1
|
Codde C, Faucher JF, Woillard JB. Use of Artificial Intelligence in Current Fight Against Antimicrobial Resistance. Microb Drug Resist 2025. [PMID: 40354275 DOI: 10.1089/mdr.2024.0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a significant global health threat, with projections indicating it could surpass cancer in mortality rates by 2050 if left unaddressed. Optimizing antimicrobial dosing is critical to mitigate resistance and improve clinical outcomes. Traditional approaches, including population pharmacokinetics (PK) models and Bayesian estimation, are limited by mechanistic hypothesis requirements and complexity. Artificial intelligence (AI) and machine learning (ML) offer transformative solutions by leveraging large datasets to predict drug exposure accurately, refine sampling strategies, and enable real-time dose adjustments through therapeutic drug monitoring. This review highlights the role of ML models, in managing PK and pharmacodynamic variability across diverse patient populations. AI models often equal or outperform traditional methods in achieving therapeutic targets while minimizing toxicity, as demonstrated in some case studies involving ganciclovir, vancomycin, and daptomycin. Despite challenges such as data quality, interpretability, and integration with clinical workflows, AI's dynamic adaptability and precision underscore its potential. Future directions emphasize integrating multi-omics data, developing bedside decision-support tools, and expanding AI applications to broader drug categories and populations. Continued research and clinical validation are essential to harness AI's full potential in advancing precision medicine and combating AMR effectively.
Collapse
Affiliation(s)
- Cyrielle Codde
- Service de Maladies Infectieuses et Tropicales, CHU Dupuytren, Limoges, France
- Inserm, Pharmacology & Transplantation, Univ. Limoges, Limoges, France
| | | | - Jean-Baptiste Woillard
- Inserm, Pharmacology & Transplantation, Univ. Limoges, Limoges, France
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, CHU Dupuytren, Limoges, France
| |
Collapse
|
2
|
Huang S, Xu Q, Yang G, Ding J, Pei Q. Machine Learning for Prediction of Drug Concentrations: Application and Challenges. Clin Pharmacol Ther 2025; 117:1236-1247. [PMID: 39901656 DOI: 10.1002/cpt.3577] [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: 06/12/2024] [Accepted: 01/13/2025] [Indexed: 02/05/2025]
Abstract
With the advancements in algorithms and increased accessibility of multi-source data, machine learning in pharmacokinetics is gaining interest. This review summarizes studies on machine learning-based pharmacokinetics analysis up to September 2024, identified from the PubMed and IEEE Xplore databases. The main focus of this review is on the use of machine learning in predicting drug concentration. This review provides a comprehensive summary of the advances in the machine learning algorithms for pharmacokinetics analysis. Specifically, we describe the common practices in data preprocessing, the application scenarios of various algorithms, and the critical challenges that require attention. Most machine learning models show comparable performance to those of population pharmacokinetics models. Tree-based algorithms and neural networks have the most applications. Furthermore, the use of ensemble modeling techniques can improve the accuracy of these models' predictions of drug concentrations, especially the ensembles of machine learning and pharmacometrics.
Collapse
Affiliation(s)
- Shuqi Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qihan Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Guoping Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Junjie Ding
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Qi Pei
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Sayadi H, Fromage Y, Labriffe M, Billat PA, Codde C, Arraki Zava S, Marquet P, Woillard JB. Estimation of Ganciclovir Exposure in Adults Transplant Patients by Machine Learning. AAPS J 2025; 27:53. [PMID: 40021573 DOI: 10.1208/s12248-025-01034-9] [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: 08/13/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025] Open
Abstract
INTRODUCTION Valganciclovir, a prodrug of ganciclovir (GCV), is used to prevent cytomegalovirus infection after transplantation, with doses adjusted based on creatinine clearance (CrCL) to target GCV AUC0-24 h of 40-60 mg*h/L. This sometimes leads to overexposure or underexposure. This study aimed to train, test and validate machine learning (ML) algorithms for accurate GCV AUC0-24 h estimation in solid organ transplantation. METHODS We simulated patients for different dosing regimen (900 mg/24 h, 450 mg/24 h, 450 mg/48 h, 450 mg/72 h) using two literature population pharmacokinetic models, allocating 75% for training and 25% for testing. Simulations from two other literature models and real patients provided validation datasets. Three independent sets of ML algorithms were created for each regimen, incorporating CrCL and 2 or 3 concentrations. We evaluated their performance on testing and validation datasets and compared them with MAP-BE. RESULTS XGBoost using 3 concentrations generated the most accurate predictions. In testing dataset, they exhibited a relative bias of -0.02 to 1.5% and a relative RMSE of 2.6 to 8.5%. In the validation dataset, a relative bias of 1.5 to 5.8% and 8.9 to 16.5%, and a relative RMSE of 8.5 to 9.6% and 10.7% to 19.7% were observed depending on the model used. XGBoost algorithms outperformed or matched MAP-BE, showing enhanced generalization and robustness in their estimates. When applied to real patients' data, algorithms using 2 concentrations showed relative bias of 1.26% and relative RMSE of 12.68%. CONCLUSIONS XGBoost ML models accurately estimated GCV AUC0-24 h from limited samples and CrCL, providing a strategy for optimized therapeutic drug monitoring.
Collapse
Affiliation(s)
- Hamza Sayadi
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Yeleen Fromage
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Marc Labriffe
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
| | - Pierre-André Billat
- INERIS, Experimental Toxicology and Modeling Unit (TEAM), Parc ALATA BP2, Verneuil en Halatte, France
| | - Cyrielle Codde
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
- Service de Maladies Infectieuses et Tropicales, CHU Dupuytren, Limoges, France
| | - Selim Arraki Zava
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Pierre Marquet
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
| | - Jean-Baptiste Woillard
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France.
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France.
| |
Collapse
|
4
|
Alsultan A, Aljutayli A, Aljouie A, Albassam A, Woillard JB. Leveraging machine learning in limited sampling strategies for efficient estimation of the area under the curve in pharmacokinetic analysis: a review. Eur J Clin Pharmacol 2025; 81:183-201. [PMID: 39570408 DOI: 10.1007/s00228-024-03780-9] [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: 07/02/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE Limited sampling strategies are widely employed in clinical practice to minimize the number of blood samples required for the accurate area under the curve calculations, as obtaining these samples can be costly and challenging. Traditionally, the maximum a posteriori Bayesian estimation has been the standard method for the area under the curve estimation based on limited samples. However, machine learning is emerging as a promising alternative for this purpose. Here, we review studies that utilize machine learning approaches to develop limited sampling strategies and compare the strengths and weaknesses of these machine learning methods. METHODS We searched the literature for studies that used machine learning to estimate the area under the curve using a limited sampling strategy approach. RESULTS We identified ten studies that developed machine learning models to estimate the area under the curve for six different drugs. Several of these models demonstrated good accuracy and precision in area under the curve estimation in reference to the traditional Bayesian approach, highlighting the potential of machine learning models in precision dosing. CONCLUSIONS Despite these promising early results, the development of machine learning for limited sampling strategies is still in its early stages. Further research might be needed to validate machine learning models with larger, high-quality clinical datasets to ensure their reliability and applicability in clinical settings.
Collapse
Affiliation(s)
- Abdullah Alsultan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
- Clinical Pharmacokinetics and Pharmacodynamics Unit, King Saud University Medical City, Riyadh, Saudi Arabia.
| | - Abdullah Aljutayli
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Abdulrhman Aljouie
- Department of Data Management, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Artificial Intelligence and Bioinformatics, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Health Informatics, College of Public Health and Health Informatics, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Albassam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - 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
| |
Collapse
|
5
|
Bassani D, Parrott NJ, Manevski N, Zhang JD. Another string to your bow: machine learning prediction of the pharmacokinetic properties of small molecules. Expert Opin Drug Discov 2024; 19:683-698. [PMID: 38727016 DOI: 10.1080/17460441.2024.2348157] [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: 10/23/2023] [Accepted: 04/23/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Prediction of pharmacokinetic (PK) properties is crucial for drug discovery and development. Machine-learning (ML) models, which use statistical pattern recognition to learn correlations between input features (such as chemical structures) and target variables (such as PK parameters), are being increasingly used for this purpose. To embed ML models for PK prediction into workflows and to guide future development, a solid understanding of their applicability, advantages, limitations, and synergies with other approaches is necessary. AREAS COVERED This narrative review discusses the design and application of ML models to predict PK parameters of small molecules, especially in light of established approaches including in vitro-in vivo extrapolation (IVIVE) and physiologically based pharmacokinetic (PBPK) models. The authors illustrate scenarios in which the three approaches are used and emphasize how they enhance and complement each other. In particular, they highlight achievements, the state of the art and potentials of applying machine learning for PK prediction through a comphrehensive literature review. EXPERT OPINION ML models, when carefully crafted, regularly updated, and appropriately used, empower users to prioritize molecules with favorable PK properties. Informed practitioners can leverage these models to improve the efficiency of drug discovery and development process.
Collapse
Affiliation(s)
- Davide Bassani
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Neil John Parrott
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Nenad Manevski
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Jitao David Zhang
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
6
|
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
|
7
|
Ahmadi M, Alizadeh B, Ayyoubzadeh SM, Abiyarghamsari M. Predicting Pharmacokinetics of Drugs Using Artificial Intelligence Tools: A Systematic Review. Eur J Drug Metab Pharmacokinet 2024; 49:249-262. [PMID: 38457092 DOI: 10.1007/s13318-024-00883-7] [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: 01/29/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND AND OBJECTIVE Pharmacokinetic studies encompass the examination of the absorption, distribution, metabolism, and excretion of bioactive compounds. The pharmacokinetics of drugs exert a substantial influence on their efficacy and safety. Consequently, the investigation of pharmacokinetics holds great importance. However, laboratory-based assessment necessitates the use of numerous animals, various materials, and significant time. To mitigate these challenges, alternative methods such as artificial intelligence have emerged as a promising approach. This systematic review aims to review existing studies, focusing on the application of artificial intelligence tools in predicting the pharmacokinetics of drugs. METHODS A pre-prepared search strategy based on related keywords was used to search different databases (PubMed, Scopus, Web of Science). The process involved combining articles, eliminating duplicates, and screening articles based on their titles, abstracts, and full text. Articles were selected based on inclusion and exclusion criteria. Then, the quality of the included articles was assessed using an appraisal tool. RESULTS Ultimately, 23 relevant articles were included in this study. The clearance parameter received the highest level of investigation, followed by the area under the concentration-time curve (AUC) parameter, in pharmacokinetic studies. Among the various models employed in the articles, Random Forest and eXtreme Gradient Boosting (XGBoost) emerged as the most commonly utilized ones. Generalized Linear Models and Elastic Nets (GLMnet) and Random Forest models showed the most performance in predicting clearance. CONCLUSION Overall, artificial intelligence tools offer a robust, rapid, and precise means of predicting various pharmacokinetic parameters based on a dataset containing information of patients or drugs.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Alizadeh
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ayyoubzadeh
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Health Information Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdiye Abiyarghamsari
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, 1991953381, Iran.
| |
Collapse
|
8
|
Huang Z, Denti P, Mistry H, Kloprogge F. Machine Learning and Artificial Intelligence in PK-PD Modeling: Fad, Friend, or Foe? Clin Pharmacol Ther 2024; 115:652-654. [PMID: 38179832 PMCID: PMC11146679 DOI: 10.1002/cpt.3165] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/22/2023] [Indexed: 01/06/2024]
Affiliation(s)
- Zhonghui Huang
- Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Paolo Denti
- Division of Clinical Pharmacology, Department of MedicineUniversity of CapeCape TownSouth Africa
| | - Hitesh Mistry
- Division of PharmacyUniversity of ManchesterManchesterUK
| | - Frank Kloprogge
- Institute for Global HealthUniversity College LondonLondonUK
| |
Collapse
|
9
|
Li G, Sun Y, Zhu L. Application of machine learning combined with population pharmacokinetics to improve individual prediction of vancomycin clearance in simulated adult patients. Front Pharmacol 2024; 15:1352113. [PMID: 38562463 PMCID: PMC10982467 DOI: 10.3389/fphar.2024.1352113] [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: 12/12/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Background and aim Vancomycin, a glycopeptide antimicrobial drug. PPK has problems such as difficulty in accurately reflecting inter-individual differences, and the PPK model may not be accurate enough to predict individual pharmacokinetic parameters. Therefore, the aim of this study is to investigate whether the application of machine learning combined with the PPK method can improve the prediction of vancomycin CL in adult Chinese patients. Methods In the first step, a vancomycin CL prediction model for Chinese adult patients is given by PPK and Hamilton Monte Carlo sampling is used to obtain the reference CL of 1,000 patients; the second step is to obtain the final prediction model by machine learning using an appropriate model for the predictive factor and the reference CL; and the third step is to randomly select, in the simulated data, a total of 250 patients for prediction effect evaluation. Results XGBoost model is selected as final machine learning model. More than four-fifths of the subjects' predictive values regarding vancomycin CL are improved by machine learning combined with PPK. Machine learning combined with PPK models is more stable in performance than the PPK method alone for predicting models. Conclusion The first combination of PPK and machine learning for predictive modeling of vancomycin clearance in adult patients. It provides a reference for clinical pharmacists or clinicians to optimize the initial dosage given to ensure the effectiveness and safety of drug therapy for each patient.
Collapse
Affiliation(s)
- Guodong Li
- Department of Mathematics, Guilin University of Electronic Technology, Guilin, China
| | - Yubo Sun
- Department of Mathematics, Guilin University of Electronic Technology, Guilin, China
| | - Liping Zhu
- Department of Mathematics, Changji University, Xinjiang, China
| |
Collapse
|
10
|
Destere A, Teisseyre M, Merino D, Cremoni M, Gérard AO, Crepin T, Jourde-Chiche N, Graça D, Zorzi K, Fernandez C, Brglez V, Benzaken S, Esnault VL, Benito S, Drici MD, Seitz-Polski B. Optimization of Rituximab Therapy in Adult Patients With PLA2R1-Associated Membranous Nephropathy With Artificial Intelligence. Kidney Int Rep 2024; 9:134-144. [PMID: 38312797 PMCID: PMC10831377 DOI: 10.1016/j.ekir.2023.10.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 02/06/2024] Open
Abstract
Introduction Rituximab is a first-line treatment for membranous nephropathy. Nephrotic syndrome limits rituximab exposure due to urinary drug loss. Rituximab underdosing (serum level <2 μg/ml at month-3) is a risk factor for treatment failure. We developed a machine learning algorithm to predict the risk of underdosing based on patients' characteristics at rituximab infusion. We investigated the relationship between the predicted risk of underdosing and the cumulative dose of rituximab required to achieve remission. Methods Rituximab concentrations were measured at month-3 in 92 sera from adult patients with primary membranous nephropathy, split into a training (75%) and a testing set (25%). A forward-backward machine-learning procedure determined the best combination of variables to predict rituximab underdosing in the training data set, which was tested in the test set. The performances were evaluated for accuracy, sensitivity, and specificity in 10-fold cross-validation training and test sets. Results The best variables combination to predict rituximab underdosing included age, gender, body surface area (BSA), anti-phospholipase A2 receptor type 1 (anti-PLA2R1) antibody titer on day-0, serum albumin on day-0 and day-15, and serum creatinine on day-0 and day-15. The accuracy, sensitivity, and specificity were respectively 79.4%, 78.7%, and 81.0% (training data set), and 79.2%, 84.6% and 72.7% (testing data set). In both sets, the algorithm performed significantly better than chance (P < 0.05). Patients with an initial high probability of underdosing experienced a longer time to remission with higher rituximab cumulative doses required to achieved remission. Conclusion This algorithm could allow for early intensification of rituximab regimen in patients at high estimated risk of underdosing to increase the likelihood of remission.
Collapse
Affiliation(s)
- Alexandre Destere
- Département de Pharmacologie et de Pharmacovigilance, CHU de Nice, Université Côte d’Azur, France
- Université Côte d’Azur, Inria, CNRS, Laboratoire J.A. Dieudonné, Maasai team, Nice, France
| | - Maxime Teisseyre
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
- Département de Néphrologie, Dialyse et Transplantation, CHU de Nice, Université Côte d’Azur, France
| | - Diane Merino
- Département de Pharmacologie et de Pharmacovigilance, CHU de Nice, Université Côte d’Azur, France
| | - Marion Cremoni
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, France
| | - Alexandre O Gérard
- Département de Pharmacologie et de Pharmacovigilance, CHU de Nice, Université Côte d’Azur, France
- Département de Néphrologie, Dialyse et Transplantation, CHU de Nice, Université Côte d’Azur, France
| | - Thomas Crepin
- Département de Néphrologie, Dialyse et Transplantation, CHU de Besançon, Besançon, France
| | - Noémie Jourde-Chiche
- Département de Néphrologie, Dialyse et Transplantation, Hôpital de la Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Daisy Graça
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, France
| | - Kévin Zorzi
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
| | - Céline Fernandez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
| | - Vesna Brglez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, France
| | - Sylvia Benzaken
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, France
| | - Vincent L.M. Esnault
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Département de Néphrologie, Dialyse et Transplantation, CHU de Nice, Université Côte d’Azur, France
| | | | - Milou-Daniel Drici
- Département de Pharmacologie et de Pharmacovigilance, CHU de Nice, Université Côte d’Azur, France
| | - Barbara Seitz-Polski
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, CHU de Nice, France
- Département de Néphrologie, Dialyse et Transplantation, CHU de Nice, Université Côte d’Azur, France
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, France
| |
Collapse
|
11
|
Le Tilly O, Woillard JB. Evaluating Use of Artificial Intelligence for Drug Exposure and Effect Prediction. Kidney Int Rep 2024; 9:1-3. [PMID: 38312781 PMCID: PMC10831393 DOI: 10.1016/j.ekir.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024] Open
Affiliation(s)
- Olivier Le Tilly
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, Tours, France
- Service de Pharmacologie Médicale, CHRU Tours, Tours, France
| | - Jean-Baptiste Woillard
- P&T, Unité Mixte de Recherche 1248 Université de Limoges, Institut National de la Santé et de la Recherche Médicale, Limoges, France
- Service de Pharmacologie, Toxicologie et Pharmacovigilance, Centre Hospitalier Universitaire de Limoges, Limoges, France
| |
Collapse
|
12
|
Hughes JH, Tong DMH, Burns V, Daly B, Razavi P, Boelens JJ, Goswami S, Keizer RJ. Clinical decision support for chemotherapy-induced neutropenia using a hybrid pharmacodynamic/machine learning model. CPT Pharmacometrics Syst Pharmacol 2023; 12:1764-1776. [PMID: 37503916 PMCID: PMC10681461 DOI: 10.1002/psp4.13019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
Consensus guidelines recommend use of granulocyte colony stimulating factor in patients deemed at risk of chemotherapy-induced neutropenia, however, these risk models are limited in the factors they consider and miss some cases of neutropenia. Clinical decision making could be supported using models that better tailor their predictions to the individual patient using the wealth of data available in electronic health records (EHRs). Here, we present a hybrid pharmacokinetic/pharmacodynamic (PKPD)/machine learning (ML) approach that uses predictions and individual Bayesian parameter estimates from a PKPD model to enrich an ML model built on her data. We demonstrate this approach using models developed on a large real-world data set of 9121 patients treated for lymphoma, breast, or thoracic cancer. We also investigate the benefits of augmenting the training data using synthetic data simulated with the PKPD model. We find that PKPD-enrichment of ML models improves prediction of grade 3-4 neutropenia, as measured by higher precision (61%) and recall (39%) compared to PKPD model predictions (47%, 33%) or base ML model predictions (51%, 31%). PKPD augmentation of ML models showed minor improvements in recall (44%) but not precision (56%), and data augmentation required careful tuning to control overfitting its predictions to the PKPD model. PKPD enrichment of ML shows promise for leveraging both the physiology-informed predictions of PKPD and the ability of ML to learn predictor-outcome relationships from large data sets to predict patient response to drugs in a clinical precision dosing context.
Collapse
Affiliation(s)
| | | | | | - Bobby Daly
- Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Pedram Razavi
- Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | | | | | | |
Collapse
|
13
|
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: 1.5] [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
|
14
|
Habiballah S, Reisfeld B. Adapting physiologically-based pharmacokinetic models for machine learning applications. Sci Rep 2023; 13:14934. [PMID: 37696914 PMCID: PMC10495394 DOI: 10.1038/s41598-023-42165-3] [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: 04/08/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Both machine learning and physiologically-based pharmacokinetic models are becoming essential components of the drug development process. Integrating the predictive capabilities of physiologically-based pharmacokinetic (PBPK) models within machine learning (ML) pipelines could offer significant benefits in improving the accuracy and scope of drug screening and evaluation procedures. Here, we describe the development and testing of a self-contained machine learning module capable of faithfully recapitulating summary pharmacokinetic (PK) parameters produced by a full PBPK model, given a set of input drug-specific and regimen-specific information. Because of its widespread use in characterizing the disposition of orally administered drugs, the PBPK model chosen to demonstrate the methodology was an open-source implementation of a state-of-the-art compartmental and transit model called OpenCAT. The model was tested for drug formulations spanning a large range of solubility and absorption characteristics, and was evaluated for concordance against predictions of OpenCAT and relevant experimental data. In general, the values predicted by the ML models were within 20% of those of the PBPK model across the range of drug and formulation properties. However, summary PK parameter predictions from both the ML model and full PBPK model were occasionally poor with respect to those derived from experiments, suggesting deficiencies in the underlying PBPK model.
Collapse
Affiliation(s)
- Sohaib Habiballah
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523-1301, USA
| | - Brad Reisfeld
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523-1301, USA.
- School of Public Health, Colorado State University, Fort Collins, CO, 80523-1612, USA.
| |
Collapse
|
15
|
Li D, Zhao J, Xu B, Zheng Y, Liu M, Huang H, Han S, Wu X. Predicting busulfan exposure in patients undergoing hematopoietic stem cell transplantation using machine learning techniques. Expert Rev Clin Pharmacol 2023; 16:751-761. [PMID: 37326641 DOI: 10.1080/17512433.2023.2226866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
PURPOSE This study aimed to establish an optimal model to predict the busulfan (BU) area under the curve at steady state (AUCss) by using machine learning (ML). PATIENTS AND METHODS Seventy-nine adult patients (age ≥18 years) who received BU intravenously and underwent therapeutic drug monitoring from 2013 to 2021 at Fujian Medical University Union Hospital were enrolled in this retrospective study. The whole dataset was divided into a training group and test group at the ratio of 8:2. BU AUCss were considered as the target variable. Nine different ML algorithms and one population pharmacokinetic (pop PK) model were developed and validated, and their predictive performance was compared. RESULTS All ML models were superior to the pop PK model (R2 = 0.751, MSE = 0.722, 14 and RMSE = 0.830) in model fitting and had better predictive accuracy. The ML model of BU AUCss established through support vector regression (SVR) and gradient boosted regression trees (GBRT) had the best predictive ability (R2 = 0.953 and 0.953, MSE = 0.323 and 0.326, and RMSE = 0.423 and 0.425). CONCLUSION All the ML models can potentially be used to estimate BU AUCss with the aim of facilitating rational use of BU on the individualized level, especially models built by SVR and GBRT algorithms.
Collapse
Affiliation(s)
- Dandan Li
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jingtong Zhao
- School of Economics, Renmin University of China, Beijing, China
| | - Baohua Xu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - You Zheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huiping Huang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Song Han
- School of Economics, Renmin University of China, Beijing, China
| | - Xuemei Wu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
16
|
Destere A, Marquet P, Labriffe M, Drici MD, Woillard JB. A Hybrid Algorithm Combining Population Pharmacokinetic and Machine Learning for Isavuconazole Exposure Prediction. Pharm Res 2023; 40:951-959. [PMID: 36991227 DOI: 10.1007/s11095-023-03507-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVES Maximum a posteriori Bayesian estimation (MAP-BE) based on a limited sampling strategy and a population pharmacokinetic (POPPK) model is used to estimate individual pharmacokinetic parameters. Recently, we proposed a methodology that combined population pharmacokinetic and machine learning (ML) to decrease the bias and imprecision in individual iohexol clearance prediction. The aim of this study was to confirm the previous results by developing a hybrid algorithm combining POPPK, MAP-BE and ML that accurately predicts isavuconazole clearance. METHODS A total of 1727 isavuconazole rich PK profiles were simulated using a POPPK model from the literature, and MAP-BE was used to estimate the clearance based on: (i) the full PK profiles (refCL); and (ii) C24h only (C24h-CL). Xgboost was trained to correct the error between refCL and C24h-CL in the training dataset (75%). C24h-CL as well as ML-corrected C24h-CL were evaluated in a testing dataset (25%) and then in a set of PK profiles simulated using another published POPPK model. RESULTS A strong decrease in mean predictive error (MPE%), imprecision (RMSE%) and the number of profiles outside ± 20% MPE% (n-out20%) was observed with the hybrid algorithm (decreased in MPE% by 95.8% and 85.6%; RMSE% by 69.5% and 69.0%; n-out20% by 97.4% and 100% in the training and testing sets, respectively. In the external validation set, the hybrid algorithm decreased MPE% by 96%, RMSE% by 68% and n-out20% by 100%. CONCLUSION The hybrid model proposed significantly improved isavuconazole AUC estimation over MAP-BE based on the sole C24h and may improve dose adjustment.
Collapse
Affiliation(s)
- Alexandre Destere
- Pharmacology and Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, 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, CHU de 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, CHU de Limoges, Limoges, France
| | - Milou-Daniel Drici
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, 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, CHU de Limoges, Limoges, France.
| |
Collapse
|
17
|
Destere A, Merino D, Bonesso L, Lavrut T, Bernasconni A, Garraffo R, Gérard AO, Drici MD. A HPLC-DAD method to facilitate large-scale therapeutic drug monitoring of dalbavancin. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1222:123694. [PMID: 37060815 DOI: 10.1016/j.jchromb.2023.123694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/08/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Dalbavancin, a long-acting lipoglycopeptide antibiotic targeting susceptible Gram-positive bacteria, is WHO critically important antibiotic, increasingly used in critical situations such as osteoarticular infections. To ensure its effectiveness and its safety, the therapeutic drug monitoring (TDM) of dalbavancin is strongly recommended. In the absence of an available minimum inhibitory concentration (MIC), the European Committee on Antimicrobial Susceptibility Testing (EUCAST) recommends a breakpoint of 0.125 mg/L for Staphylococcus aureus, corresponding to a trough target concentration of 25 mg/L. Nowadays, the TDM is usually performed using a high-performance liquid chromatography (HPLC) method coupled with a tandem mass spectrometry. However, this expensive and specialized equipment and reagents may be difficult to acquire for non-specialized laboratories. The use of HPLC coupled with diode array detector (DAD) facilitates TDM with a lower cost, while preserving the reliability of the results. Our aim was to provide a sensitive and specific method, relying on HPLC-DAD for extending the TDM of dalbavancin beyond non-specialized labs, therefore maximizing its efficiency and Benefit/risk ratio. Our method complied with the European Medicines Agency guidelines of bioanalytical validation. Irrespective of the concentrations of dalbavancin, the coefficient of variation < 10% confirmed the reliability of this analytical method, with a calibration curve ranging from 5 to 100 mg/L. No interferences nor carryover was observed. Our HPLC-DAD method, combined with a simple extraction, provides a widely usable, inexpensive and easy-to-implement new asset for the TDM of Dalbavancin.
Collapse
Affiliation(s)
- Alexandre Destere
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Diane Merino
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Laurent Bonesso
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Thibaud Lavrut
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Anaïs Bernasconni
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Rodolphe Garraffo
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Alexandre O Gérard
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France
| | - Milou-Daniel Drici
- Department of Pharmacology and Pharmacovigilance Center, Côte d'Azur University Medical Center, Nice, France.
| |
Collapse
|