1
|
Li R, Sun F, Feng Z, Zhang Y, Lan Y, Yu H, Li Y, Mao J, Zhang W. Evaluation and application of population pharmacokinetic models for optimising linezolid treatment in non-adherence multidrug-resistant tuberculosis patients. Eur J Pharm Sci 2024; 203:106915. [PMID: 39341464 DOI: 10.1016/j.ejps.2024.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Population pharmacokinetic (popPK) models can optimise linezolid dosage regimens in patients with multidrug-resistant tuberculosis (MDR-TB); however, unknown cross-centre precision and poor adherence remain problematic. This study aimed to assess the predictive ability of published models and use the most suitable model to optimise dosage regimens and manage compliance. METHODS One hundred fifty-eight linezolid plasma concentrations from 27 patients with MDR-TB were used to assess the predictive performance of published models. Prediction-based metrics and simulation-based visual predictive checks were conducted to evaluate predictive ability. Individualised remedial dosing regimens for various delayed scenarios were optimised using the most suitable model and Monte Carlo simulations. The influence of covariates, scheduled dosing intervals, and patient compliance were assessed. RESULTS Seven popPK models were identified. Body weight and creatinine clearance were the most frequently identified covariates influencing linezolid clearance. The model with the best performance had a median prediction error (PE%) of -1.62 %, median absolute PE of 29.50 %, and percentages of PE within 20 % (F20, 36.97 %) and 30 % (F30, 51.26 %). Monte Carlo simulations indicated that a twice-daily 300 mg linezolid dose may be more efficient than 600 mg once daily. For the 'typical' patient treated with 300 mg twice daily, half the dosage should be taken after a delay of ≥ 3 h. CONCLUSIONS Monte Carlo simulations based on popPK models can propose remedial regimens for delayed doses of linezolid in patients with MDR-TB. Model-based compliance management patterns are useful for balancing efficacy, adverse reactions, and resistance suppression.
Collapse
Affiliation(s)
- Rong Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng Sun
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Feng
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yilin Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanbo Lan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; Department of Tuberculosis, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Hongying Yu
- Department of Infectious Diseases, Hunan University of Medicine General Hospital, Huaihua, Hunan, 418000, China
| | - Yang Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| | - Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Centre for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Ponthier L, Franck B, Autmizguine J, Labriffe M, Ovetchkine P, Marquet P, Åsberg A, Woillard JB. Application of machine-learning models to predict the ganciclovir and valganciclovir exposure in children using a limited sampling strategy. Antimicrob Agents Chemother 2024; 68:e0086024. [PMID: 39194260 PMCID: PMC11459947 DOI: 10.1128/aac.00860-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Intravenous ganciclovir and oral valganciclovir display significant variability in ganciclovir pharmacokinetics, particularly in children. Therapeutic drug monitoring currently relies on the area under the concentration-time (AUC). Machine-learning (ML) algorithms represent an interesting alternative to Maximum-a-Posteriori Bayesian-estimators for AUC estimation. The goal of our study was to develop and validate an ML-based limited sampling strategy (LSS) approach to determine ganciclovir AUC0-24 after administration of either intravenous ganciclovir or oral valganciclovir in children. Pharmacokinetic parameters from four published population pharmacokinetic models, in addition to the World Health Organization growth curve for children, were used in the mrgsolve R package to simulate 10,800 pharmacokinetic profiles of children. Different ML algorithms were trained to predict AUC0-24 based on different combinations of two or three samples. Performances were evaluated in a simulated test set and in an external data set of real patients. The best estimation performances in the test set were obtained with the Xgboost algorithm using a 2 and 6 hours post dose LSS for oral valganciclovir (relative mean prediction error [rMPE] = 0.4% and relative root mean square error [rRMSE] = 5.7%) and 0 and 2 hours post dose LSS for intravenous ganciclovir (rMPE = 0.9% and rRMSE = 12.4%). In the external data set, the performance based on these two sample LSS was acceptable: rMPE = 0.2% and rRMSE = 16.5% for valganciclovir and rMPE = -9.7% and rRMSE = 17.2% for intravenous ganciclovir. The Xgboost algorithm developed resulted in a clinically relevant individual estimation using only two blood samples. This will improve the implementation of AUC-targeted ganciclovir therapeutic drug monitoring in children.
Collapse
Affiliation(s)
- Laure Ponthier
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, Limoges, France
- Department of Pediatrics, University Hospital of Limoges, Limoges, France
| | - Bénédicte Franck
- Department of Clinical and Biological Pharmacology and Pharmacovigilance, Clinical Investigation Center CIC-P 1414, Rennes, France
| | - Julie Autmizguine
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
- Research Center, Center Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
- Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Marc Labriffe
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Philippe Ovetchkine
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
| | - Pierre Marquet
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital—Rikshospitalet, Oslo, Norway
- Section of Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Jean-Baptiste Woillard
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| |
Collapse
|
3
|
Marquet P. Getting Tacrolimus Dosing Right. Ther Drug Monit 2024:00007691-990000000-00270. [PMID: 39357034 DOI: 10.1097/ftd.0000000000001266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024]
Abstract
ABSTRACT Tacrolimus (TAC) dosing is typically guided by the trough concentration (C0). Yet, significant relationships between TAC C0 and clinical outcomes have seldom been reported or only with adverse events. Large retrospective studies found a moderate correlation between TAC C0 and the area under the curve (AUC), where, for any given C0 value, the AUC varied 3- to 4-fold between patients (and vice versa). However, no randomized controlled trial evaluating the dose adjustment based on TAC AUC has been conducted yet. A few observational studies have shown that the AUC is associated with efficacy and, to a lesser extent, adverse effects. Other studies showed the feasibility of reaching predefined target ranges and reducing underexposure and overexposure. TAC AUC0-12 h is now most often assessed using Bayesian estimation, but machine learning is a promising approach. Microsampling devices are well accepted by patients and represent a valuable alternative to venous blood sample collection during hospital visits, especially when a limited sampling strategy is required. As AUC monitoring cannot be proposed very frequently, C0 monitoring has to be used in the interim, which has led to fluctuating doses in patients with an AUC/C0 ratio far from the population mean, because of different dose recommendations between the 2 biomarkers. We proposed estimating the individual AUC/C0 ratio and derived individual C0 targets to be used in between or as a replacement for AUC monitoring. Existing technology and evidence are now sufficient to propose AUC monitoring interspersed with individualized-C0 monitoring for all patients with kidney transplants while collecting real-world data to strengthen the evidence.
Collapse
Affiliation(s)
- Pierre Marquet
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, France; and
- Pharmacology & Transplantation, UMR1248 Inserm, Université de Limoges, CHU de Limoges, France
| |
Collapse
|
4
|
Ma P, Shang S, Liu R, Dong Y, Wu J, Gu W, Yu M, Liu J, Li Y, Chen Y. Prediction of teicoplanin plasma concentration in critically ill patients: a combination of machine learning and population pharmacokinetics. J Antimicrob Chemother 2024:dkae292. [PMID: 39207798 DOI: 10.1093/jac/dkae292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Teicoplanin has been widely used in patients with infections caused by Staphylococcus aureus, especially for critically ill patients. The pharmacokinetics (PK) of teicoplanin vary between individuals and within the same individual. We aim to establish a prediction model via a combination of machine learning and population PK (PPK) to support personalized medication decisions for critically ill patients. METHODS A retrospective study was performed incorporating 33 variables, including PPK parameters (clearance and volume of distribution). Multiple algorithms and Shapley additive explanations were employed for feature selection of variables to determine the strongest driving factors. RESULTS The performance of each algorithm with PPK parameters was superior to that without PPK parameters. The composition of support vector regression, categorical boosting and a backpropagation neural network (7:2:1) with the highest R2 (0.809) was determined as the final ensemble model. The model included 15 variables after feature selection, of which the predictive performance was superior to that of models considering all variables or using only PPK. The R2, mean absolute error, mean squared error, absolute accuracy (±5 mg/L) and relative accuracy (±30%) of external validation were 0.649, 3.913, 28.347, 76.12% and 76.12%, respectively. CONCLUSIONS Our study offers a non-invasive, fast and cost-effective prediction model of teicoplanin plasma concentration in critically ill patients. The model serves as a fundamental tool for clinicians to determine the effective plasma concentration range of teicoplanin and formulate individualized dosing regimens accordingly.
Collapse
Affiliation(s)
- Pan Ma
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Shenglan Shang
- Department of Clinical Pharmacy, General Hospital of Central Theater Command, Wuhan, Hubei Province 430070, China
| | - Ruixiang Liu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Yuzhu Dong
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Jiangfan Wu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenrui Gu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Mengchen Yu
- Department of Clinical Pharmacy, General Hospital of Central Theater Command, Wuhan, Hubei Province 430070, China
| | - Jing Liu
- Department of Clinical Pharmacy, General Hospital of Central Theater Command, Wuhan, Hubei Province 430070, China
| | - Ying Li
- Medical Big Data and Artificial Intelligence Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Yongchuan Chen
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| |
Collapse
|
5
|
Rivals F, Goutelle S, Codde C, Garreau R, Ponthier L, Marquet P, Ferry T, Labriffe M, Destere A, Woillard JB. A Machine Learning Algorithm to Predict the Starting Dose of Daptomycin. Clin Pharmacokinet 2024; 63:1137-1146. [PMID: 39085523 DOI: 10.1007/s40262-024-01405-z] [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] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND AND OBJECTIVE The dosage of daptomycin is usually based on body weight. However, it has been shown that this approach yields too high an exposure in obese patients. Pharmacokinetic and pharmacodynamic indexes (PK/PD) have been proposed for daptomycin's antibacterial effect (AUC/CMI >666) and toxicity (C0 > 24.3 mg/L). We previously developed machine learning (ML) algorithms to predict starting doses based on Monte Carlo simulations. We propose a new way to perform probability of target attainment based on an ML algorithm to predict the daptomycin starting dose. METHODS The Dvorchik model of daptomycin was implemented in the mrgsolve R package and 4950 pharmacokinetic profiles were simulated with doses ranging from 4 to 12 mg/kg. We trained and benchmarked four machine learning algorithms and selected the best to iteratively search for the optimal dose of daptomycin maximizing the event (AUC/CMI > 666 and C0 < 24.3 mg/L). The ML algorithm was evaluated in simulations and an external database of real patients in comparison with population pharmacokinetics. RESULTS The performance of the Xgboost algorithms developed to predict the event (ROC AUC) in the training and test set were 0.762 and 0.761, respectively. The most important prediction variables were dose, creatinine clearance, body weight and sex. In the external database of real patients, the starting dose administered based on the ML algorithm significantly improved the target attainment by 7.9% (p-value = 0.02929) in comparison with the dose administered based on body weight. CONCLUSION The developed algorithm improved the target attainment for daptomycin in comparison with weight-based dosing. We built a Shiny app to calculate the optimal starting dose.
Collapse
Affiliation(s)
- Florence Rivals
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Limoges, France
| | - Sylvain Goutelle
- Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France
- UMR CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, Univ Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
- Faculté de Médecine et de Pharmacie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Cyrielle Codde
- Service de Maladies Infectieuses et Tropicales, CHU Limoges, Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Transplantation, U 12482 rue du Pr Descottes, 87000, Limoges, France
| | - Romain Garreau
- Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France
- UMR CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, Univ Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
- Faculté de Médecine et de Pharmacie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laure Ponthier
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Transplantation, U 12482 rue du Pr Descottes, 87000, Limoges, France
| | - Pierre Marquet
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Transplantation, U 12482 rue du Pr Descottes, 87000, Limoges, France
| | - Tristan Ferry
- Faculté de Médecine et de Pharmacie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Service des Maladies Infectieuses et Tropicales, Centre de Référence pour la prise en charge des Infections Ostéo-Articulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Groupement Hospitalier Nord, Hôpital de la Croix-Rousse, Lyon, France
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université́ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, 69007, Lyon, France
| | - Marc Labriffe
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Transplantation, U 12482 rue du Pr Descottes, 87000, Limoges, France
| | | | - Jean-Baptiste Woillard
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Limoges, France.
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Transplantation, U 12482 rue du Pr Descottes, 87000, Limoges, France.
| |
Collapse
|
6
|
Liao R, Chen L, Cheng X, Li H, Wang T, Dong Y, Dong H. Estimation of linezolid exposure in patients with hepatic impairment using machine learning based on a population pharmacokinetic model. Eur J Clin Pharmacol 2024; 80:1241-1251. [PMID: 38717625 DOI: 10.1007/s00228-024-03698-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/01/2024] [Indexed: 07/06/2024]
Abstract
PURPOSE To investigate the pharmacokinetic changes of linezolid in patients with hepatic impairment and to explore a method to predict linezolid exposure. METHODS Patients with hepatic impairment who received linezolid were recruited. A population pharmacokinetic model (PPK) was then built using NONMEM software. And based on the final model, virtual patients with rich concentration values was constructed through Monte Carlo simulations (MCS), which were used to build machine learning (ML) models to predict linezolid exposure levels. Finally, we investigated the risk factors for thrombocytopenia in patients included. RESULTS A PPK model with population typical values of 3.83 L/h and 34.1 L for clearance and volume of distribution was established, and the severe hepatic impairment was identified as a significant covariate of clearance. Then, we built a series of ML models to predict the area under 0 -24 h concentration-time curve (AUC0-24) of linezolid based on virtual patients from MCS. The results showed that the Xgboost models showed the best predictive performance and were superior to the methods for estimating linezolid AUC0-24 based on though concentration or daily dose. Finally, we found that baseline platelet count, linezolid AUC0-24, and combination with fluoroquinolones were independent risk factors for thrombocytopenia, and based on this, we proposed a method for calculating the toxicity threshold of linezolid. CONCLUSION In this study, we successfully constructed a PPK model for patients with hepatic impairment and used ML algorithm to estimate linezolid AUC0-24 based on limited data. Finally, we provided a method to determine the toxicity threshold of linezolid.
Collapse
Affiliation(s)
- Ru Liao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lihong Chen
- Department of International Medical Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaoliang Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Houli Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Taotao Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Haiyan Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
7
|
Tang BH, Zhang XF, Fu SM, Yao BF, Zhang W, Wu YE, Zheng Y, Zhou Y, van den Anker J, Huang HR, Hao GX, Zhao W. Machine Learning Approach in Dosage Individualization of Isoniazid for Tuberculosis. Clin Pharmacokinet 2024; 63:1055-1063. [PMID: 38990504 DOI: 10.1007/s40262-024-01400-4] [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] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Isoniazid is a first-line antituberculosis agent with high variability, which would profit from individualized dosing. Concentrations of isoniazid at 2 h (C2h), as an indicator of safety and efficacy, are important for optimizing therapy. OBJECTIVE The objective of this study was to establish machine learning (ML) models to predict the C2h, that can be used for establishing an individualized dosing regimen in clinical practice. METHODS Published population pharmacokinetic (PopPK) models for adults were searched based on PubMed and ultimately four reliable models were selected for simulating individual C2h datasets under different conditions (demographics, genotype, ethnicity, etc.). Machine learning models were trained on simulated C2h obtained from the four PopPK models. Five different algorithms were used for ML model building to predict C2h. Real-world data were used for predictive performance evaluations. Virtual trials were used to compare ML-optimized doses with PopPK model-optimized doses. RESULTS Categorical boosting (CatBoost) exhibited the highest prediction ability. Target C2h can be predicted using the ML model combined with the dosing regimen and three covariates (N-acetyltransferase 2 [NAT2] genotypes, weight and race [Asians and Africans]). Real-world data validation results showed that the ML model can achieve an overall prediction accuracy of 93.4%. Using the final ML model, the mean absolute prediction error value decreased by 45.7% relative to the average of PopPK models. Using the ML-optimized dosing regimen, the probability of target attainment increased by 43.7% relative to the PopPK model-optimized dosing regimens. CONCLUSION Machine learning models were developed with great predictive performance, which can be used to determine the individualized initial dose of isoniazid in adult patients.
Collapse
Affiliation(s)
- Bo-Hao Tang
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin-Fang Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shu-Meng Fu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
- Departments of Pediatrics, Pharmacology and Physiology, Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Hai-Rong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China.
| |
Collapse
|
8
|
Wang YP, Lu XL, Shao K, Shi HQ, Zhou PJ, Chen B. Improving prediction of tacrolimus concentration using a combination of population pharmacokinetic modeling and machine learning in chinese renal transplant recipients. Front Pharmacol 2024; 15:1389271. [PMID: 38783953 PMCID: PMC11111944 DOI: 10.3389/fphar.2024.1389271] [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: 02/21/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Aims The population pharmacokinetic (PPK) model-based machine learning (ML) approach offers a novel perspective on individual concentration prediction. This study aimed to establish a PPK-based ML model for predicting tacrolimus (TAC) concentrations in Chinese renal transplant recipients. Methods Conventional TAC monitoring data from 127 Chinese renal transplant patients were divided into training (80%) and testing (20%) datasets. A PPK model was developed using the training group data. ML models were then established based on individual pharmacokinetic data derived from the PPK basic model. The prediction performances of the PPK-based ML model and Bayesian forecasting approach were compared using data from the test group. Results The final PPK model, incorporating hematocrit and CYP3A5 genotypes as covariates, was successfully established. Individual predictions of TAC using the PPK basic model, postoperative date, CYP3A5 genotype, and hematocrit showed improved rankings in ML model construction. XGBoost, based on the TAC PPK, exhibited the best prediction performance. Conclusion The PPK-based machine learning approach emerges as a superior option for predicting TAC concentrations in Chinese renal transplant recipients.
Collapse
Affiliation(s)
- Yu-Ping Wang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiao-Ling Lu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Kun Shao
- Center for Organ Transplantation, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Hao-Qiang Shi
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Pei-Jun Zhou
- Center for Organ Transplantation, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Bing Chen
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Codde C, Rivals F, Destere A, Fromage Y, Labriffe M, Marquet P, Benoist C, Ponthier L, Faucher JF, Woillard JB. A machine learning approach to predict daptomycin exposure from two concentrations based on Monte Carlo simulations. Antimicrob Agents Chemother 2024; 68:e0141523. [PMID: 38501807 PMCID: PMC11064575 DOI: 10.1128/aac.01415-23] [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: 10/30/2023] [Accepted: 02/23/2024] [Indexed: 03/20/2024] Open
Abstract
Daptomycin is a concentration-dependent lipopeptide antibiotic for which exposure/effect relationships have been shown. Machine learning (ML) algorithms, developed to predict the individual exposure to drugs, have shown very good performances in comparison to maximum a posteriori Bayesian estimation (MAP-BE). The aim of this work was to predict the area under the blood concentration curve (AUC) of daptomycin from two samples and a few covariates using XGBoost ML algorithm trained on Monte Carlo simulations. Five thousand one hundred fifty patients were simulated from two literature population pharmacokinetics models. Data from the first model were split into a training set (75%) and a testing set (25%). Four ML algorithms were built to learn AUC based on daptomycin blood concentration samples at pre-dose and 1 h post-dose. The XGBoost model (best ML algorithm) with the lowest root mean square error (RMSE) in a 10-fold cross-validation experiment was evaluated in both the test set and the simulations from the second population pharmacokinetic model (validation). The ML model based on the two concentrations, the differences between these concentrations, and five other covariates (sex, weight, daptomycin dose, creatinine clearance, and body temperature) yielded very good AUC estimation in the test (relative bias/RMSE = 0.43/7.69%) and validation sets (relative bias/RMSE = 4.61/6.63%). The XGBoost ML model developed allowed accurate estimation of daptomycin AUC using C0, C1h, and a few covariates and could be used for exposure estimation and dose adjustment. This ML approach can facilitate the conduct of future therapeutic drug monitoring (TDM) studies.
Collapse
Affiliation(s)
- Cyrielle Codde
- Service de Maladies Infectieuses et Tropicales, CHU Dupuytren, Limoges, France
| | - Florence Rivals
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
| | | | - Yeleen Fromage
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
| | - Marc Labriffe
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Toxicology, Limoges, France
| | - Pierre Marquet
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Toxicology, Limoges, France
| | - Clément Benoist
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Toxicology, Limoges, France
| | - Laure Ponthier
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Toxicology, Limoges, France
| | | | - Jean-Baptiste Woillard
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
- Inserm, Univ. Limoges, CHU Limoges, Pharmacology & Toxicology, Limoges, France
| |
Collapse
|
10
|
Li QY, Tang BH, Wu YE, Yao BF, Zhang W, Zheng Y, Zhou Y, van den Anker J, Hao GX, Zhao W. Machine Learning: A New Approach for Dose Individualization. Clin Pharmacol Ther 2024; 115:727-744. [PMID: 37713106 DOI: 10.1002/cpt.3049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023]
Abstract
The application of machine learning (ML) has shown promising results in precision medicine due to its exceptional performance in dealing with complex multidimensional data. However, using ML for individualized dosing of medicines is still in its early stage, meriting further exploration. A systematic review of study designs and modeling details of using ML for individualized dosing of different drugs was performed. We have summarized the status of the study populations, predictive targets, and data sources for ML modeling, the selection of ML algorithms and features, and the evaluation and validation of their predictive performance. We also used the Prediction model Risk of Bias Assessment Tool (PROBAST) to assess the risk of bias of included studies. Currently, ML can be used for both a priori and a posteriori dose selection and optimization, and it can also assist the implementation of therapeutic drug monitoring. However, studies are mainly focused on drugs with narrow therapeutic windows, predominantly immunosuppressants (N = 23, 35.9%) and anti-infectives (N = 21, 32.8%), and there is currently only very limited attention for special populations, such as children (N = 22, 34.4%). Most studies showed poor methodological quality and a high risk of bias. The lack of external validation and clinical utility evaluation currently limits the further clinical implementation of ML for dose individualization. We therefore have proposed several ways to improve the clinical relevance of the studies and facilitate the translation of ML models into clinical practice.
Collapse
Affiliation(s)
- Qiu-Yue Li
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
- Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| |
Collapse
|
11
|
Ponthier L, Autmizguine J, Franck B, Åsberg A, Ovetchkine P, Destere A, Marquet P, Labriffe M, Woillard JB. Optimization of Ganciclovir and Valganciclovir Starting Dose in Children by Machine Learning. Clin Pharmacokinet 2024:10.1007/s40262-024-01362-7. [PMID: 38492206 DOI: 10.1007/s40262-024-01362-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Ganciclovir (GCV) and valganciclovir (VGCV) show large interindividual pharmacokinetic variability, particularly in children. The objectives of this study were (1) to develop machine learning (ML) algorithms trained on simulated pharmacokinetics profiles obtained by Monte Carlo simulations to estimate the best ganciclovir or valganciclovir starting dose in children and (2) to compare its performances on real-world profiles to previously published equation derived from literature population pharmacokinetic (POPPK) models achieving about 20% of profiles within the target. MATERIALS AND METHODS The pharmacokinetic parameters of four literature POPPK models in addition to the World Health Organization (WHO) growth curve for children were used in the mrgsolve R package to simulate 10,800 pharmacokinetic profiles. ML algorithms were developed and benchmarked to predict the probability to reach the steady-state, area-under-the-curve target (AUC0-24 within 40-60 mg × h/L) based on demographic characteristics only. The best ML algorithm was then used to calculate the starting dose maximizing the target attainment. Performances were evaluated for ML and literature formula in a test set and in an external set of 32 and 31 actual patients (GCV and VGCV, respectively). RESULTS A combination of Xgboost, neural network, and random forest algorithms yielded the best performances and highest target attainment in the test set (36.8% for GCV and 35.3% for the VGCV). In actual patients, the best GCV ML starting dose yielded the highest target attainment rate (25.8%) and performed equally for VGCV with the Franck model formula (35.3% for both). CONCLUSION The ML algorithms exhibit good performances in comparison with previously validated models and should be evaluated prospectively.
Collapse
Affiliation(s)
- Laure Ponthier
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, 87000, Limoges, France
- Department of Pediatrics, University Hospital of Limoges, Limoges, France
| | - Julie Autmizguine
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Benedicte Franck
- Department of Clinical and Biological Pharmacology and Pharmacovigilance, Clinical Investigation Center, CIC-P 1414, Rennes, France
- University of Rennes, Centre Hospitalier Universitaire Rennes, École des Hautes Études en Santé Publique, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR S 1085, Rennes, France
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Section of Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Philippe Ovetchkine
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Alexandre Destere
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Nice, Nice, France
| | - Pierre Marquet
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Marc Labriffe
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Jean-Baptiste Woillard
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, 87000, Limoges, France.
- Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France.
| |
Collapse
|
12
|
Stankevičiūtė K, Woillard JB, Peck RW, Marquet P, van der Schaar M. Bridging the Worlds of Pharmacometrics and Machine Learning. Clin Pharmacokinet 2023; 62:1551-1565. [PMID: 37803104 DOI: 10.1007/s40262-023-01310-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/08/2023]
Abstract
Precision medicine requires individualized modeling of disease and drug dynamics, with machine learning-based computational techniques gaining increasing popularity. The complexity of either field, however, makes current pharmacological problems opaque to machine learning practitioners, and state-of-the-art machine learning methods inaccessible to pharmacometricians. To help bridge the two worlds, we provide an introduction to current problems and techniques in pharmacometrics that ranges from pharmacokinetic and pharmacodynamic modeling to pharmacometric simulations, model-informed precision dosing, and systems pharmacology, and review some of the machine learning approaches to address them. We hope this would facilitate collaboration between experts, with complementary strengths of principled pharmacometric modeling and flexibility of machine learning leading to synergistic effects in pharmacological applications.
Collapse
Affiliation(s)
- Kamilė Stankevičiūtė
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Avenue, Cambridge, CB3 0FD, UK
| | - Jean-Baptiste Woillard
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France.
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France.
| | - Richard W Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Pharma Research and Development, Roche Innovation Center, Basel, Switzerland
| | - Pierre Marquet
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France
| | - Mihaela van der Schaar
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| |
Collapse
|
13
|
Vora LK, Gholap AD, Jetha K, Thakur RRS, Solanki HK, Chavda VP. Artificial Intelligence in Pharmaceutical Technology and Drug Delivery Design. Pharmaceutics 2023; 15:1916. [PMID: 37514102 PMCID: PMC10385763 DOI: 10.3390/pharmaceutics15071916] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Artificial intelligence (AI) has emerged as a powerful tool that harnesses anthropomorphic knowledge and provides expedited solutions to complex challenges. Remarkable advancements in AI technology and machine learning present a transformative opportunity in the drug discovery, formulation, and testing of pharmaceutical dosage forms. By utilizing AI algorithms that analyze extensive biological data, including genomics and proteomics, researchers can identify disease-associated targets and predict their interactions with potential drug candidates. This enables a more efficient and targeted approach to drug discovery, thereby increasing the likelihood of successful drug approvals. Furthermore, AI can contribute to reducing development costs by optimizing research and development processes. Machine learning algorithms assist in experimental design and can predict the pharmacokinetics and toxicity of drug candidates. This capability enables the prioritization and optimization of lead compounds, reducing the need for extensive and costly animal testing. Personalized medicine approaches can be facilitated through AI algorithms that analyze real-world patient data, leading to more effective treatment outcomes and improved patient adherence. This comprehensive review explores the wide-ranging applications of AI in drug discovery, drug delivery dosage form designs, process optimization, testing, and pharmacokinetics/pharmacodynamics (PK/PD) studies. This review provides an overview of various AI-based approaches utilized in pharmaceutical technology, highlighting their benefits and drawbacks. Nevertheless, the continued investment in and exploration of AI in the pharmaceutical industry offer exciting prospects for enhancing drug development processes and patient care.
Collapse
Affiliation(s)
- Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Keshava Jetha
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
- Ph.D. Section, Gujarat Technological University, Ahmedabad 382424, Gujarat, India
| | | | - Hetvi K Solanki
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
14
|
Wang W, Battini V, Carnovale C, Noordam R, van Dijk KW, Kragholm KH, van Heemst D, Soeorg H, Sessa M. A novel approach for pharmacological substantiation of safety signals using plasma concentrations of medication and administrative/healthcare databases: a case study using Danish registries for an FDA warning on lamotrigine. Pharmacol Res 2023:106811. [PMID: 37268178 DOI: 10.1016/j.phrs.2023.106811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/04/2023]
Abstract
PHARMACOM-EPI is a novel framework to predict plasma concentrations of drugs at the time of occurrence of clinical outcomes. In early 2021, the U.S. Food and Drug Administration (FDA) issued a warning on the antiseizure drug lamotrigine claiming that it has the potential to increase the risk of arrhythmias and related sudden cardiac death due to a pharmacological sodium channel-blocking effect. We hypothesized that the risk of arrhythmias and related death is due to toxicity. We used the PHARMACOM-EPI framework to investigate the relationship between lamotrigine's plasma concentrations and the risk of death in older patients using real-world data. Danish nationwide administrative and healthcare registers were used as data sources and individuals aged 65 years or older during the period 1996 - 2018 were included in the study. According to the PHARMACOM-EPI framework, plasma concentrations of lamotrigine were predicted at the time of death and patients were categorized into non-toxic and toxic groups based on the therapeutic range of lamotrigine (3-15mg/L). Over 1 year of treatment, the incidence rate ratio (IRR) of all-cause mortality was calculated between the propensities score matched toxic and non-toxic groups. In total, 7286 individuals were diagnosed with epilepsy and were exposed to lamotrigine, 432 of which had at least one plasma concentration measurement The pharmacometric model by Chavez et al. was used to predict lamotrigine's plasma concentrations considering the lowest absolute percentage error among identified models (14.25%, 95% CI: 11.68-16.23). The majority of lamotrigine associated deaths were cardiovascular-related and occurred among individuals with plasma concentrations in the toxic range. The IRR of mortality between the toxic group and non-toxic group was 3.37 [95% CI: 1.44-8.32] and the cumulative incidence of all-cause mortality exponentially increased in the toxic range. Application of our novel framework PHARMACOM-EPI provided strong evidence to support our hypothesis that the increased risk of all-cause and cardiovascular death was associated with a toxic plasma concentration level of lamotrigine among older lamotrigine users.
Collapse
Affiliation(s)
- Wenyi Wang
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands; Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Vera Battini
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy; Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Italy
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics; Leiden University Medical Center, Leiden, Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands; Department of Internal Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, Netherlands; Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | | | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics; Leiden University Medical Center, Leiden, Netherlands
| | - Hiie Soeorg
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Estonia.
| | - Maurizio Sessa
- Department of Drug Design and Pharmacology, University of Copenhagen, Denmark.
| |
Collapse
|
15
|
Li ZR, Shen CH, Li RD, Wang B, Li J, Niu WJ, Zhang LJ, Zhong MK, Wang ZX, Qiu XY. Individual dose recommendations for drug interaction between tacrolimus and voriconazole in adult liver transplant recipients: A semiphysiologically based population pharmacokinetic modeling approach. Eur J Pharm Sci 2023; 184:106405. [PMID: 36775255 DOI: 10.1016/j.ejps.2023.106405] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/18/2022] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
The magnitude of drug-drug interaction between tacrolimus and voriconazole is highly variable, and individually tailoring the tacrolimus dose when concomitantly administered with voriconazole remains difficult. This study aimed to develop a semiphysiologically based population pharmacokinetic (semi-PBPK) model and a web-based dashboard to identify the dynamic inhibition of tacrolimus metabolism caused by voriconazole and provide individual tacrolimus regimens for Chinese adult liver transplant recipients. A total of 264 tacrolimus concentrations and 146 voriconazole concentrations were prospectively collected from 32 transplant recipients. A semi-PBPK model with physiological compartments including the gut wall, portal vein, and liver was developed using the nonlinear mixed-effects modeling software NONMEM (version 7.4). A web-based dashboard was established in R software (version 3.6.1) to recommend the individual tacrolimus regimens when concomitantly administered with voriconazole. The reversible inhibition of tacrolimus metabolism caused by voriconazole was investigated in both the liver and the gut wall. Moreover, voriconazole could highly inhibit the CYP3A activity in the gut wall more than in the liver. BMI and postoperative days were identified as significant covariates on intrinsic intestinal and hepatic clearance of tacrolimus, respectively. Age and postoperative days were identified as significant covariates on the volume of distribution of voriconazole. The individual tacrolimus regimens when concomitantly administered with voriconazole could be recommended in the dashboard (https://tac-vor-ddi.shinyapps.io/shinyapp3/). In conclusion, the semi-PBPK model successfully described the dynamic inhibition process between tacrolimus and voriconazole, and the web-based dashboard could provide individual tacrolimus regimens when concomitantly administered with voriconazole.
Collapse
Affiliation(s)
- Zi-Ran Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Cong-Huan Shen
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai 200040, China; Institute of Organ Transplant, Fudan University, Shanghai 200040, China
| | - Rui-Dong Li
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai 200040, China; Institute of Organ Transplant, Fudan University, Shanghai 200040, China
| | - Bei Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Juan Li
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai 200040, China; Institute of Organ Transplant, Fudan University, Shanghai 200040, China
| | - Wan-Jie Niu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Li-Jun Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Zheng-Xin Wang
- Department of General Surgery and Liver Transplant Center, Huashan Hospital, Fudan University, Shanghai 200040, China; Institute of Organ Transplant, Fudan University, Shanghai 200040, China.
| | - Xiao-Yan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
16
|
Li ZR, Wang CY, Lin WW, Chen YT, Liu XQ, Jiao Z. Handling Delayed or Missed Dose of Antiseizure Medications: A Model-Informed Individual Remedial Dosing. Neurology 2023; 100:e921-e931. [PMID: 36450606 PMCID: PMC9990430 DOI: 10.1212/wnl.0000000000201604] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Delayed or missed antiseizure medications (ASMs) doses are common during long-term or lifelong antiepilepsy treatment. This study aims to explore optimal individualized remedial dosing regimens for delayed or missed doses of 11 commonly used ASMs. METHODS To explore remedial dosing regimens, Monte Carlo simulation was used based on previously identified and published population pharmacokinetic models. Six remedial strategies for delayed or missed doses were investigated. The deviation time outside the individual therapeutic range was used to evaluate each remedial regimen. The influences of patients' demographics, concomitant medication, and scheduled dosing intervals on remedial regimens were assessed. RxODE and Shiny in R were used to perform Monte Carlo simulation and recommend individual remedial regimens. RESULTS The recommended remedial regimens were highly correlated with delayed time, scheduled dosing interval, and half-life of the ASM. Moreover, the optimal remedial regimens for pediatric and adult patients were different. The renal function, along with concomitant medication that affects the clearance of the ASM, may also influence the remedial regimens. A web-based dashboard was developed to provide individualized remedial regimens for the delayed or missed dose, and a user-defined module with all parameters that could be defined flexibly by the user was also built. DISCUSSION Monte Carlo simulation based on population pharmacokinetic models may provide a rational approach to propose remedial regimens for delayed or missed doses of ASMs in pediatric and adult patients with epilepsy.
Collapse
Affiliation(s)
- Zi-Ran Li
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China.
| | - Chen-Yu Wang
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China
| | - Wei-Wei Lin
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China.
| | - Yue-Ting Chen
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China
| | - Xiao-Qin Liu
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China
| | - Zheng Jiao
- From the Department of Pharmacy (Z.L., C.W., Y.C., X.L., Z.J.), Shanghai Chest Hospital, Shanghai Jiao Tong University, China; Department of Pharmacy (Z.L., X.L.), Huashan Hospital, Fudan University, Shanghai, China; Department of Pharmacy (W.L.), The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; and School of Basic Medicine and Clinical Pharmacy (Y.C.), China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
17
|
Ruiz-Ramos J, Gras-Martín L, Ramírez P. Antimicrobial Pharmacokinetics and Pharmacodynamics in Critical Care: Adjusting the Dose in Extracorporeal Circulation and to Prevent the Genesis of Multiresistant Bacteria. Antibiotics (Basel) 2023; 12:antibiotics12030475. [PMID: 36978342 PMCID: PMC10044431 DOI: 10.3390/antibiotics12030475] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Critically ill patients suffering from severe infections are prone to pathophysiological pharmacokinetic changes that are frequently associated with inadequate antibiotic serum concentrations. Minimum inhibitory concentrations (MICs) of the causative pathogens tend to be higher in intensive care units. Both pharmacokinetic changes and high antibiotic resistance likely jeopardize the efficacy of treatment. The use of extracorporeal circulation devices to support hemodynamic, respiratory, or renal failure enables pharmacokinetic changes and makes it even more difficult to achieve an adequate antibiotic dose. Besides a clinical response, antibiotic pharmacokinetic optimization is important to reduce the selection of strains resistant to common antibiotics. In this review, we summarize the present knowledge regarding pharmacokinetic changes in critically ill patients and we discuss the effects of extra-corporeal devices on antibiotic treatment together with potential solutions.
Collapse
Affiliation(s)
- Jesus Ruiz-Ramos
- Pharmacy Department, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Laura Gras-Martín
- Pharmacy Department, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Paula Ramírez
- Intensive Care Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Correspondence:
| |
Collapse
|
18
|
Application of machine learning to predict tacrolimus exposure in liver and kidney transplant patients given the MeltDose formulation. Eur J Clin Pharmacol 2023; 79:311-319. [PMID: 36564549 DOI: 10.1007/s00228-022-03445-5] [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: 10/04/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022]
Abstract
PURPOSE Machine Learning (ML) algorithms represent an interesting alternative to maximum a posteriori Bayesian estimators (MAP-BE) for tacrolimus AUC estimation, but it is not known if training an ML model using a lower number of full pharmacokinetic (PK) profiles (= "true" reference AUC) provides better performances than using a larger dataset of less accurate AUC estimates. The objectives of this study were: to develop and benchmark ML algorithms trained using full PK profiles to estimate MeltDose®-tacrolimus individual AUCs using 2 or 3 blood concentrations; and to compare their performance to MAP-BE. METHODS Data from liver (n = 113) and kidney (n = 97) transplant recipients involved in MeltDose-tacrolimus PK studies were used for the training and evaluation of ML algorithms. "True" AUC0-24 h was calculated for each patient using the trapezoidal rule on the full PK profile. ML algorithms were trained to estimate tacrolimus true AUC using 2 or 3 blood concentrations. Performances were evaluated in 2 external sets of 16 (renal) and 48 (liver) transplant patients. RESULTS Best estimation performances were obtained with the MARS algorithm and the following limited sampling strategies (LSS): predose (0), 8, and 12 h post-dose (rMPE = - 1.28%, rRMSE = 7.57%), or 0 and 12 h (rMPE = - 1.9%, rRMSE = 10.06%). In the external dataset, the performances of the final ML algorithms based on two samples in kidney (rMPE = - 3.1%, rRMSE = 11.1%) or liver transplant recipients (rMPE = - 3.4%, rRMSE = 9.86%) were as good as or better than those of MAP-BEs based on three time points. CONCLUSION The MARS ML models developed using "true" MeltDose®-tacrolimus AUCs yielded accurate individual estimations using only two blood concentrations.
Collapse
|
19
|
Huang Q, Lin X, Wang Y, Chen X, Zheng W, Zhong X, Shang D, Huang M, Gao X, Deng H, Li J, Zeng F, Mo X. Tacrolimus pharmacokinetics in pediatric nephrotic syndrome: A combination of population pharmacokinetic modelling and machine learning approaches to improve individual prediction. Front Pharmacol 2022; 13:942129. [DOI: 10.3389/fphar.2022.942129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Background and Aim: Tacrolimus (TAC) is a first-line immunosuppressant for the treatment of refractory nephrotic syndrome (RNS), but the pharmacokinetics of TAC varies widely among individuals, and there is still no accurate model to predict the pharmacokinetics of TAC in RNS. Therefore, this study aimed to combine population pharmacokinetic (PPK) model and machine learning algorithms to develop a simple and accurate prediction model for TAC.Methods: 139 children with RNS from August 2013 to December 2018 were included, and blood samples of TAC trough and partial peak concentrations were collected. The blood concentration of TAC was determined by enzyme immunoassay; CYP3A5 was genotyped by polymerase chain reaction-restriction fragment length polymorphism method; MYH9, LAMB2, ACTN4 and other genotypes were determined by MALDI-TOF MS method; PPK model was established by nonlinear mixed-effects method. Based on this, six machine learning algorithms, including eXtreme Gradient Boosting (XGBoost), Random Forest (RF), Extra-Trees, Gradient Boosting Decision Tree (GBDT), Adaptive boosting (AdaBoost) and Lasso, were used to establish the machine learning model of TAC clearance.Results: A one-compartment model of first-order absorption and elimination adequately described the pharmacokinetics of TAC. Age, co-administration of Wuzhi capsules, CYP3A5 *3/*3 genotype and CTLA4 rs4553808 genotype were significantly affecting the clearance of TAC. Among the six machine learning models, the Lasso algorithm model performed the best (R2 = 0.42).Conclusion: For the first time, a clearance prediction model of TAC in pediatric patients with RNS was established using PPK combined with machine learning, by which the individual clearance of TAC can be predicted more accurately, and the initial dose of administration can be optimized to achieve the goal of individualized treatment.
Collapse
|
20
|
Qian L, Jiao Z, Zhong M. Effect of Meal Timings and Meal Content on the AUC 0-12h of Mycophenolic Acid: A Simulation Study. Clin Pharmacol Drug Dev 2022; 11:1331-1340. [PMID: 36045559 DOI: 10.1002/cpdd.1141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/20/2022] [Indexed: 01/27/2023]
Abstract
Meal timings and content related to gallbladder emptying in the enterohepatic circulation are important for explaining the high variability in mycophenolic acid exposure. The limited sampling strategy (LSS) was established to estimate the area under the plasma concentration-time curve from time 0 to 12 hours (AUC0-12h ) of mycophenolic acid in therapeutic drug monitoring. The aim of this study is to investigate the effect of meal timings and content on the AUC0-12h of mycophenolic acid and to assess the influence of meals on LSS. A mycophenolic acid pharmacokinetic model with a mechanism-based enterohepatic circulation process was employed to perform simulations under various assumed meal scenarios. The simulations were compared to evaluate the effect of meal timings and meal content on mycophenolic acid AUC0-12h . Monte Carlo simulations were performed using the meal parameter with the greatest impact on mycophenolic acid AUC0-12h as a variable. The corresponding LSS equations were established, and the predictive performance was assessed. Both the meal timings and meal content affected the mycophenolic acid AUC0-12h , and the postdose fasting period had the greatest impact. The predictive performance of the LSS is sensitive to the postdose fasting period. Therefore, meal timings may improve the estimation of mycophenolic acid AUC0-12h and the efficacy of therapeutic drug monitoring.
Collapse
Affiliation(s)
- Lixuan Qian
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
van der Lee M, Swen JJ. Artificial intelligence in pharmacology research and practice. Clin Transl Sci 2022; 16:31-36. [PMID: 36181380 PMCID: PMC9841296 DOI: 10.1111/cts.13431] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 02/04/2023] Open
Abstract
In recent years, the use of artificial intelligence (AI) in health care has risen steadily, including a wide range of applications in the field of pharmacology. AI is now used throughout the entire continuum of pharmacology research and clinical practice and from early drug discovery to real-world datamining. The types of AI models used range from unsupervised clustering of drugs or patients aimed at identifying potential drug compounds or suitable patient populations, to supervised machine learning approaches to improve therapeutic drug monitoring. Additionally, natural language processing is increasingly used to mine electronic health records to obtain real-world data. In this mini-review, we discuss the basics of AI followed by an outline of its application in pharmacology research and clinical practice.
Collapse
Affiliation(s)
- Maaike van der Lee
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jesse J. Swen
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
22
|
Zhu X, Zhang M, Wen Y, Shang D. Machine learning advances the integration of covariates in population pharmacokinetic models: Valproic acid as an example. Front Pharmacol 2022; 13:994665. [PMID: 36324679 PMCID: PMC9621318 DOI: 10.3389/fphar.2022.994665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Background and Aim: Many studies associated with the combination of machine learning (ML) and pharmacometrics have appeared in recent years. ML can be used as an initial step for fast screening of covariates in population pharmacokinetic (popPK) models. The present study aimed to integrate covariates derived from different popPK models using ML. Methods: Two published popPK models of valproic acid (VPA) in Chinese epileptic patients were used, where the population parameters were influenced by some covariates. Based on the covariates and a one-compartment model that describes the pharmacokinetics of VPA, a dataset was constructed using Monte Carlo simulation, to develop an XGBoost model to estimate the steady-state concentrations (Css) of VPA. We utilized SHapley Additive exPlanation (SHAP) values to interpret the prediction model, and calculated estimates of VPA exposure in four assumed scenarios involving different combinations of CYP2C19 genotypes and co-administered antiepileptic drugs. To develop an easy-to-use model in the clinic, we built a simplified model by using CYP2C19 genotypes and some noninvasive clinical parameters, and omitting several features that were infrequently measured or whose clinically available values were inaccurate, and verified it on our independent external dataset. Results: After data preprocessing, the finally generated combined dataset was divided into a derivation cohort and a validation cohort (8:2). The XGBoost model was developed in the derivation cohort and yielded excellent performance in the validation cohort with a mean absolute error of 2.4 mg/L, root-mean-squared error of 3.3 mg/L, mean relative error of 0%, and percentages within ±20% of actual values of 98.85%. The SHAP analysis revealed that daily dose, time, CYP2C19*2 and/or *3 variants, albumin, body weight, single dose, and CYP2C19*1*1 genotype were the top seven confounding factors influencing the Css of VPA. Under the simulated dosage regimen of 500 mg/bid, the VPA exposure in patients who had CYP2C19*2 and/or *3 variants and no carbamazepine, phenytoin, or phenobarbital treatment, was approximately 1.74-fold compared to those with CYP2C19*1/*1 genotype and co-administered carbamazepine + phenytoin + phenobarbital. The feasibility of the simplified model was fully illustrated by its performance in our external dataset. Conclusion: This study highlighted the bridging role of ML in big data and pharmacometrics, by integrating covariates derived from different popPK models.
Collapse
Affiliation(s)
- Xiuqing Zhu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Ming Zhang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- *Correspondence: Yuguan Wen, ; Dewei Shang,
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- *Correspondence: Yuguan Wen, ; Dewei Shang,
| |
Collapse
|
23
|
Zhu X, Hu J, Xiao T, Huang S, Wen Y, Shang D. An interpretable stacking ensemble learning framework based on multi-dimensional data for real-time prediction of drug concentration: The example of olanzapine. Front Pharmacol 2022; 13:975855. [PMID: 36238557 PMCID: PMC9552071 DOI: 10.3389/fphar.2022.975855] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Aim: Therapeutic drug monitoring (TDM) has evolved over the years as an important tool for personalized medicine. Nevertheless, some limitations are associated with traditional TDM. Emerging data-driven model forecasting [e.g., through machine learning (ML)-based approaches] has been used for individualized therapy. This study proposes an interpretable stacking-based ML framework to predict concentrations in real time after olanzapine (OLZ) treatment. Methods: The TDM-OLZ dataset, consisting of 2,142 OLZ measurements and 472 features, was formed by collecting electronic health records during the TDM of 927 patients who had received OLZ treatment. We compared the performance of ML algorithms by using 10-fold cross-validation and the mean absolute error (MAE). The optimal subset of features was analyzed by a random forest-based sequential forward feature selection method in the context of the top five heterogeneous regressors as base models to develop a stacked ensemble regressor, which was then optimized via the grid search method. Its predictions were explained by using local interpretable model-agnostic explanations (LIME) and partial dependence plots (PDPs). Results: A state-of-the-art stacking ensemble learning framework that integrates optimized extra trees, XGBoost, random forest, bagging, and gradient-boosting regressors was developed for nine selected features [i.e., daily dose (OLZ), gender_male, age, valproic acid_yes, ALT, K, BW, MONO#, and time of blood sampling after first administration]. It outperformed other base regressors that were considered, with an MAE of 0.064, R-square value of 0.5355, mean squared error of 0.0089, mean relative error of 13%, and ideal rate (the percentages of predicted TDM within ± 30% of actual TDM) of 63.40%. Predictions at the individual level were illustrated by LIME plots, whereas the global interpretation of associations between features and outcomes was illustrated by PDPs. Conclusion: This study highlights the feasibility of the real-time estimation of drug concentrations by using stacking-based ML strategies without losing interpretability, thus facilitating model-informed precision dosing.
Collapse
Affiliation(s)
- Xiuqing Zhu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Jinqing Hu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Tao Xiao
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Research, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shanqing Huang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- *Correspondence: Yuguan Wen, ; Dewei Shang,
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- *Correspondence: Yuguan Wen, ; Dewei Shang,
| |
Collapse
|
24
|
Ponthier L, Ensuque P, Destere A, Marquet P, Labriffe M, Jacqz-Aigrain E, Woillard JB. Optimization of Vancomycin Initial Dose in Term and Preterm Neonates by Machine Learning. Pharm Res 2022; 39:2497-2506. [PMID: 35918452 DOI: 10.1007/s11095-022-03351-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/23/2022] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Vancomycin is one of the antibiotics most used in neonates. Continuous infusion has many advantages over intermittent infusions, but no consensus has been achieved regarding the optimal initial dose. The objectives of this study were: to develop a Machine learning (ML) algorithm based on pharmacokinetic profiles obtained by Monte Carlo simulations using a population pharmacokinetic model (POPPK) from the literature, in order to derive the best vancomycin initial dose in preterm and term neonates, and to compare ML performances with those of an literature equation (LE) derived from a POPPK previously published. MATERIALS AND METHODS The parameters of a previously published POPPK model of vancomycin in children and neonates were used in the mrgsolve R package to simulate 1900 PK profiles. ML algorithms were developed from these simulations using Xgboost, GLMNET and MARS in parallel, benchmarked and used to calculate the ML first dose. Performances were evaluated in a second simulation set and in an external set of 82 real patients and compared to those of a LE. RESULTS The Xgboost algorithm yielded numerically best performances and target attainment rates: 46.9% in the second simulation set of 400-600 AUC/MIC ratio vs. 41.4% for the LE model (p = 0.0018); and 35.3% vs. 28% in real patients (p = 0.401), respectively). The Xgboost model resulted in less AUC/MIC > 600, thus decreasing the risk of nephrotoxicity. CONCLUSION The Xgboost algorithm developed to estimate the initial dose of vancomycin in term or preterm infants has better performances than a previous validated LE and should be evaluated prospectively.
Collapse
Affiliation(s)
- Laure Ponthier
- Pharmacology & Transplantation, University Limoges, INSERM U1248 P&T, 2 rue du Pr Descottes, F-87000, Limoges, France.,Department of Pediatrics, University Hospital of Limoges, Limoges, France
| | - Pauline Ensuque
- Department of Pediatrics, University Hospital of Limoges, Limoges, France
| | - Alexandre Destere
- Pharmacology & Transplantation, University Limoges, INSERM U1248 P&T, 2 rue du Pr Descottes, F-87000, Limoges, France.,Department of Pharmacology and Toxicology, University Hospital of Nice, Nice, France
| | - Pierre Marquet
- Pharmacology & Transplantation, University Limoges, INSERM U1248 P&T, 2 rue du Pr Descottes, F-87000, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Marc Labriffe
- Pharmacology & Transplantation, University Limoges, INSERM U1248 P&T, 2 rue du Pr Descottes, F-87000, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France
| | - Evelyne Jacqz-Aigrain
- Pediatric Pharmacology, Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, Saint-Louis, France
| | - Jean-Baptiste Woillard
- Pharmacology & Transplantation, University Limoges, INSERM U1248 P&T, 2 rue du Pr Descottes, F-87000, Limoges, France. .,Department of Pharmacology, Toxicology and Pharmacovigilance, University Hospital of Limoges, Limoges, France.
| |
Collapse
|
25
|
Koppe L, Soulage CO. Protein-bound uremic toxins: putative modulators of calcineurin inhibitors exposure. Nephrol Dial Transplant 2022; 37:2044-2047. [PMID: 35916444 DOI: 10.1093/ndt/gfac229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Laetitia Koppe
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France.,Univ. Lyon, CarMeN lab, INSA-Lyon, INSERM U1060, INRA, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Christophe O Soulage
- Univ. Lyon, CarMeN lab, INSA-Lyon, INSERM U1060, INRA, Université Claude Bernard Lyon 1, Villeurbanne, France
| |
Collapse
|
26
|
Labriffe M, Woillard J, Debord J, Marquet P. Machine learning algorithms to estimate everolimus exposure trained on simulated and patient pharmacokinetic profiles. CPT Pharmacometrics Syst Pharmacol 2022; 11:1018-1028. [PMID: 35599364 PMCID: PMC9381914 DOI: 10.1002/psp4.12810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022] Open
Abstract
Everolimus is an immunosuppressant with a small therapeutic index and large between‐patient variability. The area under the concentration versus time curve (AUC) is the best marker of exposure but measuring it requires collecting many blood samples. The objective of this study was to train machine learning (ML) algorithms using pharmacokinetic (PK) profiles from kidney transplant recipients, simulated profiles, or both types, and compare their performance for everolimus AUC0‐12h estimation using a limited number of predictors, as compared to an independent set of full PK profiles from patients, as well as to the corresponding maximum a posteriori Bayesian estimates (MAP‐BE). XGBoost was first trained on 508 patient interdose AUCs estimated using MAP‐BE, and then on 500–10,000 rich interdose PK profiles simulated using previously published population PK parameters. The predictors used were: predose, ~1 h, and ~2 h whole blood concentrations, differences between these concentrations, relative deviations from theoretical sampling times, morning dose, patient age, and time elapsed since transplantation. The best results were obtained with XGBoost trained on 5016 simulated profiles. AUC estimation achieved in an external dataset of 114 full‐PK profiles was excellent (root mean squared error [RMSE] = 10.8 μg*h/L) and slightly better than MAP‐BE (RMSE = 11.9 μg*h/L). Using more profiles (n = 10,035) did not improve the ML algorithm performance. The contribution of mixing patient and simulated profiles was significant only when they were in balanced numbers, with ~500 for each (RMSE = 12.5 μg*h/L), compared with patient data alone (RMSE = 18.0 μg*h/L).
Collapse
Affiliation(s)
- Marc Labriffe
- Pharmacology & Transplantation, INSERM U1248 Université de Limoges Limoges France
- Department of Pharmacology, Toxicology and Pharmacovigilance CHU de Limoges Limoges France
| | - Jean‐Baptiste Woillard
- Pharmacology & Transplantation, INSERM U1248 Université de Limoges Limoges France
- Department of Pharmacology, Toxicology and Pharmacovigilance CHU de Limoges Limoges France
| | - Jean Debord
- Pharmacology & Transplantation, INSERM U1248 Université de Limoges Limoges France
- Department of Pharmacology, Toxicology and Pharmacovigilance CHU de Limoges Limoges France
| | - Pierre Marquet
- Pharmacology & Transplantation, INSERM U1248 Université de Limoges Limoges France
- Department of Pharmacology, Toxicology and Pharmacovigilance CHU de Limoges Limoges France
| |
Collapse
|
27
|
Keutzer L, You H, Farnoud A, Nyberg J, Wicha SG, Maher-Edwards G, Vlasakakis G, Moghaddam GK, Svensson EM, Menden MP, Simonsson USH. Machine Learning and Pharmacometrics for Prediction of Pharmacokinetic Data: Differences, Similarities and Challenges Illustrated with Rifampicin. Pharmaceutics 2022; 14:pharmaceutics14081530. [PMID: 35893785 PMCID: PMC9330804 DOI: 10.3390/pharmaceutics14081530] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Pharmacometrics (PM) and machine learning (ML) are both valuable for drug development to characterize pharmacokinetics (PK) and pharmacodynamics (PD). Pharmacokinetic/pharmacodynamic (PKPD) analysis using PM provides mechanistic insight into biological processes but is time- and labor-intensive. In contrast, ML models are much quicker trained, but offer less mechanistic insights. The opportunity of using ML predictions of drug PK as input for a PKPD model could strongly accelerate analysis efforts. Here exemplified by rifampicin, a widely used antibiotic, we explore the ability of different ML algorithms to predict drug PK. Based on simulated data, we trained linear regressions (LASSO), Gradient Boosting Machines, XGBoost and Random Forest to predict the plasma concentration-time series and rifampicin area under the concentration-versus-time curve from 0–24 h (AUC0–24h) after repeated dosing. XGBoost performed best for prediction of the entire PK series (R2: 0.84, root mean square error (RMSE): 6.9 mg/L, mean absolute error (MAE): 4.0 mg/L) for the scenario with the largest data size. For AUC0–24h prediction, LASSO showed the highest performance (R2: 0.97, RMSE: 29.1 h·mg/L, MAE: 18.8 h·mg/L). Increasing the number of plasma concentrations per patient (0, 2 or 6 concentrations per occasion) improved model performance. For example, for AUC0–24h prediction using LASSO, the R2 was 0.41, 0.69 and 0.97 when using predictors only (no plasma concentrations), 2 or 6 plasma concentrations per occasion as input, respectively. Run times for the ML models ranged from 1.0 s to 8 min, while the run time for the PM model was more than 3 h. Furthermore, building a PM model is more time- and labor-intensive compared with ML. ML predictions of drug PK could thus be used as input into a PKPD model, enabling time-efficient analysis.
Collapse
Affiliation(s)
- Lina Keutzer
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
| | - Huifang You
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
| | - Ali Farnoud
- Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany; (A.F.); (M.P.M.)
| | - Joakim Nyberg
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (J.N.); (E.M.S.)
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Gareth Maher-Edwards
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
| | - Georgios Vlasakakis
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
| | - Gita Khalili Moghaddam
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Elin M. Svensson
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (J.N.); (E.M.S.)
- Department of Pharmacy, Radboud Institute of Health Sciences, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
| | - Michael P. Menden
- Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany; (A.F.); (M.P.M.)
- Department of Biology, Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
| | - Ulrika S. H. Simonsson
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
- Correspondence:
| | | |
Collapse
|
28
|
A Machine Learning Approach to Predict Interdose Vancomycin Exposure. Pharm Res 2022; 39:721-731. [DOI: 10.1007/s11095-022-03252-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
|
29
|
Therapeutic drug monitoring of immunosuppressive drugs in hepatology and gastroenterology. Best Pract Res Clin Gastroenterol 2021; 54-55:101756. [PMID: 34874840 DOI: 10.1016/j.bpg.2021.101756] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 01/31/2023]
Abstract
Immunosuppressive drugs have been key to the success of liver transplantation and are essential components of the treatment of inflammatory bowel disease (IBD) and autoimmune hepatitis (AIH). For many but not all immunosuppressants, therapeutic drug monitoring (TDM) is recommended to guide therapy. In this article, the rationale and evidence for TDM of tacrolimus, mycophenolic acid, the mammalian target of rapamycin inhibitors, and azathioprine in liver transplantation, IBD, and AIH is reviewed. New developments, including algorithm-based/computer-assisted immunosuppressant dosing, measurement of immunosuppressants in alternative matrices for whole blood, and pharmacodynamic monitoring of these agents is discussed. It is expected that these novel techniques will be incorporate into the standard TDM in the next few years.
Collapse
|