, ∼50% were predicted within 1.25‐fold of observed, and 75% within 2‐fold. Dissolution rate and precipitation time were commonly optimized parameters when PBPK modeling was not able to capture the food effect. The current work presents a knowledgebase for documenting PBPK experience to predict food effect.
Collapse
Affiliation(s)
- Mengyao Li
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA.,Merck & Co, Inc, Kennilworth, New Jersey, USA
| | - Ping Zhao
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA.,Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Yuzhuo Pan
- Office of Generic Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christian Wagner
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA.,Current affiliation: Merck KGaA, Darmstadt, Germany
| |
Collapse
|
157
|
Marsousi N, Desmeules JA, Rudaz S, Daali Y. Prediction of drug-drug interactions using physiologically-based pharmacokinetic models of CYP450 modulators included in Simcyp software. Biopharm Drug Dispos 2017; 39:3-17. [PMID: 28960401 DOI: 10.1002/bdd.2107] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/28/2017] [Accepted: 09/11/2017] [Indexed: 01/16/2023]
Abstract
In recent years, physiologically based PharmacoKinetic (PBPK) modeling has received growing interest as a useful tool for the assessment of drug pharmacokinetics. It has been demonstrated to be informative and helpful to quantify the modification in drug exposure due to specific physio-pathological conditions, age, genetic polymorphisms, ethnicity and particularly drug-drug interactions (DDIs). In this paper, the prediction success of DDIs involving various cytochrome P450 isoenzyme (CYP) modulators namely ketoconazole (a competitive inhibitor of CYP3A), itraconazole (a competitive inhibitor of CYP3A), clarithromycin (a mechanism-based inhibitor of CYP3A), quinidine (a competitive inhibitor of CYP2D6), paroxetine (a mechanism-based inhibitor of CYP2D6), ciprofloxacin (a competitive inhibitor of CYP1A2), fluconazole (a competitive inhibitor of CYP2C9/2C19) and rifampicin (an inducer of CYP3A) were assessed using Simcyp® software. The aim of this report was to establish confidence in each CYP-specific modulator file so they can be used in the future for the prediction of DDIs involving new victim compounds. Our evaluation of these PBPK models suggested that they can be successfully used to evaluate DDIs in untested scenarios. The only noticeable exception concerned a quinidine inhibitor model that requires further improvement. Additionally, other important aspects such as model validation criteria were discussed.
Collapse
Affiliation(s)
- Niloufar Marsousi
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Switzerland.,School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Switzerland
| | - Jules A Desmeules
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Switzerland.,School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Switzerland.,Swiss Center for Applied Human Toxicology (SCAHT), University of Basel, Switzerland.,Faculty of Medicine, Geneva University, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Switzerland.,Swiss Center for Applied Human Toxicology (SCAHT), University of Basel, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Switzerland.,School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Switzerland.,Swiss Center for Applied Human Toxicology (SCAHT), University of Basel, Switzerland.,Faculty of Medicine, Geneva University, Switzerland
| |
Collapse
|
158
|
Zhou W, Johnson TN, Bui KH, Cheung SYA, Li J, Xu H, Al-Huniti N, Zhou D. Predictive Performance of Physiologically Based Pharmacokinetic (PBPK) Modeling of Drugs Extensively Metabolized by Major Cytochrome P450s in Children. Clin Pharmacol Ther 2017; 104:188-200. [PMID: 29027194 DOI: 10.1002/cpt.905] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
The accuracy of physiologically based pharmacokinetic (PBPK) model prediction in children, especially those younger than 2 years old, has not been systematically evaluated. The aim of this study was to characterize the pediatric predictive performance of the PBPK approach for 10 drugs extensively metabolized by CYP1A2 (theophylline), CYP2C8 (desloratidine, montelukast), CYP2C9 (diclofenac), CYP2C19 (esomeprazole, lansoprazole), CYP2D6 (tramadol), and CYP3A4 (itraconazole, ondansetron, sufentanil). Model performance in children was evaluated by comparing simulated plasma concentration-time profiles with observed clinical results for each drug and age group. PBPK models reasonably predicted the pharmacokinetics of desloratadine, diclofenac, itraconazole, lansoprazole, montelukast, ondansetron, sufentanil, theophylline, and tramadol across all age groups. Collectively, 58 out of 67 predictions were within 2-fold and 43 out of 67 predictions within 1.5-fold of observed values. Developed PBPK models can reasonably predict exposure in children age 1 month and older for an array of predominantly CYP metabolized drugs.
Collapse
Affiliation(s)
- Wangda Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | | | - Khanh H Bui
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - S Y Amy Cheung
- Quantitative Clinical Pharmacology, AstraZeneca, Cambridge, UK
| | - Jianguo Li
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Hongmei Xu
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Nidal Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Diansong Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| |
Collapse
|
159
|
Lu T, Fraczkiewicz G, Salphati L, Budha N, Dalziel G, Smelick GS, Morrissey KM, Davis JD, Jin JY, Ware JA. Combining "Bottom-up" and "Top-down" Approaches to Assess the Impact of Food and Gastric pH on Pictilisib (GDC-0941) Pharmacokinetics. CPT Pharmacometrics Syst Pharmacol 2017; 6:747-755. [PMID: 28748626 PMCID: PMC5702897 DOI: 10.1002/psp4.12228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 08/19/2017] [Accepted: 07/19/2017] [Indexed: 12/27/2022] Open
Abstract
Pictilisib, a weakly basic compound, is an orally administered, potent, and selective pan-inhibitor of phosphatidylinositol 3-kinases for oncology indications. To investigate the significance of high-fat food and gastric pH on pictilisib pharmacokinetics (PK) and enable label recommendations, a dedicated clinical study was conducted in healthy volunteers, whereby both top-down (population PK, PopPK) and bottom-up (physiologically based PK, PBPK) approaches were applied to enhance confidence of recommendation and facilitate the clinical development through scenario simulations. The PopPK model identified food (for absorption rate constant (Ka )) and proton pump inhibitors (PPI, for relative bioavailability (Frel ) and Ka ) as significant covariates. Food and PPI also impacted the variability of Frel . The PBPK model accounted for the supersaturation tendency of pictilisib, and gastric emptying physiology successfully predicted the food and PPI effect on pictilisib absorption. Our research highlights the importance of applying both quantitative approaches to address critical drug development questions.
Collapse
Affiliation(s)
- Tong Lu
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | | | - Laurent Salphati
- Department of Drug Metabolism and PharmacokineticsGenentech IncSouth San FranciscoCaliforniaUSA
| | - Nageshwar Budha
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - Gena Dalziel
- Department of Small Molecule Pharmaceutical SciencesGenentech IncSouth San FranciscoCaliforniaUSA
| | - Gillian S. Smelick
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - Kari M. Morrissey
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - John D. Davis
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - Jin Y. Jin
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - Joseph A. Ware
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| |
Collapse
|
160
|
Huang W, Nakano M, Sager J, Ragueneau-Majlessi I, Isoherranen N. Physiologically Based Pharmacokinetic Model of the CYP2D6 Probe Atomoxetine: Extrapolation to Special Populations and Drug-Drug Interactions. Drug Metab Dispos 2017; 45:1156-1165. [PMID: 28860113 PMCID: PMC5637815 DOI: 10.1124/dmd.117.076455] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/28/2017] [Indexed: 01/18/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling of drug disposition and drug-drug interactions (DDIs) has become a key component of drug development. PBPK modeling has also been considered as an approach to predict drug disposition in special populations. However, whether models developed and validated in healthy populations can be extrapolated to special populations is not well established. The goal of this study was to determine whether a drug-specific PBPK model validated using healthy populations could be used to predict drug disposition in specific populations and in organ impairment patients. A full PBPK model of atomoxetine was developed using a training set of pharmacokinetic (PK) data from CYP2D6 genotyped individuals. The model was validated using drug-specific acceptance criteria and a test set of 14 healthy subject PK studies. Population PBPK models were then challenged by simulating the effects of ethnicity, DDIs, pediatrics, and renal and hepatic impairment on atomoxetine PK. Atomoxetine disposition was successfully predicted in 100% of healthy subject studies, 88% of studies in Asians, 79% of DDI studies, and 100% of pediatric studies. However, the atomoxetine area under the plasma concentration versus time curve (AUC) was overpredicted by 3- to 4-fold in end stage renal disease and hepatic impairment. The results show that validated PBPK models can be extrapolated to different ethnicities, DDIs, and pediatrics but not to renal and hepatic impairment patients, likely due to incomplete understanding of the physiologic changes in these conditions. These results show that systematic modeling efforts can be used to further refine population models to improve the predictive value in this area.
Collapse
Affiliation(s)
- Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Mariko Nakano
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Jennifer Sager
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | | | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
161
|
Prediction of drug–drug interaction potential using physiologically based pharmacokinetic modeling. Arch Pharm Res 2017; 40:1356-1379. [DOI: 10.1007/s12272-017-0976-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
|
162
|
Yang F, Wang B, Liu Z, Xia X, Wang W, Yin D, Sheng L, Li Y. Prediction of a Therapeutic Dose for Buagafuran, a Potent Anxiolytic Agent by Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling Starting from Pharmacokinetics in Rats and Human. Front Pharmacol 2017; 8:683. [PMID: 29066968 PMCID: PMC5641330 DOI: 10.3389/fphar.2017.00683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/13/2017] [Indexed: 01/29/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK)/pharmacodynamic (PD) models can contribute to animal-to-human extrapolation and therapeutic dose predictions. Buagafuran is a novel anxiolytic agent and phase I clinical trials of buagafuran have been completed. In this paper, a potentially effective dose for buagafuran of 30 mg t.i.d. in human was estimated based on the human brain concentration predicted by a PBPK/PD modeling. The software GastroPlusTM was used to build the PBPK/PD model for buagafuran in rat which related the brain tissue concentrations of buagafuran and the times of animals entering the open arms in the pharmacological model of elevated plus-maze. Buagafuran concentrations in human plasma were fitted and brain tissue concentrations were predicted by using a human PBPK model in which the predicted plasma profiles were in good agreement with observations. The results provided supportive data for the rational use of buagafuran in clinic.
Collapse
Affiliation(s)
- Fen Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Center of Drug Clinical Trial, Peking University Cancer Hospital and Institute, Beijing, China.,Clinical Pharmacology Research Center, Peking Union Medical College Hospital and Chinese Academy of Medical Sciences, Beijing, China
| | - Baolian Wang
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihao Liu
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuejun Xia
- Department of Drug Delivery System, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijun Wang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dali Yin
- Department of Synthetic Medicinal Chemistry, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Sheng
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
163
|
Wiśniowska B, Tylutki Z, Polak S. Humans Vary, So Cardiac Models Should Account for That Too! Front Physiol 2017; 8:700. [PMID: 28983251 PMCID: PMC5613127 DOI: 10.3389/fphys.2017.00700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/30/2017] [Indexed: 12/25/2022] Open
Abstract
The utilization of mathematical modeling and simulation in drug development encompasses multiple mathematical techniques and the location of a drug candidate in the development pipeline. Historically speaking they have been used to analyze experimental data (i.e., Hill equation) and clarify the involved physical and chemical processes (i.e., Fick laws and drug molecule diffusion). In recent years the advanced utilization of mathematical modeling has been an important part of the regulatory review process. Physiologically based pharmacokinetic (PBPK) models identify the need to conduct specific clinical studies, suggest specific study designs and propose appropriate labeling language. Their application allows the evaluation of the influence of intrinsic (e.g., age, gender, genetics, disease) and extrinsic [e.g., dosing schedule, drug-drug interactions (DDIs)] factors, alone or in combinations, on drug exposure and therefore provides accurate population assessment. A similar pathway has been taken for the assessment of drug safety with cardiac safety being one the most advanced examples. Mechanistic mathematical model-informed safety evaluation, with a focus on drug potential for causing arrhythmias, is now discussed as an element of the Comprehensive in vitro Proarrhythmia Assay. One of the pillars of this paradigm is the use of an in silico model of the adult human ventricular cardiomyocyte to integrate in vitro measured data. Existing examples (in vitro—in vivo extrapolation with the use of PBPK models) suggest that deterministic, epidemiological and clinical data based variability models can be merged with the mechanistic models describing human physiology. There are other methods available, based on the stochastic approach and on population of models generated by randomly assigning specific parameter values (ionic current conductance and kinetic) and further pruning. Both approaches are briefly characterized in this manuscript, in parallel with the drug-specific variability.
Collapse
Affiliation(s)
- Barbara Wiśniowska
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical CollegeKrakow, Poland
| | - Zofia Tylutki
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical CollegeKrakow, Poland
| | - Sebastian Polak
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical CollegeKrakow, Poland.,SimcypCertara, Sheffield, United Kingdom
| |
Collapse
|
164
|
Marsousi N, Desmeules JA, Rudaz S, Daali Y. Usefulness of PBPK Modeling in Incorporation of Clinical Conditions in Personalized Medicine. J Pharm Sci 2017; 106:2380-2391. [DOI: 10.1016/j.xphs.2017.04.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/14/2022]
|
165
|
Paini A, Leonard JA, Kliment T, Tan YM, Worth A. Investigating the state of physiologically based kinetic modelling practices and challenges associated with gaining regulatory acceptance of model applications. Regul Toxicol Pharmacol 2017; 90:104-115. [PMID: 28866268 PMCID: PMC5656087 DOI: 10.1016/j.yrtph.2017.08.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 01/14/2023]
Abstract
Physiologically based kinetic (PBK) models are used widely throughout a number of working sectors, including academia and industry, to provide insight into the dosimetry related to observed adverse health effects in humans and other species. Use of these models has increased over the last several decades, especially in conjunction with emerging alternative methods to animal testing, such as in vitro studies and data-driven in silico quantitative-structure-activity-relationship (QSAR) predictions. Experimental information derived from these new approach methods can be used as input for model parameters and allows for increased confidence in models for chemicals that did not have in vivo data for model calibration. Despite significant advancements in good modelling practice (GMP) for model development and evaluation, there remains some reluctance among regulatory agencies to use such models during the risk assessment process. Here, the results of a survey disseminated to the modelling community are presented in order to inform the frequency of use and applications of PBK models in science and regulatory submission. Additionally, the survey was designed to identify a network of investigators involved in PBK modelling and knowledgeable of GMP so that they might be contacted in the future for peer review of PBK models, especially in regards to vetting the models to such a degree as to gain a greater acceptance for regulatory purposes. Physiologically Based kinetic (PBK) models are used widely in academia, industry, and government. Good modelling practice (GMP) for model development and evaluation continues to expand. Further guidance for establishing GMP is called for. There remains some reluctance among regulatory agencies to use PBK models. The next generation of PBK models could be developed using only data from in vitro and in silico methods.
Collapse
Affiliation(s)
- Alicia Paini
- European Commission, Joint Research Centre, Directorate Health, Consumers and Reference Materials, Via E Fermi 2749, 21027 Ispra, Italy.
| | - Jeremy A Leonard
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA
| | | | - Yu-Mei Tan
- U.S. Environmental Protection Agency, National Research Laboratory, Research Triangle Park, NC 27709, USA
| | - Andrew Worth
- European Commission, Joint Research Centre, Directorate Health, Consumers and Reference Materials, Via E Fermi 2749, 21027 Ispra, Italy
| |
Collapse
|
166
|
Clinical Drug–Drug Interaction Evaluations to Inform Drug Use and Enable Drug Access. J Pharm Sci 2017; 106:2214-2218. [DOI: 10.1016/j.xphs.2017.04.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 11/18/2022]
|
167
|
Yoshida K, Zhao P, Zhang L, Abernethy DR, Rekić D, Reynolds KS, Galetin A, Huang SM. In Vitro–In Vivo Extrapolation of Metabolism- and Transporter-Mediated Drug–Drug Interactions—Overview of Basic Prediction Methods. J Pharm Sci 2017; 106:2209-2213. [DOI: 10.1016/j.xphs.2017.04.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 10/19/2022]
|
168
|
Pathak SM, Ruff A, Kostewicz ES, Patel N, Turner DB, Jamei M. Model-Based Analysis of Biopharmaceutic Experiments To Improve Mechanistic Oral Absorption Modeling: An Integrated in Vitro in Vivo Extrapolation Perspective Using Ketoconazole as a Model Drug. Mol Pharm 2017; 14:4305-4320. [DOI: 10.1021/acs.molpharmaceut.7b00406] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Shriram M. Pathak
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, United Kingdom
| | - Aaron Ruff
- Department
of Pharmaceutical Technology, Johann Wolfgang Goethe University, Max-von-Laue-Strasse
9, Frankfurt am Main 60438, Germany
| | - Edmund S. Kostewicz
- Department
of Pharmaceutical Technology, Johann Wolfgang Goethe University, Max-von-Laue-Strasse
9, Frankfurt am Main 60438, Germany
| | - Nikunjkumar Patel
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, United Kingdom
| | - David B. Turner
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, United Kingdom
| | - Masoud Jamei
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU, United Kingdom
| |
Collapse
|
169
|
Pan Y, Hsu V, Grimstein M, Zhang L, Arya V, Sinha V, Grillo JA, Zhao P. The Application of Physiologically Based Pharmacokinetic Modeling to Predict the Role of Drug Transporters: Scientific and Regulatory Perspectives. J Clin Pharmacol 2017; 56 Suppl 7:S122-31. [PMID: 27385170 DOI: 10.1002/jcph.740] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 01/24/2023]
Abstract
Transporters play an important role in drug absorption, disposition, and drug action. The evaluation of drug transporters requires a comprehensive understanding of transporter biology and pharmacology. Physiologically based pharmacokinetic (PBPK) models may offer an integrative platform to quantitatively evaluate the role of drug transporters and its interplay with other drug disposition processes such as passive drug diffusion and elimination by metabolizing enzymes. To date, PBPK modeling and simulations integrating drug transporters lag behind that for drug-metabolizing enzymes. In addition, predictive performance of PBPK has not been well established for predicting the role of drug transporters in the pharmacokinetics of a drug. To enhance overall predictive performance of transporter-based PBPK models, it is necessary to have a detailed understanding of transporter biology for proper representation in the models and to have a quantitative understanding of the contribution of transporters in the absorption and metabolism of a drug. This article summarizes PBPK-based submissions evaluating the role of drug transporters to the Office of Clinical Pharmacology of the US Food and Drug Administration.
Collapse
Affiliation(s)
- Yuzhuo Pan
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.,Current affiliation: Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Vicky Hsu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Manuela Grimstein
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Vikram Arya
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Vikram Sinha
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Joseph A Grillo
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ping Zhao
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
170
|
Lee SC, Arya V, Yang X, Volpe DA, Zhang L. Evaluation of transporters in drug development: Current status and contemporary issues. Adv Drug Deliv Rev 2017; 116:100-118. [PMID: 28760687 DOI: 10.1016/j.addr.2017.07.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 01/22/2023]
Abstract
Transporters govern the access of molecules to cells or their exit from cells, thereby controlling the overall distribution of drugs to their intracellular site of action. Clinically relevant drug-drug interactions mediated by transporters are of increasing interest in drug development. Drug transporters, acting alone or in concert with drug metabolizing enzymes, can play an important role in modulating drug absorption, distribution, metabolism and excretion, thus affecting the pharmacokinetics and/or pharmacodynamics of a drug. The drug interaction guidance documents from regulatory agencies include various decision criteria that may be used to predict the need for in vivo assessment of transporter-mediated drug-drug interactions. Regulatory science research continues to assess the prediction performances of various criteria as well as to examine the strength and limitations of each prediction criterion to foster discussions related to harmonized decision criteria that may be used to facilitate global drug development. This review discusses the role of transporters in drug development with a focus on methodologies in assessing transporter-mediated drug-drug interactions, challenges in both in vitro and in vivo assessments of transporters, and emerging transporter research areas including biomarkers, assessment of tissue concentrations, and effect of diseases on transporters.
Collapse
Affiliation(s)
- Sue-Chih Lee
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Vikram Arya
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Donna A Volpe
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
171
|
Wagner C, Zhao P, Arya V, Mullick C, Struble K, Au S. Physiologically Based Pharmacokinetic Modeling for Predicting the Effect of Intrinsic and Extrinsic Factors on Darunavir or Lopinavir Exposure Coadministered With Ritonavir. J Clin Pharmacol 2017; 57:1295-1304. [PMID: 28569994 DOI: 10.1002/jcph.936] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/10/2017] [Indexed: 11/11/2022]
Abstract
Management of comorbidities and medications is complex in HIV-1-infected patients. The overall objective of this project was to develop separate physiologically based pharmacokinetic (PBPK) substrate models for the protease inhibitors darunavir and lopinavir. These protease inhibitors are used in the treatment of HIV infection. Both darunavir and lopinavir are coadministered with another medication that inhibits cytochrome (CYP) 3A. The current project focused on PBPK modeling for darunavir and lopinavir coadministered with ritonavir. Darunavir and lopinavir PBPK models that accounted for ritonavir CYP3A inhibition effects (linked PBPK models) were developed. The linked PBPK models were then used to predict the effect on darunavir or lopinavir exposure from CYP modulators. In the next step, the predicted effect of hepatic impairment was evaluated. Additional exploratory analyses predicted CYP3A inhibition effects on darunavir or lopinavir exposure in simulated hepatically impaired subjects. The linked PBPK models reasonably predicted darunavir or lopinavir exposure based on simulations with CYP inhibitors or inducers. Exploratory simulations using the linked darunavir or lopinavir PBPK models indicated CYP3A inhibition may further increase darunavir or lopinavir exposure in patients with hepatic impairment.
Collapse
Affiliation(s)
- Christian Wagner
- Division of Clinical Pharmacology IV, Office of Clinical Pharmacology, Office of Translational Sciences, CDER, FDA, Silver Spring, MD, USA
| | - Ping Zhao
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, CDER, FDA, Silver Spring, MD, USA
| | - Vikram Arya
- Division of Clinical Pharmacology IV, Office of Clinical Pharmacology, Office of Translational Sciences, CDER, FDA, Silver Spring, MD, USA
| | - Charu Mullick
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, CDER, FDA, Silver Spring, MD, USA
| | - Kimberly Struble
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, CDER, FDA, Silver Spring, MD, USA
| | - Stanley Au
- Division of Clinical Pharmacology IV, Office of Clinical Pharmacology, Office of Translational Sciences, CDER, FDA, Silver Spring, MD, USA
| |
Collapse
|
172
|
Sato M, Ochiai Y, Kijima S, Nagai N, Ando Y, Shikano M, Nomura Y. Quantitative Modeling and Simulation in PMDA: A Japanese Regulatory Perspective. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:413-415. [PMID: 28568566 PMCID: PMC5529733 DOI: 10.1002/psp4.12203] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/25/2022]
Abstract
In Japan in October 2016, the Pharmaceuticals and Medical Devices Agency (PMDA) began to receive electronic data in new drug applications (NDAs). These electronic data are useful to conduct regulatory assessment of sponsors’ submissions and contribute to the PMDA's research. In this article, we summarize the number of submissions of quantitative modeling and simulation (M&S) documents in NDAs in Japan, and we describe our current thinking and activities about quantitative M&S in PMDA.
Collapse
Affiliation(s)
- M Sato
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - Y Ochiai
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - S Kijima
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - N Nagai
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - Y Ando
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - M Shikano
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| | - Y Nomura
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo, Japan
| |
Collapse
|
173
|
Olafuyi O, Coleman M, Badhan RKS. Development of a paediatric physiologically based pharmacokinetic model to assess the impact of drug-drug interactions in tuberculosis co-infected malaria subjects: A case study with artemether-lumefantrine and the CYP3A4-inducer rifampicin. Eur J Pharm Sci 2017; 106:20-33. [PMID: 28546104 DOI: 10.1016/j.ejps.2017.05.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/27/2017] [Accepted: 05/20/2017] [Indexed: 12/13/2022]
Abstract
The fixed dosed combination of artemether and lumefantrine (AL) is widely used for the treatment of malaria in adults and children in sub-Sahara Africa, with lumefantrine day 7 concentrations being widely used as a marker for clinical efficacy. Both are substrates for CYP3A4 and susceptible to drug-drug interactions (DDIs); indeed, knowledge of the impact of these factors is currently sparse in paediatric population groups. Confounding malaria treatment is the co-infection of patients with tuberculosis. The concomitant treatment of AL with tuberculosis chemotherapy, which includes the CYP3A4 inducer rifampicin, increases the risk of parasite recrudescence and malaria treatment failure. This study developed a population-based PBPK model for AL in adults capable of predicting the pharmacokinetics of AL under non-DDI and DDI conditions, as well as predicting AL pharmacokinetics in paediatrics of 2-12years of age. The validated model was utilised to assess the concomitant treatment of rifampicin and lumefantrine under standard body-weight based treatment regimens for 2-5year olds, and demonstrated that no subjects attained the target day 7 concentration (Cd7) of 280ng/mL, highlighting the importance of this DDI and the potential risk of malaria-TB based DDIs. An adapted 7-day treatment regimen was simulated and resulted in 63% and 74.5% of subjects attaining the target Cd7 for 1-tablet and 2-tablet regimens respectively.
Collapse
Affiliation(s)
- Olusola Olafuyi
- Aston Healthy Research Group, Aston Pharmacy School, Aston University, Birmingham B4 7ET, United Kingdom
| | - Michael Coleman
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, United Kingdom
| | - Raj K S Badhan
- Aston Healthy Research Group, Aston Pharmacy School, Aston University, Birmingham B4 7ET, United Kingdom; Aston Pharmacy School, Aston University, Birmingham B4 7ET, United Kingdom.
| |
Collapse
|
174
|
Samant TS, Lukacova V, Schmidt S. Development and Qualification of Physiologically Based Pharmacokinetic Models for Drugs With Atypical Distribution Behavior: A Desipramine Case Study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:315-321. [PMID: 28398693 PMCID: PMC5697013 DOI: 10.1002/psp4.12180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/04/2022]
Abstract
Desipramine is a secondary tricyclic amine, which is primarily metabolized by cytochrome 2D6. It shows a high volume of distribution (Vss) (10–50 L/kg) due to its high lipophilicity, unspecific phospholipid binding, and lysosomal trapping. The objective of this study was to develop and qualify a physiologically based pharmacokinetic (PBPK) model for desipramine, which accounts for the high Vss of the drug following intravenous and oral administration of doses up to 100 mg. The model also accounts for the extended time to reach maximum concentration after oral dosing due to enterocyte trapping. Once developed and qualified in adults, we characterized the dynamic changes in metabolism and pharmacokinetics of desipramine after birth by scaling the system‐specific parameters of the model from adults to pediatrics. The developed modeling strategy provides a prototypical workflow that can also be applied to other drugs with similar properties and a high volume of distribution.
Collapse
Affiliation(s)
- T S Samant
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Lake Nona (Orlando), Florida, USA
| | - V Lukacova
- Simulations Plus, Inc., Lancaster, California, USA
| | - S Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Lake Nona (Orlando), Florida, USA
| |
Collapse
|
175
|
Galetin A, Zhao P, Huang SM. Physiologically Based Pharmacokinetic Modeling of Drug Transporters to Facilitate Individualized Dose Prediction. J Pharm Sci 2017; 106:2204-2208. [PMID: 28390843 DOI: 10.1016/j.xphs.2017.03.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 01/12/2023]
Abstract
Physiologically based pharmacokinetic modeling is a commonly used strategy in the drug development and regulatory submissions. This commentary provides a critical overview of the current status of physiologically based pharmacokinetic methodologies to predict transporter-mediated pharmacokinetics, in addition to the impact of disease and genetics with respect to local and systemic concentration.
Collapse
Affiliation(s)
- Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK; Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20993.
| | - Ping Zhao
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20993
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20993
| |
Collapse
|
176
|
Darwich AS, Ogungbenro K, Vinks AA, Powell JR, Reny JL, Marsousi N, Daali Y, Fairman D, Cook J, Lesko LJ, McCune JS, Knibbe CAJ, de Wildt SN, Leeder JS, Neely M, Zuppa AF, Vicini P, Aarons L, Johnson TN, Boiani J, Rostami-Hodjegan A. Why Has Model-Informed Precision Dosing Not Yet Become Common Clinical Reality? Lessons From the Past and a Roadmap for the Future. Clin Pharmacol Ther 2017; 101:646-656. [DOI: 10.1002/cpt.659] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/17/2022]
Affiliation(s)
- A S Darwich
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry; University of Manchester; Manchester UK
| | - K Ogungbenro
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry; University of Manchester; Manchester UK
| | - A A Vinks
- Cincinnati Children's Hospital Medical Center; Cincinnati Ohio USA
- Department of Pediatrics; University of Cincinnati School of medicine; Cincinnati Ohio USA
| | - J R Powell
- Eshelman School of Pharmacy; University of North Carolina; Chapel Hill North Carolina USA
| | - J-L Reny
- Geneva Platelet Group, School of Medicine; University of Geneva; Geneva Switzerland
- Department of Internal Medicine, Rehabilitation and Geriatrics; Geneva University Hospitals; Geneva Switzerland
| | - N Marsousi
- Clinical Pharmacology and Toxicology; Geneva University Hospitals; Geneva Switzerland
| | - Y Daali
- Geneva Platelet Group, School of Medicine; University of Geneva; Geneva Switzerland
- Clinical Pharmacology and Toxicology; Geneva University Hospitals; Geneva Switzerland
| | - D Fairman
- Clinical Pharmacology Modeling and Simulation, GSK Stevenage; UK
| | - J Cook
- Clinical Pharmacology, Pfizer Inc; Groton Connecticut USA
| | - L J Lesko
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology; University of Florida at Lake Nona (Orlando); Orlando Florida USA
| | - J S McCune
- University of Washington Department of Pharmaceutics and Fred Hitchinson Cancer Research Center Clinical Research Division; Seattle Washington USA
| | - C A J Knibbe
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands and Division of Pharmacology, Leiden Academic Centre for Drug Research; Leiden University; the Netherlands
| | - S N de Wildt
- Department of Pharmacology and Toxicology; Radboud University; Nijmegen the Netherlands
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital; Rotterdam the Netherlands
| | - J S Leeder
- Division of Pediatric Pharmacology and Medical Toxicology, Department of Pediatrics, Children's Mercy Hospitals and Clinics; Kansas City Missouri USA
- Department of Pharmacology; University of Missouri-Kansas City; Kansas City Missouri USA
| | - M Neely
- University of Southern California and the Children's Hospital of Los Angeles; Los Angeles California USA
| | - A F Zuppa
- Children's Hospital of Philadelphia; Philadelphia Pennsylvania USA
| | - P Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK, MedImmune; Cambridge UK
| | - L Aarons
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry; University of Manchester; Manchester UK
| | - T N Johnson
- Certara, Blades Enterprise Centre; Sheffield UK
| | - J Boiani
- Epstein Becker & Green; Washington DC USA
| | - A Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry; University of Manchester; Manchester UK
- Epstein Becker & Green; Washington DC USA
| |
Collapse
|
177
|
Yoshida K, Budha N, Jin JY. Impact of physiologically based pharmacokinetic models on regulatory reviews and product labels: Frequent utilization in the field of oncology. Clin Pharmacol Ther 2017; 101:597-602. [PMID: 28074611 PMCID: PMC5414891 DOI: 10.1002/cpt.622] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/27/2016] [Accepted: 01/02/2017] [Indexed: 02/04/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling can be used to predict drug pharmacokinetics in virtual populations using models that integrate understanding of physiological systems. PBPK models have been widely utilized for predicting pharmacokinetics in clinically untested scenarios during drug applications and regulatory reviews in recent years. Here, we provide a comprehensive review of the application of PBPK in new drug application (NDA) review documents from the US Food and Drug Administration (FDA) in the past 4 years.
Collapse
Affiliation(s)
- K Yoshida
- Department of Clinical PharmacologyGenentech Research and Early DevelopmentSouth San FranciscoCaliforniaUSA
| | - N Budha
- Department of Clinical PharmacologyGenentech Research and Early DevelopmentSouth San FranciscoCaliforniaUSA
| | - JY Jin
- Department of Clinical PharmacologyGenentech Research and Early DevelopmentSouth San FranciscoCaliforniaUSA
| |
Collapse
|
178
|
Ge S, Wei Y, Yin T, Xu B, Gao S, Hu M. Transport–Glucuronidation Classification System and PBPK Modeling: New Approach To Predict the Impact of Transporters on Disposition of Glucuronides. Mol Pharm 2017; 14:2884-2898. [DOI: 10.1021/acs.molpharmaceut.6b00941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shufan Ge
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Yingjie Wei
- Key
Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, 100 Shizi Street, Nanjing 210028, China
| | - Taijun Yin
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Beibei Xu
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Song Gao
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| | - Ming Hu
- Department
of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 1441 Moursund Street, Houston, Texas 77030, United States
| |
Collapse
|
179
|
Yamamoto Y, Danhof M, de Lange ECM. Microdialysis: the Key to Physiologically Based Model Prediction of Human CNS Target Site Concentrations. AAPS JOURNAL 2017; 19:891-909. [DOI: 10.1208/s12248-017-0050-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/25/2017] [Indexed: 01/03/2023]
|
180
|
Scotcher D, Jones CR, Galetin A, Rostami-Hodjegan A. Delineating the Role of Various Factors in Renal Disposition of Digoxin through Application of Physiologically Based Kidney Model to Renal Impairment Populations. J Pharmacol Exp Ther 2017; 360:484-495. [PMID: 28057840 PMCID: PMC5370399 DOI: 10.1124/jpet.116.237438] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
Development of submodels of organs within physiologically-based pharmacokinetic (PBPK) principles and beyond simple perfusion limitations may be challenging because of underdeveloped in vitro-in vivo extrapolation approaches or lack of suitable clinical data for model refinement. However, advantage of such models in predicting clinical observations in divergent patient groups is now commonly acknowledged. Mechanistic understanding of altered renal secretion in renal impairment is one area that may benefit from such models, despite knowledge gaps in renal pathophysiology. In the current study, a PBPK kidney model was developed for digoxin, accounting for the roles of organic anion transporting peptide 4C1 (OATP4C1) and P-glycoprotein (P-gp) in its tubular secretion, with the aim to investigate the impact of age and renal impairment (moderate to severe) on renal drug disposition. Initial PBPK simulations based on changes in glomerular filtration rate (GFR) underestimated the observed reduction in digoxin renal excretion clearance (CLR) in subjects with moderately impaired renal function relative to healthy. Reduction in either proximal tubule cell number or the OATP4C1 abundance in the mechanistic kidney model successfully predicted 59% decrease in digoxin CLR, in particular when these changes were proportional to reduction in GFR. In contrast, predicted proximal tubule concentration of digoxin was only sensitive to changes in the transporter expression/ million proximal tubule cells. Based on the mechanistic modeling, reduced proximal tubule cellularity and OATP4C1 abundance, and inhibition of OATP4C1-mediated transport, are proposed as possible causes of reduced digoxin renal secretion in renally impaired patients.
Collapse
Affiliation(s)
- Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (D.S., A.G., A.R.-H.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, United Kingdom (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, United Kingdom (A.R.-H.)
| | - Christopher R Jones
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (D.S., A.G., A.R.-H.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, United Kingdom (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, United Kingdom (A.R.-H.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (D.S., A.G., A.R.-H.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, United Kingdom (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, United Kingdom (A.R.-H.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (D.S., A.G., A.R.-H.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, United Kingdom (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, United Kingdom (A.R.-H.)
| |
Collapse
|
181
|
Zhao P. Report from the EMA workshop on qualification and reporting of physiologically based pharmacokinetic (PBPK) modeling and simulation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:71-72. [PMID: 28035755 PMCID: PMC5321806 DOI: 10.1002/psp4.12166] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 11/29/2022]
Abstract
On Nov 21, 2016, the European Medicines Agency (EMA) hosted a workshop to discuss its draft guideline on qualification and reporting of physiologically based pharmacokinetic (PBPK) analysis.1 Published on July 21, 2016, the draft PBPK guideline is currently under the period of public comments.
Collapse
Affiliation(s)
- P Zhao
- US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
182
|
Evaluation of Drug-Drug Interaction Potential Between Sacubitril/Valsartan (LCZ696) and Statins Using a Physiologically Based Pharmacokinetic Model. J Pharm Sci 2017; 106:1439-1451. [PMID: 28089685 DOI: 10.1016/j.xphs.2017.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/23/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023]
Abstract
Sacubitril/valsartan (LCZ696) has been approved for the treatment of heart failure. Sacubitril is an in vitro inhibitor of organic anion-transporting polypeptides (OATPs). In clinical studies, LCZ696 increased atorvastatin Cmax by 1.7-fold and area under the plasma concentration-time curve by 1.3-fold, but had little or no effect on simvastatin or simvastatin acid exposure. A physiologically based pharmacokinetics modeling approach was applied to explore the underlying mechanisms behind the statin-specific LCZ696 drug interaction observations. The model incorporated OATP-mediated clearance (CLint,T) for simvastatin and simvastatin acid to successfully describe the pharmacokinetic profiles of either analyte in the absence or presence of LCZ696. Moreover, the model successfully described the clinically observed drug effect with atorvastatin. The simulations clarified the critical parameters responsible for the observation of a low, yet clinically relevant, drug-drug interaction DDI between sacubitril and atorvastatin and the lack of effect with simvastatin acid. Atorvastatin is administered in its active form and rapidly achieves Cmax that coincide with the low Cmax of sacubitril. In contrast, simvastatin requires a hydrolysis step to the acid form and therefore is not present at the site of interactions at sacubitril concentrations that are inhibitory. Similar models were used to evaluate the drug-drug interaction risk for additional OATP-transported statins which predicted to maximally result in a 1.5-fold exposure increase.
Collapse
|
183
|
Tylutki Z, Polak S. A four-compartment PBPK heart model accounting for cardiac metabolism - model development and application. Sci Rep 2017; 7:39494. [PMID: 28051093 PMCID: PMC5209692 DOI: 10.1038/srep39494] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/21/2016] [Indexed: 12/20/2022] Open
Abstract
In the field of cardiac drug efficacy and safety assessment, information on drug concentration in heart tissue is desirable. Because measuring drug concentrations in human cardiac tissue is challenging in healthy volunteers, mathematical models are used to cope with such limitations. With a goal of predicting drug concentration in cardiac tissue, we have developed a whole-body PBPK model consisting of seventeen perfusion-limited compartments. The proposed PBPK heart model consisted of four compartments: the epicardium, midmyocardium, endocardium, and pericardial fluid, and accounted for cardiac metabolism using CYP450. The model was written in R. The plasma:tissues partition coefficients (Kp) were calculated in Simcyp Simulator. The model was fitted to the concentrations of amitriptyline in plasma and the heart. The estimated parameters were as follows: 0.80 for the absorption rate [h-1], 52.6 for Kprest, 0.01 for the blood flow through the pericardial fluid [L/h], and 0.78 for the P-parameter describing the diffusion between the pericardial fluid and epicardium [L/h]. The total cardiac clearance of amitriptyline was calculated as 0.316 L/h. Although the model needs further improvement, the results support its feasibility, and it is a first attempt to provide an active drug concentration in various locations within heart tissue using a PBPK approach.
Collapse
Affiliation(s)
- Zofia Tylutki
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688 Cracow, Poland
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9 Str., 30-688 Cracow, Poland
- Simcyp (a Certara Company) Limited, Blades Enterprise Centre, John Street, Sheffield S2 4SU, UK
| |
Collapse
|
184
|
Luzon E, Blake K, Cole S, Nordmark A, Versantvoort C, Berglund EG. Physiologically based pharmacokinetic modeling in regulatory decision-making at the European Medicines Agency. Clin Pharmacol Ther 2016; 102:98-105. [PMID: 27770430 DOI: 10.1002/cpt.539] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/11/2016] [Accepted: 10/14/2016] [Indexed: 12/19/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is a valuable tool in drug development and regulatory assessment, as it offers the opportunity to simulate the pharmacokinetics of a compound, with a mechanistic understanding, in a variety of populations and situations. This work reviews the use and impact of such modeling in selected regulatory procedures submitted to the European Medicines Agency (EMA) before the end of 2015, together with its subsequent reflection in public documents relating to the assessment of these procedures. It is apparent that the reference to PBPK modeling in regulatory public documents underrepresents its use. A positive trend over time of the number of PBPK models submitted is shown, and in a number of cases the results of these may impact the decision-making process or lead to recommendations in the product labeling. These results confirm the need for regulatory guidance in this field, which is currently under development by the EMA.
Collapse
Affiliation(s)
- E Luzon
- European Medicines Agency (EMA), London, UK
| | - K Blake
- European Medicines Agency (EMA), London, UK
| | - S Cole
- Medicines and Healthcare products Regulatory Agency (MHRA), London, UK.,Pharmacokinetics Working Party (PKWP), EMA, London, UK.,Modelling and Simulation Working Group (MSWG), EMA, London, UK
| | - A Nordmark
- Modelling and Simulation Working Group (MSWG), EMA, London, UK.,Medical Products Agency (MPA), Uppsala, Sweden
| | - C Versantvoort
- Pharmacokinetics Working Party (PKWP), EMA, London, UK.,Medicines Evaluation Board (MEB), Utrecht, Netherlands
| | - E Gil Berglund
- Pharmacokinetics Working Party (PKWP), EMA, London, UK.,Medical Products Agency (MPA), Uppsala, Sweden
| |
Collapse
|
185
|
Kuepfer L, Niederalt C, Wendl T, Schlender JF, Willmann S, Lippert J, Block M, Eissing T, Teutonico D. Applied Concepts in PBPK Modeling: How to Build a PBPK/PD Model. CPT Pharmacometrics Syst Pharmacol 2016; 5:516-531. [PMID: 27653238 PMCID: PMC5080648 DOI: 10.1002/psp4.12134] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this tutorial is to introduce the fundamental concepts of physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling with a special focus on their practical implementation in a typical PBPK model building workflow. To illustrate basic steps in PBPK model building, a PBPK model for ciprofloxacin will be constructed and coupled to a pharmacodynamic model to simulate the antibacterial activity of ciprofloxacin treatment.
Collapse
Affiliation(s)
- L Kuepfer
- Bayer Technology Services, Leverkusen, Germany
| | - C Niederalt
- Bayer Technology Services, Leverkusen, Germany
| | - T Wendl
- Bayer Technology Services, Leverkusen, Germany
| | | | | | - J Lippert
- Bayer HealthCare, Wuppertal, Germany
| | - M Block
- Bayer Technology Services, Leverkusen, Germany
| | - T Eissing
- Bayer Technology Services, Leverkusen, Germany
| | - D Teutonico
- Bayer Technology Services, Leverkusen, Germany.
| |
Collapse
|
186
|
Hartmanshenn C, Scherholz M, Androulakis IP. Physiologically-based pharmacokinetic models: approaches for enabling personalized medicine. J Pharmacokinet Pharmacodyn 2016; 43:481-504. [PMID: 27647273 DOI: 10.1007/s10928-016-9492-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/06/2016] [Indexed: 12/17/2022]
Abstract
Personalized medicine strives to deliver the 'right drug at the right dose' by considering inter-person variability, one of the causes for therapeutic failure in specialized populations of patients. Physiologically-based pharmacokinetic (PBPK) modeling is a key tool in the advancement of personalized medicine to evaluate complex clinical scenarios, making use of physiological information as well as physicochemical data to simulate various physiological states to predict the distribution of pharmacokinetic responses. The increased dependency on PBPK models to address regulatory questions is aligned with the ability of PBPK models to minimize ethical and technical difficulties associated with pharmacokinetic and toxicology experiments for special patient populations. Subpopulation modeling can be achieved through an iterative and integrative approach using an adopt, adapt, develop, assess, amend, and deliver methodology. PBPK modeling has two valuable applications in personalized medicine: (1) determining the importance of certain subpopulations within a distribution of pharmacokinetic responses for a given drug formulation and (2) establishing the formulation design space needed to attain a targeted drug plasma concentration profile. This review article focuses on model development for physiological differences associated with sex (male vs. female), age (pediatric vs. young adults vs. elderly), disease state (healthy vs. unhealthy), and temporal variation (influence of biological rhythms), connecting them to drug product formulation development within the quality by design framework. Although PBPK modeling has come a long way, there is still a lengthy road before it can be fully accepted by pharmacologists, clinicians, and the broader industry.
Collapse
Affiliation(s)
- Clara Hartmanshenn
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ, 08854, USA
| | - Megerle Scherholz
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ, 08854, USA
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ, 08854, USA. .,Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
187
|
Wiśniowska B, Polak S. Virtual Clinical Trial Toward Polytherapy Safety Assessment: Combination of Physiologically Based Pharmacokinetic/Pharmacodynamic-Based Modeling and Simulation Approach With Drug-Drug Interactions Involving Terfenadine as an Example. J Pharm Sci 2016; 105:3415-3424. [PMID: 27640752 DOI: 10.1016/j.xphs.2016.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 10/21/2022]
Abstract
A Quantitative Systems Pharmacology approach was utilized to predict the cardiac consequences of drug-drug interaction (DDI) at the population level. The Simcyp in vitro-in vivo correlation and physiologically based pharmacokinetic platform was used to predict the pharmacokinetic profile of terfenadine following co-administration of the drug. Electrophysiological effects were simulated using the Cardiac Safety Simulator. The modulation of ion channel activity was dependent on the inhibitory potential of drugs on the main cardiac ion channels and a simulated free heart tissue concentration. ten Tusscher's human ventricular cardiomyocyte model was used to simulate the pseudo-ECG traces and further predict the pharmacodynamic consequences of DDI. Consistent with clinical observations, predicted plasma concentration profiles of terfenadine show considerable intra-subject variability with recorded Cmax values below 5 ng/mL for most virtual subjects. The pharmacokinetic and pharmacodynamic effects of inhibitors were predicted with reasonable accuracy. In all cases, a combination of the physiologically based pharmacokinetic and physiology-based pharmacodynamic models was able to differentiate between the terfenadine alone and terfenadine + inhibitor scenario. The range of QT prolongation was comparable in the clinical and virtual studies. The results indicate that mechanistic in vitro-in vivo correlation can be applied to predict the clinical effects of DDI even without comprehensive knowledge on all mechanisms contributing to the interaction.
Collapse
Affiliation(s)
- Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, Kraków 30-688, Poland.
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, Kraków 30-688, Poland; Simcyp (part of Certara), Blades Enterprise Centre, John Street, Sheffield S2 4SU, UK
| |
Collapse
|
188
|
Zhuang X, Lu C. PBPK modeling and simulation in drug research and development. Acta Pharm Sin B 2016; 6:430-440. [PMID: 27909650 PMCID: PMC5125732 DOI: 10.1016/j.apsb.2016.04.004] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 01/13/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling and simulation can be used to predict the pharmacokinetic behavior of drugs in humans using preclinical data. It can also explore the effects of various physiologic parameters such as age, ethnicity, or disease status on human pharmacokinetics, as well as guide dose and dose regiment selection and aid drug-drug interaction risk assessment. PBPK modeling has developed rapidly in the last decade within both the field of academia and the pharmaceutical industry, and has become an integral tool in drug discovery and development. In this mini-review, the concept and methodology of PBPK modeling are briefly introduced. Several case studies were discussed on how PBPK modeling and simulation can be utilized through various stages of drug discovery and development. These case studies are from our own work and the literature for better understanding of the absorption, distribution, metabolism and excretion (ADME) of a drug candidate, and the applications to increase efficiency, reduce the need for animal studies, and perhaps to replace clinical trials. The regulatory acceptance and industrial practices around PBPK modeling and simulation is also discussed.
Collapse
Affiliation(s)
- Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing
Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chuang Lu
- Department of DMPK, Biogen, Inc., Cambridge, MA 02142, USA
- Corresponding author. Tel.: +1 6176793365.
| |
Collapse
|
189
|
Lavé T, Caruso A, Parrott N, Walz A. Translational PK/PD modeling to increase probability of success in drug discovery and early development. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:27-34. [PMID: 27978984 DOI: 10.1016/j.ddtec.2016.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 06/06/2023]
Abstract
In this review we present ways in which translational PK/PD modeling can address opportunities to enhance probability of success in drug discovery and early development. This is achieved by impacting efficacy and safety-driven attrition rates, through increased focus on the quantitative understanding and modeling of translational PK/PD. Application of the proposed principles early in the discovery and development phases is anticipated to bolster confidence of successfully evaluating proof of mechanism in humans and ultimately improve Phase II success. The present review is centered on the application of predictive modeling and simulation approaches during drug discovery and early development, and more specifically of mechanism-based PK/PD modeling. Case studies are presented, focused on the relevance of M&S contributions to real-world questions and the impact on decision making.
Collapse
Affiliation(s)
- Thierry Lavé
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland.
| | - Antonello Caruso
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Antje Walz
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| |
Collapse
|
190
|
Cristofoletti R, Charoo NA, Dressman JB. Exploratory Investigation of the Limiting Steps of Oral Absorption of Fluconazole and Ketoconazole in Children Using an In Silico Pediatric Absorption Model. J Pharm Sci 2016; 105:2794-2803. [DOI: 10.1016/j.xphs.2016.01.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 11/28/2022]
|
191
|
Grillo JA, Huang SM. Perspectives in regulatory science: translational and clinical pharmacology. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:67-73. [PMID: 27978990 DOI: 10.1016/j.ddtec.2016.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 06/06/2023]
Abstract
This paper focuses on the role of clinical and translational pharmacology in the drug development and the regulatory process. Contemporary regulatory issues faced by FDA's Office of Clinical Pharmacology (OCP) in fulfilling its mission to advance the science of drug response and translate patient diversity into optimal drug therapy are discussed. Specifically current focus of the following key aspects of the drug development and regulatory science processes are discussed: the OCP vision and mission, two key OCP initiatives (i.e. guidance modernization, labeling and health communications), and translational and clinical pharmacology related regulatory science issues in (i.e. uncertainty, breakthrough therapies, individualization).
Collapse
Affiliation(s)
- Joseph A Grillo
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Shiew Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States.
| |
Collapse
|
192
|
Zhou W, Johnson TN, Xu H, Cheung S, Bui KH, Li J, Al-Huniti N, Zhou D. Predictive Performance of Physiologically Based Pharmacokinetic and Population Pharmacokinetic Modeling of Renally Cleared Drugs in Children. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:475-83. [PMID: 27566992 PMCID: PMC5036422 DOI: 10.1002/psp4.12101] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/07/2016] [Accepted: 06/29/2016] [Indexed: 11/17/2022]
Abstract
Predictive performance of physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PopPK) models of drugs predominantly eliminated through kidney in the pediatric population was evaluated. After optimization using adult clinical data, the verified PBPK models can predict 33 of 34 drug clearance within twofold of the observed values in children 1 month and older. More specifically, 10 of 11 of predicted clearance values were within 1.5‐fold of those observed in children between 1 month and 2 years old. The PopPK approach also predicted 19 of 21 drug clearance within twofold of the observed values in children. In summary, our analysis demonstrated both PBPK and PopPK adult models, after verification with additional adult pharmacokinetic (PK) studies and incorporation of known ontogeny of renal filtration, could be applied for dosing regimen recommendation in children 1 month and older for renally eliminated drugs in a first‐in‐pediatric study.
Collapse
Affiliation(s)
- W Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - T N Johnson
- Simcyp (A Certara Company), Sheffield, United Kingdom
| | - H Xu
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Sya Cheung
- Quantitative Clinical Pharmacology, AstraZeneca, Cambridge, United Kingdom
| | - K H Bui
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - J Li
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - N Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - D Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA.
| |
Collapse
|
193
|
Scotcher D, Jones C, Posada M, Galetin A, Rostami-Hodjegan A. Key to Opening Kidney for In Vitro-In Vivo Extrapolation Entrance in Health and Disease: Part II: Mechanistic Models and In Vitro-In Vivo Extrapolation. AAPS JOURNAL 2016; 18:1082-1094. [PMID: 27506526 DOI: 10.1208/s12248-016-9959-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/11/2016] [Indexed: 12/11/2022]
Abstract
It is envisaged that application of mechanistic models will improve prediction of changes in renal disposition due to drug-drug interactions, genetic polymorphism in enzymes and transporters and/or renal impairment. However, developing and validating mechanistic kidney models is challenging due to the number of processes that may occur (filtration, secretion, reabsorption and metabolism) in this complex organ. Prediction of human renal drug disposition from preclinical species may be hampered by species differences in the expression and activity of drug metabolising enzymes and transporters. A proposed solution is bottom-up prediction of pharmacokinetic parameters based on in vitro-in vivo extrapolation (IVIVE), mediated by recent advances in in vitro experimental techniques and application of relevant scaling factors. This review is a follow-up to the Part I of the report from the 2015 AAPS Annual Meeting and Exhibition (Orlando, FL; 25th-29th October 2015) which focuses on IVIVE and mechanistic prediction of renal drug disposition. It describes the various mechanistic kidney models that may be used to investigate renal drug disposition. Particular attention is given to efforts that have attempted to incorporate elements of IVIVE. In addition, the use of mechanistic models in prediction of renal drug-drug interactions and potential for application in determining suitable adjustment of dose in kidney disease are discussed. The need for suitable clinical pharmacokinetics data for the purposes of delineating mechanistic aspects of kidney models in various scenarios is highlighted.
Collapse
Affiliation(s)
- Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Christopher Jones
- DMPK, Oncology iMed, AstraZeneca R&D Alderley Park, Macclesfield, Cheshire, UK
| | - Maria Posada
- Drug Disposition, Lilly Research Laboratories, Indianapolis, Indiana, 46203, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK. .,Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, UK.
| |
Collapse
|
194
|
Lin W, Heimbach T, Jain JP, Awasthi R, Hamed K, Sunkara G, He H. A Physiologically Based Pharmacokinetic Model to Describe Artemether Pharmacokinetics in Adult and Pediatric Patients. J Pharm Sci 2016; 105:3205-3213. [PMID: 27506269 DOI: 10.1016/j.xphs.2016.06.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 11/25/2022]
Abstract
Artemether is co-administered with lumefantrine as part of a fixed-dose combination therapy for malaria in both adult and pediatric patients. However, artemether exposure is higher in younger infants (1-3 months) with a lower body weight (<5 kg) as compared to older infants (3-6 months) with a higher body weight (≥5 to <10 kg), children, and adults. In contrast, lumefantrine exposure is similar in all age groups. This article describes the clinically observed artemether exposure data in pediatric populations across various age groups (1 month to 12 years) and body weights (<5 or ≥5 kg) using physiologically based pharmacokinetic (PBPK) mechanistic models. A PBPK model was developed using artemether physicochemical, biopharmaceutic, and metabolic properties together with known enzyme ontogeny and pediatric physiology. The model was verified using clinical data from adult patients after multiple doses of oral artemether, and was then applied to simulate the exposure in children and infants. The simulated PBPK concentration-time profiles captured observed clinical data. Consistent with the clinical data, the PBPK model simulations indicated a higher artemether exposure for younger infants with lower body weight. A PBPK model developed for artemether reliably described the clinical data from adult and pediatric patients.
Collapse
Affiliation(s)
- Wen Lin
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936
| | - Tycho Heimbach
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936.
| | - Jay Prakash Jain
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - Rakesh Awasthi
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936
| | - Kamal Hamed
- Global Medical Affairs, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey 07936
| | - Gangadhar Sunkara
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936
| | - Handan He
- Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936
| |
Collapse
|
195
|
Parrott NJ, Yu LJ, Takano R, Nakamura M, Morcos PN. Physiologically Based Absorption Modeling to Explore the Impact of Food and Gastric pH Changes on the Pharmacokinetics of Alectinib. AAPS JOURNAL 2016; 18:1464-1474. [PMID: 27450228 DOI: 10.1208/s12248-016-9957-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/09/2016] [Indexed: 01/06/2023]
Abstract
Alectinib, a lipophilic, basic, anaplastic lymphoma kinase (ALK) inhibitor with very low aqueous solubility, has received Food and Drug Administration-accelerated approval for the treatment of patients with ALK+ non-small-cell lung cancer. This paper describes the application of physiologically based absorption modeling during clinical development to predict and understand the impact of food and gastric pH changes on alectinib absorption. The GastroPlus™ software was used to develop an absorption model integrating in vitro and in silico data on drug substance properties. Oral pharmacokinetics was simulated by linking the absorption model to a disposition model fit to pharmacokinetic data obtained after an intravenous infusion. Simulations were compared to clinical data from a food effect study and a drug-drug interaction study with esomeprazole, a gastric acid-reducing agent. Prospective predictions of a positive food effect and negligible impact of gastric pH elevation were confirmed with clinical data, although the exact magnitude of the food effect could not be predicted with confidence. After optimization of the absorption model with clinical food effect data, a refined model was further applied to derive recommendations on the timing of dose administration with respect to a meal. The application of biopharmaceutical absorption modeling is an area with great potential to further streamline late stage drug development and with impact on regulatory questions.
Collapse
Affiliation(s)
- Neil J Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070, Basel, Switzerland.
| | - Li J Yu
- Roche Innovation Center, New York City, New York, USA
| | - Ryusuke Takano
- Pharmaceutical Technology Division, Chugai Pharmaceutical Co. Ltd., Tokyo, Japan
| | - Mikiko Nakamura
- Translational Clinical Research Division, Chugai Pharmaceutical Co. Ltd., Tokyo, Japan
| | | |
Collapse
|
196
|
Mehrotra N, Bhattaram A, Earp JC, Florian J, Krudys K, Lee JE, Lee JY, Liu J, Mulugeta Y, Yu J, Zhao P, Sinha V. Role of Quantitative Clinical Pharmacology in Pediatric Approval and Labeling. Drug Metab Dispos 2016; 44:924-33. [PMID: 27079249 DOI: 10.1124/dmd.116.069559] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
Dose selection is one of the key decisions made during drug development in pediatrics. There are regulatory initiatives that promote the use of model-based drug development in pediatrics. Pharmacometrics or quantitative clinical pharmacology enables development of models that can describe factors affecting pharmacokinetics and/or pharmacodynamics in pediatric patients. This manuscript describes some examples in which pharmacometric analysis was used to support approval and labeling in pediatrics. In particular, the role of pharmacokinetic (PK) comparison of pediatric PK to adults and utilization of dose/exposure-response analysis for dose selection are highlighted. Dose selection for esomeprazole in pediatrics was based on PK matching to adults, whereas for adalimumab, exposure-response, PK, efficacy, and safety data together were useful to recommend doses for pediatric Crohn's disease. For vigabatrin, demonstration of similar dose-response between pediatrics and adults allowed for selection of a pediatric dose. Based on model-based pharmacokinetic simulations and safety data from darunavir pediatric clinical studies with a twice-daily regimen, different once-daily dosing regimens for treatment-naïve human immunodeficiency virus 1-infected pediatric subjects 3 to <12 years of age were evaluated. The role of physiologically based pharmacokinetic modeling (PBPK) in predicting pediatric PK is rapidly evolving. However, regulatory review experiences and an understanding of the state of science indicate that there is a lack of established predictive performance of PBPK in pediatric PK prediction. Moving forward, pharmacometrics will continue to play a key role in pediatric drug development contributing toward decisions pertaining to dose selection, trial designs, and assessing disease similarity to adults to support extrapolation of efficacy.
Collapse
Affiliation(s)
- Nitin Mehrotra
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Atul Bhattaram
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Justin C Earp
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jeffry Florian
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Kevin Krudys
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jee Eun Lee
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Joo Yeon Lee
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jiang Liu
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Yeruk Mulugeta
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jingyu Yu
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Ping Zhao
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Vikram Sinha
- Division of Pharmacometrics, Office of Clinical Pharmacology (N.M., A.B., J.C.E., J.F., K.K., J.E.L., J.L., Y.M., J.Y., P.Z., V.S.), and Division of Biometrics VII, Office of Biostatistics (J.Y.L.), Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
197
|
Rioux N, Waters NJ. Physiologically Based Pharmacokinetic Modeling in Pediatric Oncology Drug Development. Drug Metab Dispos 2016; 44:934-43. [PMID: 26936973 DOI: 10.1124/dmd.115.068031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/01/2016] [Indexed: 02/13/2025] Open
Abstract
Childhood cancer represents more than 100 rare and ultra-rare diseases, with an estimated 12,400 new cases diagnosed each year in the United States. As such, this much smaller patient population has led to pediatric oncology drug development lagging behind that for adult cancers. Developing drugs for pediatric malignancies also brings with it a number of unique trial design considerations, including flexible enrollment approaches, age-appropriate formulation, acceptable sampling schedules, and balancing the need for age-stratified dosing regimens, given the smaller patient populations. The regulatory landscape for pediatric pharmacotherapy has evolved with U.S. Food and Drug Administration (FDA) legislation such as the 2012 FDA Safety and Innovation Act. In parallel, regulatory authorities have recommended the application of physiologically based pharmacokinetic (PBPK) modeling, for example, in the recently issued FDA Strategic Plan for Accelerating the Development of Therapies for Pediatric Rare Diseases. PBPK modeling provides a quantitative and systems-based framework that allows the effects of intrinsic and extrinsic factors on drug exposure to be modeled in a mechanistic fashion. The application of PBPK modeling in drug development for pediatric cancers is relatively nascent, with several retrospective analyses of cytotoxic therapies, and latterly for targeted agents such as obatoclax and imatinib. More recently, we have employed PBPK modeling in a prospective manner to inform the first pediatric trials of pinometostat and tazemetostat in genetically defined populations (mixed lineage leukemia-rearranged and integrase interactor-1-deficient sarcomas, respectively). In this review, we evaluate the application of PBPK modeling in pediatric cancer drug development and discuss the important challenges that lie ahead in this field.
Collapse
|
198
|
Sjögren E, Thörn H, Tannergren C. In Silico Modeling of Gastrointestinal Drug Absorption: Predictive Performance of Three Physiologically Based Absorption Models. Mol Pharm 2016; 13:1763-78. [PMID: 26926043 DOI: 10.1021/acs.molpharmaceut.5b00861] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Gastrointestinal (GI) drug absorption is a complex process determined by formulation, physicochemical and biopharmaceutical factors, and GI physiology. Physiologically based in silico absorption models have emerged as a widely used and promising supplement to traditional in vitro assays and preclinical in vivo studies. However, there remains a lack of comparative studies between different models. The aim of this study was to explore the strengths and limitations of the in silico absorption models Simcyp 13.1, GastroPlus 8.0, and GI-Sim 4.1, with respect to their performance in predicting human intestinal drug absorption. This was achieved by adopting an a priori modeling approach and using well-defined input data for 12 drugs associated with incomplete GI absorption and related challenges in predicting the extent of absorption. This approach better mimics the real situation during formulation development where predictive in silico models would be beneficial. Plasma concentration-time profiles for 44 oral drug administrations were calculated by convolution of model-predicted absorption-time profiles and reported pharmacokinetic parameters. Model performance was evaluated by comparing the predicted plasma concentration-time profiles, Cmax, tmax, and exposure (AUC) with observations from clinical studies. The overall prediction accuracies for AUC, given as the absolute average fold error (AAFE) values, were 2.2, 1.6, and 1.3 for Simcyp, GastroPlus, and GI-Sim, respectively. The corresponding AAFE values for Cmax were 2.2, 1.6, and 1.3, respectively, and those for tmax were 1.7, 1.5, and 1.4, respectively. Simcyp was associated with underprediction of AUC and Cmax; the accuracy decreased with decreasing predicted fabs. A tendency for underprediction was also observed for GastroPlus, but there was no correlation with predicted fabs. There were no obvious trends for over- or underprediction for GI-Sim. The models performed similarly in capturing dependencies on dose and particle size. In conclusion, it was shown that all three software packages are useful to guide formulation development. However, as a consequence of the high fraction of inaccurate predictions (prediction error >2-fold) and the clear trend toward decreased accuracy with decreased predicted fabs observed with Simcyp, the results indicate that GI-Sim and GastroPlus perform better than Simcyp in predicting the intestinal absorption of the incompletely absorbed drugs when a higher degree of accuracy is needed. In addition, this study suggests that modeling and simulation research groups should perform systematic model evaluations using their own input data to maximize confidence in model performance and output.
Collapse
Affiliation(s)
- Erik Sjögren
- Department of Pharmacy, Uppsala University , Box 580, S-751 23 Uppsala, Sweden
| | - Helena Thörn
- Pharmaceutical Technology and Development, AstraZeneca R&D Gothenburg , SE-43183 Mölndal, Sweden
| | - Christer Tannergren
- Pharmaceutical Technology and Development, AstraZeneca R&D Gothenburg , SE-43183 Mölndal, Sweden
| |
Collapse
|
199
|
Yu G, Li GF, Markowitz JS. Atomoxetine: A Review of Its Pharmacokinetics and Pharmacogenomics Relative to Drug Disposition. J Child Adolesc Psychopharmacol 2016; 26:314-26. [PMID: 26859445 PMCID: PMC4876529 DOI: 10.1089/cap.2015.0137] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Atomoxetine is a selective norepinephrine (NE) reuptake inhibitor approved for the treatment of attention-deficit/hyperactivity disorder (ADHD) in children (≥6 years of age), adolescents, and adults. Its metabolism and disposition are fairly complex, and primarily governed by cytochrome P450 (CYP) 2D6 (CYP2D6), whose protein expression varies substantially from person to person, and by race and ethnicity because of genetic polymorphism. These differences can be substantial, resulting in 8-10-fold differences in atomoxetine exposure between CYP2D6 poor metabolizers and extensive metabolizers. In this review, we have attempted to revisit and analyze all published clinical pharmacokinetic data on atomoxetine inclusive of public access documents from the new drug application submitted to the United States Food and Drug Administration (FDA). The present review focuses on atomoxetine metabolism, disposition, and genetic polymorphisms of CYP2D6 as they specifically relate to atomoxetine, and provides an in-depth discussion of the fundamental pharmacokinetics of the drug including its absorption, distribution, metabolism, and excretion in pediatric and adult populations. Further, a summary of relationships between genetic variants of CYP2D6 and to some degree, CYP2C19, are provided with respect to atomoxetine plasma concentrations, central nervous system (CNS) pharmacokinetics, and associated clinical implications for pharmacotherapy. Lastly, dosage adjustments based on pharmacokinetic principles are discussed.
Collapse
Affiliation(s)
- Guo Yu
- Laboratory of Pharmacogenomics and Pharmacokinetic Research, Subei People's Hospital, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guo-Fu Li
- Center for Drug Clinical Research, Shanghai University of Chinese Medicine, Shanghai, China
| | - John S. Markowitz
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
- Center for Pharmacogenomics, University of Florida, Gainesville, Florida
| |
Collapse
|
200
|
Jamei M. Recent Advances in Development and Application of Physiologically-Based Pharmacokinetic (PBPK) Models: a Transition from Academic Curiosity to Regulatory Acceptance. ACTA ACUST UNITED AC 2016; 2:161-169. [PMID: 27226953 PMCID: PMC4856711 DOI: 10.1007/s40495-016-0059-9] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There is a renewed surge of interest in applications of physiologically-based pharmacokinetic (PBPK) models by the pharmaceutical industry and regulatory agencies. Developing PBPK models within a systems pharmacology context allows separation of the parameters pertaining to the animal or human body (the system) from that of the drug and the study design which is essential to develop generic drug-independent models used to extrapolate PK/PD properties in various healthy and patient populations. This has expanded the classical paradigm to a ‘predict-learn-confirm-apply’ concept. Recently, a number of drug labels are informed by simulation results generated using PBPK models. These cases show that either the simulations are used in lieu of conducting clinical studies or have informed the drug label that otherwise would have been silent in some specific situations. It will not be surprising to see applications of these models in implementing precision dosing at the point of care in the near future.
Collapse
Affiliation(s)
- Masoud Jamei
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, John Street, Sheffield, S2 4SU UK
| |
Collapse
|