1
|
Falkenhagen U, Cavallari LH, Duarte JD, Kloft C, Schmidt S, Huisinga W. Leveraging QSP Models for MIPD: A Case Study for Warfarin/INR. Clin Pharmacol Ther 2024; 116:795-806. [PMID: 38655898 DOI: 10.1002/cpt.3274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
Warfarin dosing remains challenging due to substantial inter-individual variability, which can lead to unsafe or ineffective therapy with standard dosing. Model-informed precision dosing (MIPD) can help individualize warfarin dosing, requiring the selection of a suitable model. For models developed from clinical data, the dependence on the study design and population raises questions about generalizability. Quantitative system pharmacology (QSP) models promise better extrapolation abilities; however, their complexity and lack of validation on clinical data raise questions about applicability in MIPD. We have previously derived a mechanistic warfarin/international normalized ratio (INR) model from a blood coagulation QSP model. In this article, we evaluated the predictive performance of the warfarin/INR model in the context of MIPD using an external dataset with INR data from patients starting warfarin treatment. We assessed the accuracy and precision of model predictions, benchmarked against an empirically based reference model. Additionally, we evaluated covariate contributions and assessed the predictive performance separately in the more challenging outpatient data. The warfarin/INR model performed comparably to the reference model across various measures despite not being calibrated with warfarin initiation data. Including CYP2C9 and/or VKORC1 genotypes as covariates improved the prediction quality of the warfarin/INR model, even after assimilating 4 days of INR data. The outpatient INR exhibited higher unexplained variability, and predictions slightly exceeded observed values, suggesting that model adjustments might be necessary when transitioning from an inpatient to an outpatient setting. Overall, this research underscores the potential of QSP-derived models for MIPD, offering a complementary approach to empirical model development.
Collapse
Affiliation(s)
- Undine Falkenhagen
- PharMetrX Graduate Research Training Program, Berlin/Potsdam, Germany
- Institute of Mathematics, Mathematical Modelling and Systems Biology, University of Potsdam, Potsdam, Germany
| | - Larisa H Cavallari
- College of Pharmacy, Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida, USA
| | - Julio D Duarte
- College of Pharmacy, Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida, USA
| | - Charlotte Kloft
- Institute of Pharmacy, Department of Clinical Pharmacy and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stephan Schmidt
- College of Pharmacy, Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida, USA
| | - Wilhelm Huisinga
- Institute of Mathematics, Mathematical Modelling and Systems Biology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
2
|
Ding X, Xue L, Wang M, Zhu S, Zhu K, Jiang S, Wu J, Miao L. Dynamics and implications of anti-drug antibodies against adalimumab using ultra-sensitive and highly drug-tolerant assays. Front Immunol 2024; 15:1429544. [PMID: 39238635 PMCID: PMC11374634 DOI: 10.3389/fimmu.2024.1429544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Background Adalimumab induces the production of anti-drug antibodies (ADA) that may lead to reduced drug concentration and loss-of-response, posing significant clinical challenges. However, traditional immunoassays have limitations in terms of sensitivity and drug-tolerance, hindering the insights of ADA response. Methods Herein, we developed an integrated immunoassay platform combining the electrochemiluminescence immunoassay with immunomagnetic separation strategy. A longitudinal cohort study involving 49 patients with ankylosing spondylitis was carried out to analyze the dynamic profiles of ADA and to investigate the impact of ADA on adalimumab pharmacokinetics using a population pharmacokinetic model. Additionally, cross-sectional data from 12 patients were collected to validate the correlation between ADA levels and disease relapse. Results The ADA assay demonstrated high sensitivity (0.4 ng/mL) and drug-tolerance (100 μg/mL), while the neutralizing antibodies (NAB) assay showed a sensitivity of 100 ng/mL and drug-tolerance of 20 μg/mL. Analysis of the longitudinal cohort revealed that a majority of patients (44/49, 90%) developed persistent ADA within the first 24 weeks of treatment. ADA levels tended to plateau over time after an initial increase during the early immune response phase. Further, nearly all of the tested patients (26/27, 96%) were classified as NAB positive, with a strong correlation between ADA levels and neutralization capacity (R2 = 0.83, P < 0.001). Population pharmacokinetic modeling revealed a significant positive association between model-estimated individual clearance and observed ADA levels. Higher ADA levels were associated with adalimumab clearance and disease relapse in a cross-sectional cohort, suggesting a promising ADA threshold of 10 for potential clinical application. Moreover, the IgG class was the primary contributor to ADA against adalimumab and the apparent affinity exhibited an increasing trend over time, indicating a T-cell dependent mechanism for ADA elicitation by adalimumab. Conclusion In summary, this integrated immunoassay platform shows promise for in-depth analysis of ADA against biologics, offering fresh insights into immunogenicity and its clinical implications.
Collapse
Affiliation(s)
- Xiaoliang Ding
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Ling Xue
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Mingjun Wang
- Department of Rheumatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengxiong Zhu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Kouzhu Zhu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Sheng Jiang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jian Wu
- Department of Rheumatology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Xue L, He S, Singla RK, Qin Q, Ding Y, Liu L, Ding X, Bediaga-Bañeres H, Arrasate S, Durado-Sanchez A, Zhang Y, Shen Z, Shen B, Miao L, González-Díaz H. Machine learning guided prediction of warfarin blood levels for personalized medicine based on clinical longitudinal data from cardiac surgery patients: a prospective observational study. Int J Surg 2024; 110:01279778-990000000-01621. [PMID: 38833337 PMCID: PMC11487003 DOI: 10.1097/js9.0000000000001734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/19/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Warfarin is a common oral anticoagulant, and its effects vary widely among individuals. Numerous dose-prediction algorithms have been reported based on cross-sectional data generated via multiple linear regression or machine learning. This study aimed to construct an information fusion perturbation theory and machine learning prediction model of warfarin blood levels based on clinical longitudinal data from cardiac surgery patients. METHODS AND MATERIAL The data of 246 patients were obtained from electronic medical records. Continuous variables were processed by calculating the distance of the raw data with the moving average (MA ∆vki(sj)), and categorical variables in different attribute groups were processed using Euclidean distance (ED ǁ∆vk(sj)ǁ). Regression and classification analyses were performed on the raw data, MA ∆vki(sj), and ED ǁ∆vk(sj)ǁ. Different machine-learning algorithms were chosen for the STATISTICA and WEKA software. RESULTS The random forest (RF) algorithm was the best for predicting continuous outputs using the raw data. The correlation coefficients of the RF algorithm were 0.978 and 0.595 for the training and validation sets, respectively, and the mean absolute errors were 0.135 and 0.362 for the training and validation sets, respectively. The proportion of ideal predictions of the RF algorithm was 59.0%. General discriminant analysis (GDA) was the best algorithm for predicting the categorical outputs using the MA ∆vki(sj) data. The GDA algorithm's total true positive rate (TPR) was 95.4% and 95.6% for the training and validation sets, respectively, with MA ∆vki(sj) data. CONCLUSIONS An information fusion perturbation theory and machine learning model for predicting warfarin blood levels was established. A model based on the RF algorithm could be used to predict the target international normalized ratio (INR), and a model based on the GDA algorithm could be used to predict the probability of being within the target INR range under different clinical scenarios.
Collapse
Affiliation(s)
- Ling Xue
- Department of Pharmacy, the First Affiliated Hospital of Soochow University
- Department of Pharmacology, Faculty of Medicine, University of The Basque Country (UPV/EHU), Bilbao, Basque Country
| | - Shan He
- Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
- IKERDATA S.L., ZITEK, University of The Basque Country (UPV/EHU), Bilbao, Basque Country
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Qiong Qin
- Department of Pharmacy, the First Affiliated Hospital of Soochow University
| | - Yinglong Ding
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University
- Institute for Cardiovascular Science, Soochow University
| | - Linsheng Liu
- Department of Pharmacy, the First Affiliated Hospital of Soochow University
| | - Xiaoliang Ding
- Department of Pharmacy, the First Affiliated Hospital of Soochow University
| | - Harbil Bediaga-Bañeres
- IKERDATA S.L., ZITEK, University of The Basque Country (UPV/EHU), Bilbao, Basque Country
- Department of Painting, Faculty of Fine Arts, University of the Basque Country UPV/EHU, 48940, Leioa, Biscay
| | - Sonia Arrasate
- Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - Aliuska Durado-Sanchez
- IKERDATA S.L., ZITEK, University of The Basque Country (UPV/EHU), Bilbao, Basque Country
- Department of Public Law, Faculty of Law, University of The Basque Country (UPV/EHU), Leioa, Biscay, Basque, Country
| | - Yuzhen Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University
| | - Zhenya Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University
- Institute for Cardiovascular Science, Soochow University
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Liyan Miao
- Department of Pharmacy, the First Affiliated Hospital of Soochow University
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
- BIOFISIKA: Basque Center for Biophysics CSIC, University of The Basque Country (UPV/EHU), Bilbao, Basque Country
- IKERBASQUE, Basque Foundation for Science, Bilbao, Basque Country, Spain
| |
Collapse
|
4
|
Xue L, Singla RK, He S, Arrasate S, González-Díaz H, Miao L, Shen B. Warfarin-A natural anticoagulant: A review of research trends for precision medication. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155479. [PMID: 38493714 DOI: 10.1016/j.phymed.2024.155479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Warfarin is a widely prescribed anticoagulant in the clinic. It has a more considerable individual variability, and many factors affect its variability. Mathematical models can quantify the quantitative impact of these factors on individual variability. PURPOSE The aim is to comprehensively analyze the advanced warfarin dosing algorithm based on pharmacometrics and machine learning models of personalized warfarin dosage. METHODS A bibliometric analysis of the literature retrieved from PubMed and Scopus was performed using VOSviewer. The relevant literature that reported the precise dosage of warfarin calculation was retrieved from the database. The multiple linear regression (MLR) algorithm was excluded because a recent systematic review that mainly reviewed this algorithm has been reported. The following terms of quantitative systems pharmacology, mechanistic model, physiologically based pharmacokinetic model, artificial intelligence, machine learning, pharmacokinetic, pharmacodynamic, pharmacokinetics, pharmacodynamics, and warfarin were added as MeSH Terms or appearing in Title/Abstract into query box of PubMed, then humans and English as filter were added to retrieve the literature. RESULTS Bibliometric analysis revealed important co-occuring MeShH and index keywords. Further, the United States, China, and the United Kingdom were among the top countries contributing in this domain. Some studies have established personalized warfarin dosage models using pharmacometrics and machine learning-based algorithms. There were 54 related studies, including 14 pharmacometric models, 31 artificial intelligence models, and 9 model evaluations. Each model has its advantages and disadvantages. The pharmacometric model contains biological or pharmacological mechanisms in structure. The process of pharmacometric model development is very time- and labor-intensive. Machine learning is a purely data-driven approach; its parameters are more mathematical and have less biological interpretation. However, it is faster, more efficient, and less time-consuming. Most published models of machine learning algorithms were established based on cross-sectional data sourced from the database. CONCLUSION Future research on personalized warfarin medication should focus on combining the advantages of machine learning and pharmacometrics algorithms to establish a more robust warfarin dosage algorithm. Randomized controlled trials should be performed to evaluate the established algorithm of warfarin dosage. Moreover, a more user-friendly and accessible warfarin precision medicine platform should be developed.
Collapse
Affiliation(s)
- Ling Xue
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pharmacology, Faculty of Medicine, University of The Basque Country (UPV/EHU), Bilbao, Basque Country, Spain
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Shan He
- IKERDATA S.l., ZITEK, University of The Basque Country (UPVEHU), Rectorate Building, 48940, Bilbao, Basque Country, Spain; Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), P.O. Box 644, 48080, Bilbao, Basque Country, Spain
| | - Sonia Arrasate
- Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), P.O. Box 644, 48080, Bilbao, Basque Country, Spain
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, Faculty of Science and Technology, University of The Basque Country (UPV/EHU), P.O. Box 644, 48080, Bilbao, Basque Country, Spain; BIOFISIKA: Basque Center for Biophysics CSIC, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, Leioa, Bizkaia 48940, Basque Country, Spain; IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Basque Country, Spain
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China; College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Xue L, Ma G, Holford N, Qin Q, Ding Y, Hannam JA, Ding X, Fan H, Ji Z, Yang B, Shen H, Shen Z, Miao L. A Randomized Trial Comparing Standard of Care to Bayesian Warfarin Dose Individualization. Clin Pharmacol Ther 2024; 115:1316-1325. [PMID: 38439157 DOI: 10.1002/cpt.3207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/24/2024] [Indexed: 03/06/2024]
Abstract
The quality of warfarin treatment may be improved if management is guided by the use of models based upon pharmacokinetic-pharmacodynamic theory. A prospective, two-armed, single-blind, randomized controlled trial compared management aided by a web-based dose calculator (NextDose) with standard clinical care. Participants were 240 adults receiving warfarin therapy following cardiac surgery, followed up until the first outpatient appointment at least 3 months after warfarin initiation. We compared the percentage of time spent in the international normalized ratio acceptable range (%TIR) during the first 28 days following warfarin initiation, and %TIR and count of bleeding events over the entire follow-up period. Two hundred thirty-four participants were followed up to day 28 (NextDose: 116 and standard of care: 118), and 228 participants (114 per arm) were followed up to the final study visit. Median %TIR tended to be higher for participants receiving NextDose guided warfarin management during the first 28 days (63 vs. 56%, P = 0.13) and over the entire follow-up period (74 vs. 71%, P = 0.04). The hazard of clinically relevant minor bleeding events was lower for participants in the NextDose arm (hazard ratio: 0.21, P = 0.041). In NextDose, there were 89.3% of proposed doses accepted by prescribers. NextDose guided dose management in cardiac surgery patients requiring warfarin was associated with an increase in %TIR across the full follow-up period and fewer hemorrhagic events. A theory-based, pharmacologically guided approach facilitates higher quality warfarin anticoagulation. An important practical benefit is a reduced requirement for clinical experience of warfarin management.
Collapse
Affiliation(s)
- Ling Xue
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Pharmacology, Faculty of Medicine, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Guangda Ma
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Nick Holford
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Qiong Qin
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinglong Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Jacqueline A Hannam
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Xiaoliang Ding
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongyou Fan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenchun Ji
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Biwen Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Han Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| |
Collapse
|
6
|
Sanghavi K, Ribbing J, Rogers JA, Ahmed MA, Karlsson MO, Holford N, Chasseloup E, Ahamadi M, Kowalski KG, Cole S, Kerwash E, Wade JR, Liu C, Wang Y, Trame MN, Zhu H, Wilkins JJ. Covariate modeling in pharmacometrics: General points for consideration. CPT Pharmacometrics Syst Pharmacol 2024; 13:710-728. [PMID: 38566433 PMCID: PMC11098153 DOI: 10.1002/psp4.13115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/15/2024] [Accepted: 02/05/2024] [Indexed: 04/04/2024] Open
Abstract
Modeling the relationships between covariates and pharmacometric model parameters is a central feature of pharmacometric analyses. The information obtained from covariate modeling may be used for dose selection, dose individualization, or the planning of clinical studies in different population subgroups. The pharmacometric literature has amassed a diverse, complex, and evolving collection of methodologies and interpretive guidance related to covariate modeling. With the number and complexity of technologies increasing, a need for an overview of the state of the art has emerged. In this article the International Society of Pharmacometrics (ISoP) Standards and Best Practices Committee presents perspectives on best practices for planning, executing, reporting, and interpreting covariate analyses to guide pharmacometrics decision making in academic, industry, and regulatory settings.
Collapse
Affiliation(s)
| | | | | | - Mariam A. Ahmed
- Quantitative Clinical Pharmacology, Takeda PharmaceuticalCambridgeMassachusettsUSA
| | | | - Nick Holford
- Department of Pharmacology & Clinical PharmacologyUniversity of AucklandAucklandNew Zealand
| | | | | | | | - Susan Cole
- Medical and Healthcare product Regulatory Agency (MHRA)LondonUK
| | - Essam Kerwash
- Medical and Healthcare product Regulatory Agency (MHRA)LondonUK
| | | | - Chao Liu
- Applied Innovation Quantitative Solutions, BeiGeneWashingtonDCUSA
| | - Yaning Wang
- Createrna Science and TechnologyClarksburgMarylandUSA
| | - Mirjam N. Trame
- Integrated Drug Development Northeast Regional LeadCertaraMassachusettsUSA
| | - Hao Zhu
- Division of Pharmacometrics, Office of Clinical PharmacologyCenter for Drug Evaluation and Research, Food and Drug AdministrationSilver SpringsMarylandUSA
| | | | | |
Collapse
|
7
|
Geng K, Shen C, Wang X, Wang X, Shao W, Wang W, Chen T, Sun H, Xie H. A physiologically-based pharmacokinetic/pharmacodynamic modeling approach for drug-drug-gene interaction evaluation of S-warfarin with fluconazole. CPT Pharmacometrics Syst Pharmacol 2024; 13:853-869. [PMID: 38487942 PMCID: PMC11098157 DOI: 10.1002/psp4.13123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 05/18/2024] Open
Abstract
Warfarin is a widely used anticoagulant, and its S-enantiomer has higher potency compared to the R-enantiomer. S-warfarin is mainly metabolized by cytochrome P450 (CYP) 2C9, and its pharmacological target is vitamin K epoxide reductase complex subunit 1 (VKORC1). Both CYP2C9 and VKORC1 have genetic polymorphisms, leading to large variations in the pharmacokinetics (PKs) and pharmacodynamics (PDs) of warfarin in the population. This makes dosage management of warfarin difficult, especially in the case of drug-drug interactions (DDIs). This study provides a whole-body physiologically-based pharmacokinetic/PD (PBPK/PD) model of S-warfarin for predicting the effects of drug-drug-gene interactions on S-warfarin PKs and PDs. The PBPK/PD model of S-warfarin was developed in PK-Sim and MoBi. Drug-dependent parameters were obtained from the literature or optimized. Of the 34 S-warfarin plasma concentration-time profiles used, 96% predicted plasma concentrations within twofold range compared to observed data. For S-warfarin plasma concentration-time profiles with CYP2C9 genotype, 364 of 386 predicted plasma concentration values (~94%) fell within the twofold of the observed values. This model was tested in DDI predictions with fluconazole as CYP2C9 perpetrators, with all predicted DDI area under the plasma concentration-time curve to the last measurable timepoint (AUClast) ratio within twofold of the observed values. The anticoagulant effect of S-warfarin was described using an indirect response model, with all predicted international normalized ratio (INR) within twofold of the observed values. This model also incorporates a dose-adjustment method that can be used for dose adjustment and predict INR when warfarin is used in combination with CYP2C9 perpetrators.
Collapse
Affiliation(s)
- Kuo Geng
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Chaozhuang Shen
- Department of Clinical Pharmacy and Pharmacy Administration, West China College of PharmacySichuan UniversityChengduSichuanChina
| | - Xiaohu Wang
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjingChina
| | - Xingwen Wang
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Wenxin Shao
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Wenhui Wang
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Tao Chen
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Hua Sun
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
| | - Haitang Xie
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
| |
Collapse
|
8
|
Xue L, Singla RK, Qin Q, Ding Y, Liu L, Ding X, Qu W, Huang C, Shen Z, Shen B, Miao L. Exploring the complex relationship between vitamin K, gut microbiota, and warfarin variability in cardiac surgery patients. Int J Surg 2023; 109:3861-3871. [PMID: 37598356 PMCID: PMC10720796 DOI: 10.1097/js9.0000000000000673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND AND OBJECTIVES Due to the high individual variability of anticoagulant warfarin, this study aimed to investigate the effects of vitamin K concentration and gut microbiota on individual variability of warfarin in 246 cardiac surgery patients. METHODS The pharmacokinetics and pharmacodynamics (PKPD) model predicted international normalized ratio (INR) and warfarin concentration. Serum and fecal samples were collected to detect warfarin and vitamin K [VK1 and menaquinone-4 (MK4)] concentrations and gut microbiota diversity, respectively. In addition, the patient's medical records were reviewed for demographic characteristics, drug history, and CYP2C9, VKORC1, and CYP4F2 genotypes. RESULTS The PKPD model predicted ideal values of 62.7% for S-warfarin, 70.4% for R-warfarin, and 76.4% for INR. The normal VK1 level was 1.34±1.12 nmol/ml (95% CI: 0.33-4.08 nmol/ml), and the normal MK4 level was 0.22±0.18 nmol/ml (95% CI: 0.07-0.63 nmol/ml). The MK4 to total vitamin K ratio was 16.5±9.8% (95% CI: 4.3-41.5%). The S-warfarin concentration of producing 50% of maximum anticoagulation and the half-life of prothrombin complex activity tended to increase with vitamin K. Further, Prevotella and Eubacterium of gut microbiota identified as the main bacteria associated with individual variability of warfarin. The results suggest that an increase in vitamin K concentration can decrease anticoagulation, and gut microbiota may influence warfarin anticoagulation through vitamin K2 synthesis. CONCLUSION This study highlights the importance of considering vitamin K concentration and gut microbiota when prescribing warfarin. The findings may have significant implications for the personalized use of warfarin. Further research is needed to understand better the role of vitamin K and gut microbiota in warfarin anticoagulation.
Collapse
Affiliation(s)
- Ling Xue
- Department of Pharmacy
- Department of Pharmacology, Faculty of Medicine, UPV/EHU, Spain
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | | | - Yinglong Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University
| | | | | | | | | | - Zhenya Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Liyan Miao
- Department of Pharmacy
- College of Pharmaceutical Sciences
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Jiangsu
| |
Collapse
|
9
|
Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther 2023; 8:386. [PMID: 37806986 PMCID: PMC10560686 DOI: 10.1038/s41392-023-01619-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 07/20/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
Individual variability in drug response (IVDR) can be a major cause of adverse drug reactions (ADRs) and prolonged therapy, resulting in a substantial health and economic burden. Despite extensive research in pharmacogenomics regarding the impact of individual genetic background on pharmacokinetics (PK) and pharmacodynamics (PD), genetic diversity explains only a limited proportion of IVDR. The role of gut microbiota, also known as the second genome, and its metabolites in modulating therapeutic outcomes in human diseases have been highlighted by recent studies. Consequently, the burgeoning field of pharmacomicrobiomics aims to explore the correlation between microbiota variation and IVDR or ADRs. This review presents an up-to-date overview of the intricate interactions between gut microbiota and classical therapeutic agents for human systemic diseases, including cancer, cardiovascular diseases (CVDs), endocrine diseases, and others. We summarise how microbiota, directly and indirectly, modify the absorption, distribution, metabolism, and excretion (ADME) of drugs. Conversely, drugs can also modulate the composition and function of gut microbiota, leading to changes in microbial metabolism and immune response. We also discuss the practical challenges, strategies, and opportunities in this field, emphasizing the critical need to develop an innovative approach to multi-omics, integrate various data types, including human and microbiota genomic data, as well as translate lab data into clinical practice. To sum up, pharmacomicrobiomics represents a promising avenue to address IVDR and improve patient outcomes, and further research in this field is imperative to unlock its full potential for precision medicine.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China.
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, PR China.
- Central Laboratory of Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, 410013, PR China.
| |
Collapse
|
10
|
Falkenhagen U, Knöchel J, Kloft C, Huisinga W. Deriving mechanism-based pharmacodynamic models by reducing quantitative systems pharmacology models: An application to warfarin. CPT Pharmacometrics Syst Pharmacol 2023; 12:432-443. [PMID: 36866520 PMCID: PMC10088086 DOI: 10.1002/psp4.12903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 03/04/2023] Open
Abstract
Quantitative systems pharmacology (QSP) models integrate comprehensive qualitative and quantitative knowledge about pharmacologically relevant processes. We previously proposed a first approach to leverage the knowledge in QSP models to derive simpler, mechanism-based pharmacodynamic (PD) models. Their complexity, however, is typically still too large to be used in the population analysis of clinical data. Here, we extend the approach beyond state reduction to also include the simplification of reaction rates, elimination of reactions, and analytic solutions. We additionally ensure that the reduced model maintains a prespecified approximation quality not only for a reference individual but also for a diverse virtual population. We illustrate the extended approach for the warfarin effect on blood coagulation. Using the model-reduction approach, we derive a novel small-scale warfarin/international normalized ratio model and demonstrate its suitability for biomarker identification. Due to the systematic nature of the approach in comparison with empirical model building, the proposed model-reduction algorithm provides an improved rationale to build PD models also from QSP models in other applications.
Collapse
Affiliation(s)
- Undine Falkenhagen
- Institute of Mathematics, University of Potsdam, Potsdam, Germany.,Graduate Research Training Program PharMetrX: Pharmacometrics & Computational Disease Modelling, Freie Universität Berlin and University of Potsdam, Potsdam, Germany
| | - Jane Knöchel
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Wilhelm Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| |
Collapse
|
11
|
Morse JD, Cortinez LI, Anderson BJ. Considerations for Intravenous Anesthesia Dose in Obese Children: Understanding PKPD. J Clin Med 2023; 12:1642. [PMID: 36836174 PMCID: PMC9960599 DOI: 10.3390/jcm12041642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The intravenous induction or loading dose in children is commonly prescribed per kilogram. That dose recognizes the linear relationship between volume of distribution and total body weight. Total body weight comprises both fat and fat-free mass. Fat mass influences the volume of distribution and the use of total body weight fails to recognize the impact of fat mass on pharmacokinetics in children. Size metrics alternative to total body mass (e.g., fat-free and normal fat mass, ideal body weight and lean body weight) have been proposed to scale pharmacokinetic parameters (clearance, volume of distribution) for size. Clearance is the key parameter used to calculate infusion rates or maintenance dosing at steady state. Dosing schedules recognize the curvilinear relationship, described using allometric theory, between clearance and size. Fat mass also has an indirect influence on clearance through both metabolic and renal function that is independent of its effects due to increased body mass. Fat-free mass, lean body mass and ideal body mass are not drug specific and fail to recognize the variable impact of fat mass contributing to body composition in children, both lean and obese. Normal fat mass, used in conjunction with allometry, may prove a useful size metric but computation by clinicians for the individual child is not facile. Dosing is further complicated by the need for multicompartment models to describe intravenous drug pharmacokinetics and the concentration effect relationship, both beneficial and adverse, is often poorly understood. Obesity is also associated with other morbidity that may also influence pharmacokinetics. Dose is best determined using pharmacokinetic-pharmacodynamic (PKPD) models that account for these varied factors. These models, along with covariates (age, weight, body composition), can be incorporated into programmable target-controlled infusion pumps. The use of target-controlled infusion pumps, assuming practitioners have a sound understanding of the PKPD within programs, provide the best available guide to intravenous dose in obese children.
Collapse
Affiliation(s)
- James Denzil Morse
- Department of Anaesthesiology, University of Auckland, Park Road, Auckland 1023, New Zealand
| | - Luis Ignacio Cortinez
- División Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Brian Joseph Anderson
- Department of Anaesthesiology, University of Auckland, Park Road, Auckland 1023, New Zealand
- Department of Anaesthesia, Auckland Children’s Hospital, Park Road, Private Bag 92024, Auckland 1023, New Zealand
| |
Collapse
|
12
|
Cheng S, Flora DR, Rettie AE, Brundage RC, Tracy TS. A Physiological-Based Pharmacokinetic Model Embedded with a Target-Mediated Drug Disposition Mechanism Can Characterize Single-Dose Warfarin Pharmacokinetic Profiles in Subjects with Various CYP2C9 Genotypes under Different Cotreatments. Drug Metab Dispos 2023; 51:257-267. [PMID: 36379708 PMCID: PMC9901215 DOI: 10.1124/dmd.122.001048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/10/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Warfarin, a commonly prescribed oral anticoagulant medication, is highly effective in treating deep vein thrombosis and pulmonary embolism. However, the clinical dosing of warfarin is complicated by high interindividual variability in drug exposure and response and its narrow therapeutic index. CYP2C9 genetic polymorphism and drug-drug interactions (DDIs) are substantial contributors to this high variability of warfarin pharmacokinetics (PK), among numerous factors. Building a physiology-based pharmacokinetic (PBPK) model for warfarin is not only critical for a mechanistic characterization of warfarin PK but also useful for investigating the complicated dose-exposure relationship of warfarin. Thus, the objective of this study was to develop a PBPK model for warfarin that integrates information regarding CYP2C9 genetic polymorphisms and their impact on DDIs. Generic PBPK models for both S- and R-warfarin, the two enantiomers of warfarin, were constructed in R with the mrgsolve package. As expected, a generic PBPK model structure did not adequately characterize the warfarin PK profile collected up to 15 days following the administration of a single oral dose of warfarin, especially for S-warfarin. However, following the integration of an empirical target-mediated drug disposition (TMDD) component, the PBPK-TMDD model well characterized the PK profiles collected for both S- and R-warfarin in subjects with different CYP2C9 genotypes. Following the integration of enzyme inhibition and induction effects, the PBPK-TMDD model also characterized the PK profiles of both S- and R-warfarin in various DDI settings. The developed mathematic framework may be useful in building algorithms to better inform the clinical dosing of warfarin. SIGNIFICANCE STATEMENT: The present study found that a traditional physiology-based pharmacokinetic (PBPK) model cannot sufficiently characterize the pharmacokinetic profiles of warfarin enantiomers when warfarin is administered as a single dose, but a PBPK model with a target-mediated drug disposition mechanism can. After incorporating CYP2C9 genotypes and drug-drug interaction information, the developed model is anticipated to facilitate the understanding of warfarin disposition in subjects with different CYP2C9 genotypes in the absence and presence of both cytochrome P450 inhibitors and cytochrome P450 inducers.
Collapse
Affiliation(s)
- Shen Cheng
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Twin Cities, Minnesota (S.C., D.R.F., R.C.B.); Tracy Consultants, Huntsville, Alabama (T.S.T.); and Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington (A.E.R.)
| | - Darcy R Flora
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Twin Cities, Minnesota (S.C., D.R.F., R.C.B.); Tracy Consultants, Huntsville, Alabama (T.S.T.); and Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington (A.E.R.)
| | - Allan E Rettie
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Twin Cities, Minnesota (S.C., D.R.F., R.C.B.); Tracy Consultants, Huntsville, Alabama (T.S.T.); and Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington (A.E.R.)
| | - Richard C Brundage
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Twin Cities, Minnesota (S.C., D.R.F., R.C.B.); Tracy Consultants, Huntsville, Alabama (T.S.T.); and Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington (A.E.R.)
| | - Timothy S Tracy
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Twin Cities, Minnesota (S.C., D.R.F., R.C.B.); Tracy Consultants, Huntsville, Alabama (T.S.T.); and Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington (A.E.R.)
| |
Collapse
|
13
|
Eaton MP, Nadtochiy SM, Stefanos T, LeMoine D, Anderson BJ. Delayed concentration effect models for dabigatran anticoagulation. Paediatr Anaesth 2022; 32:1113-1120. [PMID: 35735989 PMCID: PMC9541555 DOI: 10.1111/pan.14511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/24/2022] [Accepted: 06/19/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Dabigatran is an anticoagulant with potential use during cardiopulmonary bypass in children and adults. The pharmacokinetic-pharmacodynamic relationship for dabigatran anticoagulation effect was investigated in an intact animal model using rabbits. METHODS Ten male New Zealand white rabbits were given a novel preparation of intravenous dabigatran 15 mg.kg-1 . Blood samples were collected for activated clotting time, thromboelastometric reaction time, and drug assay at 5, 15, 30, 60, 120, 180, 300, and 420 min. Plasma dabigatran concentrations and coagulation measures were analyzed using an integrated pharmacokinetic-pharmacodynamic model using nonlinear mixed effects. Effects (activated clotting and thromboelastometric reaction times) were described using a sigmoidal EMAX model. Pharmacokinetic parameters were scaled using allometry and standardized to a 70 kg size standard. Pharmacodynamics were investigated using both an effect compartment model and an indirect response (turnover) model. RESULTS A two-compartment model described dabigatran pharmacokinetics with a clearance (CL 0.135 L.min-1 .70 kg-1 ), intercompartment clearance (Q 0.33 L.min-1 .70 kg-1 ), central volume of distribution (V1 12.3 L.70 kg-1 ), and peripheral volume of distribution (V2 30.1 L.70 kg-1 ). The effect compartment model estimates for a sigmoid EMAX model with activated clotting time had an effect site concentration (Ce50 20.1 mg.L-1 ) eliciting half of the maximal effect (EMAX 899 s) and a Hill coefficient (N 0.66). The equilibration half time (T1/2 keo) was 1.4 min. Results for the reaction time were plasma concentration (Cp50 65.3 mg.L-1 ), EMAX 34 min, N 0.80 with a baseline thromboelastometric reaction time of 0.4 min. The equilibration half time (T1/2 keo) was 2.04 min. CONCLUSIONS Dabigatran reversibly binds to the active site on the thrombin molecule, preventing thrombin-mediated activation of coagulation factors. The effect compartment model performed slightly better than the turnover model and was able to adequately capture pharmacodynamics for both activated clotting and thromboelastometric reaction times. The equilibration half time was short (<2 min). These data can be used to inform future animal preclinical studies for those undergoing cardiopulmonary bypass. These preclinical data also demonstrate the magnitude of parameter values for a delayed effect compartment model that are applicable to humans.
Collapse
Affiliation(s)
- Michael P. Eaton
- University of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| | - Sergiy M. Nadtochiy
- University of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| | - Tatsiana Stefanos
- University of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| | - Dana LeMoine
- University of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| | | |
Collapse
|
14
|
Cheng S, Flora DR, Rettie AE, Brundage RC, Tracy TS. Pharmacokinetic Modeling of Warfarin І - Model-based Analysis of Warfarin Enantiomers with a Target Mediated Drug Disposition Model Reveals CYP2C9 Genotype-dependent Drug-drug Interactions of S-Warfarin. Drug Metab Dispos 2022; 50:DMD-AR-2022-000876. [PMID: 35798369 PMCID: PMC9488981 DOI: 10.1124/dmd.122.000876] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
The objective of this study is to characterize the impact of CYP2C9 genotype on warfarin drug-drug interactions when warfarin is taken together with fluconazole, a cytochrome P450 (CYP) inhibitor, or rifampin, a CYP inducer with a nonlinear mixed effect modeling approach. A target mediated drug disposition model with a urine compartment was necessary to characterize both S-warfarin and R-warfarin plasma and urine pharmacokinetic profiles sufficiently. Following the administration of fluconazole, our study found subjects with CYP2C9 *2 or *3 alleles experience smaller changes in S-warfarin CL compared with subjects without these alleles (69.5%, 64.8%, 59.7% and 47.8% decrease in subjects with CYP2C9 *1/*1, *1/*3, *2/*3 and *3/*3 respectively). Whereas, following the administration of rifampin, subjects with CYP2C9 *2/*3 or CYP2C9 *3/*3 experience larger changes in S-warfarin CL compared with subjects with at least one copy of CYP2C9 *1 or *1B (115%, 111%, 119%, 198% and 193% increase in subjects with CYP2C9 *1/*1, *1B/*1B, *1/*3, *2/*3 and *3/*3 respectively). The results suggest different dose adjustments are potentially required for patients with different CYP2C9 genotypes if warfarin is administered together with CYP inhibitors or inducers. Significance Statement The present study found a target mediated drug disposition model is needed to sufficiently characterize the clinical pharmacokinetic profiles of warfarin racemates under different co-treatments in subjects with various CYP2C9 genotypes, following a single dose of warfarin administration. The study also found S-warfarin, the pharmacologically more active ingredient in warfarin, exhibits CYP2C9 genotype-dependent drug-drug interactions, which indicates the dose of warfarin may need to be adjusted differently in subjects with different CYP2C9 genotypes in the presence of drug-drug interactions.
Collapse
Affiliation(s)
| | - Darcy R Flora
- Present Affiliation: GRYT Health Inc., United States
| | - Allan E Rettie
- Dept. of Medicinal Chemistry, University of Washington, United States
| | - Richard C Brundage
- Experimental and Clinical Pharmacology, University of Minnesota, United States
| | | |
Collapse
|
15
|
Cheng S, Flora DR, Rettie AE, Brundage RC, Tracy TS. Pharmacokinetic Modeling of Warfarin ІI - Model-based Analysis of Warfarin Metabolites following Warfarin Administered either Alone or Together with Fluconazole or Rifampin. Drug Metab Dispos 2022; 50:DMD-AR-2022-000877. [PMID: 35798368 PMCID: PMC9488977 DOI: 10.1124/dmd.122.000877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
The objective of this study is to conduct a population pharmacokinetic (PK) model-based analysis on 10 warfarin metabolites (4'-, 6-, 7-, 8- and 10-hydroxylated (OH)-S- and R- warfarin), when warfarin is administered alone or together with either fluconazole or rifampin. One or two compartment PK models expanded from target mediated drug disposition (TMDD) models developed previously for warfarin enantiomers were able to sufficiently characterize the PK profiles of 10 warfarin metabolites in plasma and urine under different conditions. Model-based analysis shows CYP2C9 mediated metabolic elimination pathways are more inhibitable by fluconazole (% formation CL (CLf) of 6- and 7-OH-S-warfarin decrease: 73.2% and 74.8%) but less inducible by rifampin (% CLf of 6- and 7-OH-S-warfarin increase: 85% and 75%), compared with non-CYP2C9 mediated elimination pathways (% CLf of 10-OH-S-warfarin and CLR of S-warfarin decrease in the presence of fluconazole: 65.0% and 15.3%; % CLf of 4'- 8- and 10-OH-S-warfarin increase in the presence of rifampin: 260%, 127% and 355%), which potentially explains the CYP2C9 genotype-dependent DDIs exhibited by S-warfarin, when warfarin is administrated together with fluconazole or rifampin. Additionally, for subjects with CYP2C9 *2 and *3 variants, a model-based analysis of warfarin metabolite profiles in subjects with various CYP2C9 genotypes demonstrates CYP2C9 mediated elimination is less important and non-CYP2C9 mediated elimination is more important, compared with subjects without these variants. To our knowledge, this is so far one of the most comprehensive population-based PK analyses of warfarin metabolites in subjects with various CYP2C9 genotypes under different co-medications. Significance Statement The studies we wish to publish are potentially impactful. The need for a TMDD pharmacokinetic model and the demonstration of genotyped-dependent drug interactions may explain the extensive variability in dose-response relationships that are seen in the clinical dose adjustments of warfarin.
Collapse
Affiliation(s)
| | - Darcy R Flora
- Present Affiliation: GRYT Health Inc., United States
| | - Allan E Rettie
- Dept. of Medicinal Chemistry, University of Washington, United States
| | - Richard C Brundage
- Experimental and Clinical Pharmacology, University of Minnesota, United States
| | | |
Collapse
|
16
|
A novel, rapid and simple UHPLC-MS/MS method for quantification of warfarin in dried blood spots. Anal Biochem 2022; 647:114664. [PMID: 35300971 DOI: 10.1016/j.ab.2022.114664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/01/2022]
Abstract
Warfarin is a common first line anticoagulant with a narrow therapeutic window. Because of the large blood volume needed, previous warfarin determination methods were not applicable to small animals, such as mice. To reduce the number of small animals used needed, we developed and validated a sensitive rapid assay for the simultaneous detection of warfarin enantiomers in mouse dried blood spot (DBS) samples. Analytes were extracted by tert-butyl methyl ether and then separated by a chiral Cellulose-1 column with a mobile phase of 75% acetonitrile (containing 0.1% formic acid). The total chromatographic run time was 3 min. Negative mode electrospray ionization was used for MS/MS detection, where the monitored ion transitions were m/z 307.1 → 161.0 and 341.1 → 284.0 for warfarin and coumachlor (internal standard) respectively. The calibration curves were linear with a correlation coefficient of ≥0.994 for both enantiomers over a concentration range of 10-1000 ng/mL. The satisfactory accuracy and adequate reproducibility of both warfarin enantiomers were validated in terms of intra- and interday precision with mouse DBS cards. The samples were stable at room temperature for at least 14 days. The validated method was applied to a pharmacokinetic study in mice.
Collapse
|
17
|
Drug Response Diversity: A Hidden Bacterium? J Pers Med 2021; 11:jpm11050345. [PMID: 33922920 PMCID: PMC8146020 DOI: 10.3390/jpm11050345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 11/27/2022] Open
Abstract
Interindividual heterogeneity in response to treatment is a real public health problem. It is a factor that can be responsible not only for ineffectiveness or fatal toxicity but also for hospitalization due to iatrogenic effects, thus increasing the cost of patient care. Several research teams have been interested in what may be at the origin of these phenomena, particularly at the genetic level and the basal activity of organs dedicated to the inactivation and elimination of drug molecules. Today, a new branch is being set up, explaining the enigmatic part that could not be explained before. Pharmacomicrobiomics attempts to investigate the interactions between bacteria, especially those in the gut, and drug response. In this review, we provide a state of the art on what this field has brought as new information and discuss the challenges that lie ahead to see the real application in clinical practice.
Collapse
|
18
|
Wang Z, Xiang X, Liu S, Tang Z, Sun H, Parvez M, Ghim JL, Shin JG, Cai W. A physiologically based pharmacokinetic/pharmacodynamic modeling approach for drug-drug interaction evaluation of warfarin enantiomers with sorafenib. Drug Metab Pharmacokinet 2020; 39:100362. [PMID: 34242938 DOI: 10.1016/j.dmpk.2020.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 10/23/2022]
Abstract
Sorafenib was suggested to cause drug-drug interaction (DDI) with the common anticoagulant, warfarin based on published studies. The inhibition on CYP2C9 enzyme was thought to be the mechanism, but further studies are warranted. Thus, a mechanistic PBPK/PD model for warfarin enantiomers was developed to predict DDI potential with sorafenib, aiming at providing reference for the rational use of both drugs. PBPK models of warfarin enantiomers were constructed by Simcyp software. A mechanistic PK/PD model was built in NONMEM software. PBPK model of sorafenib was fitted via a top-down method. The final PBPK/PD model of warfarin enantiomers was verified and validated by different dosing regimens, ethnicities and genetic polymorphisms, and used to perform DDI simulations between warfarin racemate and sorafenib among general populations and sub-populations with various CYP2C9 and VKORC1 genotypes. Results suggested low DDI risk between warfarin and sorafenib for general populations. Potentially serious consequence was seen for those carrying both CYP2C9 ∗2 and ∗3 and VKORC1 A/A genotypes. This PBPK/PD modeling approach for warfarin enantiomers enabled DDI evaluation with sorafenib. Close monitoring and warfarin dosage adjustment were recommended for patients carrying mutant genotypes. The novel model could be applied to investigate other drugs that may interact with warfarin.
Collapse
Affiliation(s)
- Ziteng Wang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shuaibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhijia Tang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hong Sun
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Masud Parvez
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan, 614735, Republic of Korea
| | - Jong-Lyul Ghim
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan, 614735, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan, 614735, Republic of Korea.
| | - Weimin Cai
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
19
|
Xie C, Xue L, Zhang Y, Zhu J, Zhou L, Hang Y, Ding X, Jiang B, Miao L. Comparison of the prediction performance of different warfarin dosing algorithms based on Chinese patients. Pharmacogenomics 2020; 21:23-32. [PMID: 31849278 DOI: 10.2217/pgs-2019-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Aim: To compare the prediction performance of different warfarin dosing algorithms based on Chinese patients. Materials & methods: A total of 18 algorithms were tested in 325 patients. The predictive efficacy of selected algorithms was evaluated by calculating the percentage of patients whose predicted dose fell within ±20% of their actual stable warfarin dose and the mean absolute error. Results: The percentage within ± 20% and the mean absolute error of the algorithms ranged from 11.9 to 41.2% and -0.20 (-0.29 to -0.11) mg/d to -1.63 (-1.75 to -1.50) mg/d. The algorithms established by Miao et al. and Wei et al. had optimal predictive performance. Conclusion: Algorithms based on geographical populations might be more suitable for the prediction of stable warfarin doses in local patients.
Collapse
Affiliation(s)
- Cheng Xie
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Ling Xue
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Yuzhen Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Jianguo Zhu
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Ling Zhou
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Yongfu Hang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Xiaoliang Ding
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Bin Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Liyan Miao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| |
Collapse
|
20
|
Holford N, Ma G, Metz D. TDM is dead. Long live TCI! Br J Clin Pharmacol 2020; 88:1406-1413. [PMID: 32543717 PMCID: PMC9290673 DOI: 10.1111/bcp.14434] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/28/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
Twenty years ago, target concentration intervention (TCI) was distinguished from therapeutic drug monitoring (TDM). It was proposed that TCI would bring more clinical benefit because of the precision of the approach and the ability to link TCI to principles of pharmacokinetics and pharmacodynamics to predict the dose required by an individual (1). We examine the theory and clinical trial evidence supporting the benefits of TCI over TDM and conclude that in the digital age TDM should be abandoned and replaced by TCI.
Collapse
Affiliation(s)
- Nick Holford
- Department of Pharmacology & Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Guangda Ma
- Department of Pharmacology & Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - David Metz
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
The gut microbes, Enterococcus and Escherichia-Shigella, affect the responses of heart valve replacement patients to the anticoagulant warfarin. Pharmacol Res 2020; 159:104979. [PMID: 32505835 DOI: 10.1016/j.phrs.2020.104979] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
Numerous algorithms based on patient genetic variants have been established with the aim of reducing the risk of GI bleeding and thromboembolism during warfarin administration. However, approximately 35 % of individual warfarin sensitivity still remains unexplained. Few of warfarin algorithms take into account gut microbiota profiles. The identification of certain microbiome will provide new targets and new strategies for reducing the risk of bleeding and thromboembolism during warfarin administration. In this study, we collected plasma and stool samples from 200 inpatients undergoing heart valve replacement (HVR), which were classified as low responder (LR), high responder (HR) and normal responder (NR). Significant differences were observed in the diversity and relative abundance of the gut microbiota among the three groups. The genus Escherichia-Shigella was enriched significantly in the LRs (P = 3.189e-11), while the genus Enterococcus was enriched significantly in the HRs (P = 1.249e-11). The amount of VK2 synthesized by gut microbiota in LR group was much higher than that in HR group (P = 0.005). Whole genome shotgun sequencing indicated that the relative abundance of enzymes and modules associated with VK biosynthesis was significantly higher in LRs than in HRs or NRs. The 12 microbial markers were identified through tenfold cross-validation with a random forest model. The results provided a new microbial diagnostic model that can be used to inform modulation of warfarin dosage on the basis of patient intestinal flora composition.
Collapse
|
22
|
Wright DFB, Martin JH, Cremers S. Spotlight Commentary: Model-informed precision dosing must demonstrate improved patient outcomes. Br J Clin Pharmacol 2019; 85:2238-2240. [PMID: 31400011 DOI: 10.1111/bcp.14050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
| | - Jennifer H Martin
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Serge Cremers
- Departments of Pathology and Cell Biology, and Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
23
|
Helin TA, Joutsi-Korhonen L, Asmundela H, Niemi M, Orpana A, Lassila R. Warfarin dose requirement in patients having severe thrombosis or thrombophilia. Br J Clin Pharmacol 2019; 85:1684-1691. [PMID: 30933373 DOI: 10.1111/bcp.13948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/12/2019] [Accepted: 03/23/2019] [Indexed: 12/17/2022] Open
Abstract
AIMS Warfarin dose requirement varies significantly. We compared the clinically established doses based on international normalized ratio (INR) among patients with severe thrombosis and/or thrombophilia with estimates from genetic dosing algorithms. METHODS Fifty patients with severe thrombosis and/or thrombophilia requiring permanent anticoagulation, referred to the Helsinki University Hospital Coagulation Center, were screened for thrombophilias and genotyped for CYP2C9*2 (c.430C>T, rs1799853), CYP2C9*3 (c.1075A>C, rs1057910) and VKORC1 c.-1639G>A (rs9923231) variants. The warfarin maintenance doses (target INR 2.0-3.0 in 94%, 2.5-3.5 in 6%) were estimated by the Gage and the International Warfarin Pharmacogenetics Consortium (IWPC) algorithms. The individual warfarin maintenance dose was tailored, supplementing estimates with comprehensive clinical evaluation and INR data. RESULTS Mean patient age was 47 years (range 20-76), and BMI 27 (SD 6), 68% being women. Forty-six (92%) had previous venous or arterial thrombosis, and 26 (52%) had a thrombophilia, with 22% having concurrent aspirin. A total of 40% carried the CYP2C9*2 or *3 allele and 54% carried the VKORC1-1639A allele. The daily mean maintenance dose of warfarin estimated by the Gage algorithm was 5.4 mg (95% CI 4.9-5.9 mg), and by the IWPC algorithm was 5.2 mg (95% CI 4.7-5.7 mg). The daily warfarin maintenance dose after clinical visits and follow-up was higher than the estimates, mean 6.9 mg (95% CI 5.6-8.2 mg, P < 0.006), with highest dose in patients having multiple thrombophilic factors (P < 0.03). CONCLUSIONS In severe thrombosis and/or thrombophilia, variation in thrombin generation and pharmacodynamics influences warfarin response. Pharmacogenetic dosing algorithms seem to underestimate dose requirement.
Collapse
Affiliation(s)
- Tuukka A Helin
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Heidi Asmundela
- Coagulation Disorders Unit, Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Arto Orpana
- Genetics and Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Riitta Lassila
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Coagulation Disorders Unit, Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
24
|
Awad ME, Padela MT, Sayeed Z, El-Othmani MM, Zekaj M, Darwiche HF, Saleh KJ. Pharmacogenomic Testing for Postoperative Pain Optimization Before Total Joint Arthroplasty: A Focus on Drug-Drug-Gene Interaction with Commonly Prescribed Drugs and Prior Opioid Use. JBJS Rev 2019; 7:e2. [PMID: 31094889 DOI: 10.2106/jbjs.rvw.18.00058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Mohamed E Awad
- Resident Research Partnership, Detroit, Michigan.,FAJR Scientific, Detroit, Michigan.,Michigan State University College of Osteopathic Medicine, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan
| | - Muhammad Talha Padela
- Resident Research Partnership, Detroit, Michigan.,FAJR Scientific, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery, Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois
| | - Zain Sayeed
- Resident Research Partnership, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan.,Department of Orthopaedic Surgery, Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois
| | - Mouhanad M El-Othmani
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Mark Zekaj
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Hussein F Darwiche
- Department of Orthopaedic Surgery and Sports Medicine, Detroit Medical Center, Detroit, Michigan
| | - Khaled J Saleh
- FAJR Scientific, Detroit, Michigan.,Michigan State University College of Osteopathic Medicine, Detroit, Michigan.,John D. Dingell VA Medical Center, Detroit, Michigan
| |
Collapse
|
25
|
Xue L, Zhang Y, Xie C, Zhou L, Liu L, Zhang H, Xu L, Song H, Lin M, Qiu H, Zhu J, Zhu Y, Zou J, Zhuang W, Xuan B, Chen Y, Fan Y, Wu D, Shen Z, Miao L. Relationship between warfarin dosage and international normalized ratio: a dose–response analysis and evaluation based on multicenter data. Eur J Clin Pharmacol 2019; 75:785-794. [DOI: 10.1007/s00228-019-02655-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/22/2019] [Indexed: 12/27/2022]
|
26
|
Hu K, Li Y, Ding R, Zhai Y, Chen L, Qian W, Yang J. A simple, sensitive, and high-throughput LC-APCI-MS/MS method for simultaneous determination of vitamin K 1, vitamin K 1 2,3-epoxide in human plasma and its application to a clinical pharmacodynamic study of warfarin. J Pharm Biomed Anal 2018; 159:82-91. [PMID: 29980023 DOI: 10.1016/j.jpba.2018.06.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 01/10/2023]
Abstract
Warfarin exerts its anticoagulation activity by blocking the vitamin K-epoxide cycle. A quantitative understanding of how warfarin and related genes interact with the vitamin K-epoxide cycle and the associated change of coagulation activity in the human body may help study the pharmacodynamics of warfarin. The plasma concentration of vitamin K1 (VK1) and vitamin K1 2,3-epoxide (VK1O) could reflect the status of vitamin K-epoxide cycle. However, their determination is a challenging task due to their extremely low concentrations in human plasma and the severe interferences caused by co-extracted lipids. In this study, we developed an LC-APCI-MS/MS method for the simultaneous determination of VK1 and VK1O in human plasma using stable deuterium-labeled vitamin K1 (vitamin K1-d7) as the internal standard (IS). Plasma samples were prepared through protein denaturation followed by one-step liquid extraction with cyclohexane. Chromatographic separation of analytes from isobaric interferences and endogenous ion suppressor was performed on a Synergi Hydro-RP column (150 mm × 4.6 mm, 4 μm) under the reversed-phase condition with isocratic elution. The selective reaction monitoring (SRM) transitions were chosen as m/z = 451.5→187.3 for VK1, m/z = 467.5→161.2 for VK1O, and m/z = 458.6→194.3 for IS in APCI positive mode. The assay was linear in the range of 100-10,000 pg/mL for the two analytes and achieved considerable extraction recoveries (87.8-93.3%, 91.0-96.9%, and 92.0% for VK1, VK1O, and IS, respectively), negligible matrix effects (93.6-96.0%, 96.3-100.1%, and 95.5%), and high selectivity with a small sample volume requirement (0.2 mL) and short run time (15 min). The validated method was successfully applied in a clinical pharmacodynamic study of warfarin, and the clotting activity was found to be negatively correlated with the plasma concentration ratio of VK1O to VK1.
Collapse
Affiliation(s)
- Kuan Hu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Li
- Department of Pharmacy, Xinjiang Medical University, Urumchi 830011, China
| | - Ru Ding
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yu Zhai
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Lin Chen
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Qian
- Department of Phase I Clinical Trials, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Jin Yang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
27
|
Liu S, Xue L, Shi X, Sun Z, Zhu Z, Zhang X, Tian X. Population pharmacokinetics and pharmacodynamics of ticagrelor and AR-C124910XX in Chinese healthy male subjects. Eur J Clin Pharmacol 2018; 74:745-754. [DOI: 10.1007/s00228-018-2427-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/01/2018] [Indexed: 11/28/2022]
|
28
|
McLay JS, Engelhardt T, Mohammed BS, Cameron G, Cohen MN, Galinkin JL, Christians U, Avram MJ, Henthorn TK, Dsida RM, Hawwa AF, Anderson BJ. The pharmacokinetics of intravenous ketorolac in children aged 2 months to 16 years: A population analysis. Paediatr Anaesth 2018; 28:80-86. [PMID: 29266539 DOI: 10.1111/pan.13302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Intravenous ketorolac is commonly administered to children for the control of postoperative pain. An effect site EC50 for analgesia of 0.37 mg. L-1 is described in adults. AIMS The aim of this study was to review age- and weight-related effects on ketorolac pharmacokinetic parameters in children and current dosing schedules. METHODS Pooled intravenous ketorolac (0.5 mg. kg-1 ) concentration-time data in children aged 2 months to 16 years were analyzed using nonlinear mixed-effects models. Allometry was used to scale to a 70 kg person. RESULTS There were 64 children aged 2 months to 16 years (641 plasma concentrations) available for analysis. A two-compartment mammillary model was used to describe pharmacokinetics. Clearance was 2.53 (CV 45.9%) L. h-1. 70 kg-1 and intercompartment clearance was 4.43 (CV 95.6%) L. h-1. 70 kg-1 . Both central (V1) and peripheral (V2) volumes of distribution decreased with age over the first few years of postnatal life to reach V1 6.89 (CV 30.3%) L. 70 kg-1 and V2 5.53 (CV 47.6%) L. 70 kg-1 . CONCLUSION Clearance, expressed as L. h-1. kg-1 , decreased with age from infancy. A dosing regimen of 0.5 mg. kg-1 every 6 hours maintains a trough concentration larger than 0.37 mg. L-1 in children 9 months to 16 years of age. This dosing regimen is consistent with current recommendations.
Collapse
Affiliation(s)
- James S McLay
- The Department of Child Health, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Thomas Engelhardt
- The Department of Paediatric Anaesthesia, Royal Aberdeen Children's Hospital, Aberdeen, UK
| | - Baba S Mohammed
- Pharmacology Unit, University of Development Studies, Tamale, Ghana
| | - Gary Cameron
- The Department of Child Health, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Mindy N Cohen
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jeffrey L Galinkin
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael J Avram
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas K Henthorn
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard M Dsida
- Department of Anesthesiology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | | | - Brian J Anderson
- Department of Anaesthesiology, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
Xue L, Holford N, Miao LY. Response to R-warfarin anticoagulant effect. Br J Clin Pharmacol 2017; 83:2305-2306. [PMID: 28735507 DOI: 10.1111/bcp.13344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/06/2017] [Indexed: 11/28/2022] Open
Affiliation(s)
- Ling Xue
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Nick Holford
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Li-Yan Miao
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Erratum. Br J Clin Pharmacol 2017; 83:1602. [DOI: 10.1111/bcp.13325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
31
|
Abstract
This tutorial describes sources of pharmacokinetic variability that are not obviously linked to genetic differences. The sources of variability are therefore described as environmental. The major quantitative sources of environmental variability are body size (including body composition), maturation and organ function. Size should be considered in all patients. Maturation is mainly relevant to neonates and infants less than 2 years of age. Renal function is the most important predictable source of variability due to differences in organ function.
Collapse
Affiliation(s)
- Nick Holford
- Department of Pharmacology & Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Padrini R, Quintieri L. R-warfarin anticoagulant effect. Br J Clin Pharmacol 2017; 83:2303-2304. [PMID: 28493597 DOI: 10.1111/bcp.13300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/27/2017] [Accepted: 04/04/2017] [Indexed: 01/07/2023] Open
Affiliation(s)
- Roberto Padrini
- Dipartimento di Medicina - DIMED, Università degli Studi di Padova, via Giustiniani 2, 35128, Padova, Italy
| | - Luigi Quintieri
- Dipartimento di Scienze del Farmaco - DSF, Università degli Studi di Padova, via Marzolo 5, 35131, Padova, Italy
| |
Collapse
|
33
|
Ferrari M, Pengo V, Barolo M, Bezzo F, Padrini R. Assessing the relative potency of (S)- and (R)-warfarin with a new PK-PD model, in relation to VKORC1 genotypes. Eur J Clin Pharmacol 2017; 73:699-707. [PMID: 28382498 DOI: 10.1007/s00228-017-2248-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE The purpose of this study is to develop a new pharmacokinetic-pharmacodynamic (PK-PD) model to characterise the contribution of (S)- and (R)-warfarin to the anticoagulant effect on patients in treatment with rac-warfarin. METHODS Fifty-seven patients starting warfarin (W) therapy were studied, from the first dose and during chronic treatment at INR stabilization. Plasma concentrations of (S)- and (R)-W and INRs were measured 12, 36 and 60 h after the first dose and at steady state 12-14 h after dosing. Patients were also genotyped for the G>A VKORC1 polymorphism. The PK-PD model assumed a linear relationship between W enantiomer concentration and INR and included a scaling factor k to account for a different potency of (R)-W. Two parallel compartment chains with different transit times (MTT1 and MTT2) were used to model the delay in the W effect. PD parameters were estimated with the maximum likelihood approach. RESULTS The model satisfactorily described the mean time-course of INR, both after the initial dose and during long-term treatment. (R)-W contributed to the rac-W anticoagulant effect with a potency of about 27% that of (S)-W. This effect was independent of VKORC1 genotype. As expected, the slope of the PK/PD linear correlation increased stepwise from GG to GA and from GA to AA VKORC1 genotype (0.71, 0.90 and 1.49, respectively). CONCLUSIONS Our PK-PD linear model can quantify the partial pharmacodynamic activity of (R)-W in patients contemporaneously exposed to therapeutic (S)-W plasma levels. This concept may be useful in improving the performance of future algorithms aiming at identifying the most appropriate W maintenance dose.
Collapse
Affiliation(s)
- Myriam Ferrari
- Computer-Aided Process Engineering Laboratory (CAPE-lab), Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131, Padua, Italy
| | - Vittorio Pengo
- Department of Cardiological, Thoracic and Vascular Sciences, University of Padova Medical School, Via Giustiniani 2, 35128, Padua, Italy
| | - Massimiliano Barolo
- Computer-Aided Process Engineering Laboratory (CAPE-lab), Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131, Padua, Italy
| | - Fabrizio Bezzo
- Computer-Aided Process Engineering Laboratory (CAPE-lab), Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131, Padua, Italy
| | - Roberto Padrini
- Department of Medicine (DIMED), University of Padova Medical School, Via Giustiniani 2, 35128, Padua, Italy.
| |
Collapse
|
34
|
Xue L, Holford N, Ding XL, Shen ZY, Huang CR, Zhang H, Zhang JJ, Guo ZN, Xie C, Zhou L, Chen ZY, Liu LS, Miao LY. Theory-based pharmacokinetics and pharmacodynamics of S- and R-warfarin and effects on international normalized ratio: influence of body size, composition and genotype in cardiac surgery patients. Br J Clin Pharmacol 2016; 83:823-835. [PMID: 27763679 DOI: 10.1111/bcp.13157] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 10/09/2016] [Accepted: 10/15/2016] [Indexed: 11/30/2022] Open
Abstract
AIMS The aims of this study are to apply a theory-based mechanistic model to describe the pharmacokinetics (PK) and pharmacodynamics (PD) of S- and R-warfarin. METHODS Clinical data were obtained from 264 patients. Total concentrations for S- and R-warfarin were measured by ultra-high performance liquid tandem mass spectrometry. Genotypes were measured using pyrosequencing. A sequential population PK parameter with data method was used to describe the international normalized ratio (INR) time course. Data were analyzed with NONMEM. Model evaluation was based on parameter plausibility and prediction-corrected visual predictive checks. RESULTS Warfarin PK was described using a one-compartment model. CYP2C9 *1/*3 genotype had reduced clearance for S-warfarin, but increased clearance for R-warfarin. The in vitro parameters for the relationship between prothrombin complex activity (PCA) and INR were markedly different (A = 0.560, B = 0.386) from the theory-based values (A = 1, B = 0). There was a small difference between healthy subjects and patients. A sigmoid Emax PD model inhibiting PCA synthesis as a function of S-warfarin concentration predicted INR. Small R-warfarin effects was described by competitive antagonism of S-warfarin inhibition. Patients with VKORC1 AA and CYP4F2 CC or CT genotypes had lower C50 for S-warfarin. CONCLUSION A theory-based PKPD model describes warfarin concentrations and clinical response. Expected PK and PD genotype effects were confirmed. The role of predicted fat free mass with theory-based allometric scaling of PK parameters was identified. R-warfarin had a minor effect compared with S-warfarin on PCA synthesis. INR is predictable from 1/PCA in vivo.
Collapse
Affiliation(s)
- Ling Xue
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Nick Holford
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Xiao-Liang Ding
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhen-Ya Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chen-Rong Huang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hua Zhang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jing-Jing Zhang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhe-Ning Guo
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Xie
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ling Zhou
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhi-Yao Chen
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Lin-Sheng Liu
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Li-Yan Miao
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| |
Collapse
|