1
|
Centanni M, Reijnhout N, Thijs A, Karlsson MO, Friberg LE. Pharmacogenetic Testing or Therapeutic Drug Monitoring: A Quantitative Framework. Clin Pharmacokinet 2024; 63:871-884. [PMID: 38842789 PMCID: PMC11222190 DOI: 10.1007/s40262-024-01382-3] [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: 05/05/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Pharmacogenetic profiling and therapeutic drug monitoring (TDM) have both been proposed to manage inter-individual variability (IIV) in drug exposure. However, determining the most effective approach for estimating exposure for a particular drug remains a challenge. This study aimed to quantitatively assess the circumstances in which pharmacogenetic profiling may outperform TDM in estimating drug exposure, under three sources of variability (IIV, inter-occasion variability [IOV], and residual unexplained variability [RUV]). METHODS Pharmacokinetic models were selected from the literature corresponding to drugs for which pharmacogenetic profiling and TDM are both clinically considered approaches for dose individualization. The models were used to simulate relevant drug exposures (trough concentration or area under the curve [AUC]) under varying degrees of IIV, IOV, and RUV. RESULTS Six drug cases were selected from the literature. Model-based simulations demonstrated that the percentage of patients for whom pharmacogenetic exposure prediction is superior to TDM differs for each drug case: tacrolimus (11.0%), tamoxifen (12.7%), efavirenz (49.2%), vincristine (49.6%), risperidone (48.1%), and 5-fluorouracil (5-FU) (100%). Generally, in the presence of higher unexplained IIV in combination with lower RUV and IOV, exposure was best estimated by TDM, whereas, under lower unexplained IIV in combination with higher IOV or RUV, pharmacogenetic profiling was preferred. CONCLUSIONS For the drugs with relatively low RUV and IOV (e.g., tamoxifen and tacrolimus), TDM estimated true exposure the best. Conversely, for drugs with similar or lower unexplained IIV (e.g., efavirenz or 5-FU, respectively) combined with relatively high RUV, pharmacogenetic profiling provided the most accurate estimate for most patients. However, genotype prevalence and the relative influence of genotypes on the PK, as well as the ability of TDM to accurately estimate AUC with a limited number of samples, had an impact. The results could be used to support clinical decision making when considering other factors, such as the probability for severe side effects.
Collapse
Affiliation(s)
- Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, 751 23, Uppsala, Sweden
| | - Niels Reijnhout
- Department of Pharmacy, Uppsala University, Box 580, 751 23, Uppsala, Sweden
| | - Abel Thijs
- Department of Internal Medicine, Amsterdam UMC, Location VU University, Amsterdam, The Netherlands
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, Box 580, 751 23, Uppsala, Sweden
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, 751 23, Uppsala, Sweden.
| |
Collapse
|
2
|
Zhang X, Sui Y, Yu L, Zhou M, Zhang C, Liu D, Chen X, Yang L, Sui Y. Population Pharmacokinetic Analysis of Follicle-Stimulating Hormone During Ovarian Stimulation: Relation with Weight, Prolactin and Gene Polymorphism in THADA and ADIPOQ. Clin Pharmacokinet 2023; 62:1493-1507. [PMID: 37632631 DOI: 10.1007/s40262-023-01299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Personalisation strategies of ovarian stimulation for in vitro fertilisation (IVF)/ intracytoplasmic sperm injection (ICSI) treatments using exogenous follicle-stimulating hormone (FSH) have been extensively studied over the past 20 years. This research aimed to develop a FSH population pharmacokinetic (PPK) model taking into account the contribution of gene polymorphisms in Chinese reproductive-age women. METHODS Data from 173 patients undergoing GnRH agonist down-regulation long protocols of IVF/ICSI treatment were collected. PPK analysis was subsequently conducted using the nonlinear mixed-effect model (NONMEM) software. Several covariates, including 18 single nucleotide polymorphisms, demographic factors and biological characteristics, were evaluated. The final PPK model was extensively validated using bootstrapping and normalised prediction error distribution, as well as external validation on an independent group of 35 patients. RESULTS FSH PPK was accurately described by a one-compartment model with first-order absorption. The typical population value of apparent clearance was estimated to be 0.81 L/h [relative standard errors (RSE) 5.3%] with an inter-individual variability (IIV) of 16.0%. The typical apparent distribution volume was 8.36 L (RSE 9.7%, 59.7% IIV), and the absorption rate constant was estimated to be 0.0444 h-1 (RSE 9.1%). Body weight, basal prolactin concentration and the gene ADIPOQ (rs1501299) showed a significant covariate effect on the FSH clearance rate and exposure concentration. Genotypes of THADA (rs12478601) significantly influenced the distribution volume. Simulation results indicated that patients with the TT genotype of THADA (rs12478601) required a longer time to reach steady state and had less fluctuation in FSH levels. Model evaluations showed that the final model accurately and precisely described the observed data and demonstrated effective prediction performance. CONCLUSION PPK models of FSH have been developed, which could potentially be used for FSH dosage individualisation in the clinical setting. CLINICAL TRIAL REGISTRATION This study has been registered with the Chinese Clinical Trials Registry (ChiCTR2100049142).
Collapse
Affiliation(s)
- Xiaowei Zhang
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China.
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China.
| | - Yu Sui
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Institute of Health Sciences, China Medical University, Shenyang, China
| | - Lei Yu
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Zhou
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
| | - Chong Zhang
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
| | - Danhua Liu
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
| | - Xinren Chen
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
| | - Liqun Yang
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China
| | - Yang Sui
- NHC Key Laboratory of Reproductive Health and Medical Genetics, China Medical University, Shenyang, China.
- Reproductive Affiliated Hospital of China Medical University, PuHe Street 10, Huanggu District, Shenyang, 110031, China.
| |
Collapse
|
3
|
Li ZR, Li RD, Niu WJ, Zheng XY, Wang ZX, Zhong MK, Qiu XY. Population Pharmacokinetic Modeling Combined With Machine Learning Approach Improved Tacrolimus Trough Concentration Prediction in Chinese Adult Liver Transplant Recipients. J Clin Pharmacol 2023; 63:314-325. [PMID: 36097320 DOI: 10.1002/jcph.2156] [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: 06/21/2022] [Accepted: 09/05/2022] [Indexed: 12/30/2022]
Abstract
This study aimed to develop and evaluate a population pharmacokinetic (PPK) combined machine learning approach to predict tacrolimus trough concentrations for Chinese adult liver transplant recipients in the early posttransplant period. Tacrolimus trough concentrations were retrospectively collected from routine monitoring records of liver transplant recipients and divided into the training data set (1287 concentrations in 145 recipients) and the test data set (296 concentrations in 36 recipients). A PPK model was first established using NONMEM. Then a machine learning model of Xgboost was adapted to fit the estimated individual pharmacokinetic parameters obtained from the PPK model with Bayesian forecasting. The performance of the final PPK model and Xgboost model was compared in the test data set. In the final PPK model, tacrolimus daily dose, postoperative days, hematocrit, aspartate aminotransferase, and concomitant voriconazole, were identified to significantly influence the clearance. The postoperative days along with hematocrit significantly influence the volume of distribution. In the Xgboost model, the first 5 predictors for predicting the clearance were concomitant with voriconazole, sex, single nucleotide polymorphisms of CYP3A4*1G and CYP3A5*3 in recipients, and tacrolimus daily dose, for the volume of distribution were postoperative days, age, weight, total bilirubin and graft : recipient weight ratio. In the test data set, the Xgboost model showed the minimum median prediction error of tacrolimus concentrations, less than the PPK model with or without Bayesian forecasting. In conclusion, a PPK combined machine learning approach could improve the prediction of tacrolimus concentrations for Chinese adult liver transplant recipients in the early posttransplant period.
Collapse
Affiliation(s)
- Zi-Ran Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Rui-Dong Li
- Liver Transplant Centre, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Wan-Jie Niu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Xin-Yi Zheng
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Zheng-Xin Wang
- Liver Transplant Centre, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Xiao-Yan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
4
|
Optimized Dosing: The Next Step in Precision Medicine in Non-Small-Cell Lung Cancer. Drugs 2021; 82:15-32. [PMID: 34894338 DOI: 10.1007/s40265-021-01654-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
In oncology, and especially in the treatment of non-small-cell lung cancer (NSCLC), dose optimization is often a neglected part of precision medicine. Many drugs are still being administered in "one dose fits all" regimens or based on parameters that are often only minor determinants for systemic exposure. These dosing approaches often introduce additional pharmacokinetic variability and do not add to treatment outcomes. Fortunately, pharmacological knowledge is increasing, providing valuable information regarding the potential of, for example, therapeutic drug monitoring. This article focuses on the evidence for the most promising and easily implemented optimized dosing approaches for the small-molecule inhibitors, chemotherapeutic agents, and monoclonal antibodies as treatment options currently approved for NSCLC. Despite limitations such as investigations having been conducted in oncological diseases other than NSCLC or the retrospective origin of many analyses, an alternative dosing regimen could be beneficial for treatment outcomes, prescriber convenience, or financial burden on healthcare systems. This review of the literature provides recommendations on the implementation of dose optimization and advice regarding promising strategies that deserve further research in NSCLC.
Collapse
|
5
|
Le Louedec F, Puisset F, Thomas F, Chatelut É, White-Koning M. Easy and reliable maximum a posteriori Bayesian estimation of pharmacokinetic parameters with the open-source R package mapbayr. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1208-1220. [PMID: 34342170 PMCID: PMC8520754 DOI: 10.1002/psp4.12689] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/28/2022]
Abstract
Pharmacokinetic (PK) parameter estimation is a critical and complex step in the model‐informed precision dosing (MIPD) approach. The mapbayr package was developed to perform maximum a posteriori Bayesian estimation (MAP‐BE) in R from any population PK model coded in mrgsolve. The performances of mapbayr were assessed using two approaches. First, “test” models with different features were coded, for example, first‐order and zero‐order absorption, lag time, time‐varying covariates, Michaelis–Menten elimination, combined and exponential residual error, parent drug and metabolite, and small or large inter‐individual variability (IIV). A total of 4000 PK profiles (combining single/multiple dosing and rich/sparse sampling) were simulated from each test model, and MAP‐BE of parameters was performed in both mapbayr and NONMEM. Second, a similar procedure was conducted with seven “real” previously published models to compare mapbayr and NONMEM on a PK outcome used in MIPD. For the test models, 98% of mapbayr estimations were identical to those given by NONMEM. Some discordances could be observed when dose‐related parameters were estimated or when models with large IIV were used. The exploration of objective function values suggested that mapbayr might outdo NONMEM in specific cases. For the real models, a concordance close to 100% on PK outcomes was observed. The mapbayr package provides a reliable solution to perform MAP‐BE of PK parameters in R. It also includes functions dedicated to data formatting and reporting and enables the creation of standalone Shiny web applications dedicated to MIPD, whatever the model or the clinical protocol and without additional software other than R.
Collapse
Affiliation(s)
- Félicien Le Louedec
- Inserm UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Faculty of Pharmacy, Université Paul Sabatier Toulouse III, Toulouse, France.,Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Florent Puisset
- Inserm UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Faculty of Pharmacy, Université Paul Sabatier Toulouse III, Toulouse, France.,Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Fabienne Thomas
- Inserm UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Faculty of Pharmacy, Université Paul Sabatier Toulouse III, Toulouse, France.,Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Étienne Chatelut
- Inserm UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Faculty of Pharmacy, Université Paul Sabatier Toulouse III, Toulouse, France.,Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Mélanie White-Koning
- Inserm UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Faculty of Pharmacy, Université Paul Sabatier Toulouse III, Toulouse, France
| |
Collapse
|
6
|
Tauzin M, Tréluyer JM, Nabbout R, Chemaly N, Billette de Villemeur T, Desguerre I, Lui G, Gana I, Boujaafar S, Zheng Y, Benaboud S, Bouazza N, Chenevier-Gobeaux C, Freihuber C, Hirt D. Predictive Performance of Population Pharmacokinetic Models of Levetiracetam in Children and Evaluation of Dosing Regimen. J Clin Pharmacol 2021; 61:1366-1375. [PMID: 33997989 DOI: 10.1002/jcph.1910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 05/11/2021] [Indexed: 11/08/2022]
Abstract
Levetiracetam is a broad-spectrum antiepileptic drug that exhibits high interindividual variability in serum concentrations in children. A population pharmacokinetic approach can be used to explain this variability and optimize dosing schemes. The objectives are to identify the best predictive population pharmacokinetic model for children and to evaluate recommended doses using simulations and Bayesian forecasting. A validation cohort included children treated with levetiracetam who had a serum drug concentration assayed during therapeutic drug monitoring. We assessed the predictive performance of all the population pharmacokinetic models published in the literature using mean prediction errors, root mean squared errors, and visual predictive checks. A population model was finally constructed on the data, and dose simulations were performed to evaluate doses. We included 267 levetiracetam concentrations ranging from 2 to 69 mg/L from 194 children in the validation cohort. Six published models were externally evaluated. Most of the models underestimated the variability of our population. A 1-compartment model with first-order absorption and elimination with allometric scaling was finally fitted on our data. In our cohort, 57% of patients had a trough concentration <12 mg/L and 12% <5 mg/L. To reach a trough concentration >5 mg/L, doses ≥30 mg/kg/d for patients ≤50 kg and ≥2000 mg/d for patients >50 kg are required. In our population, a high percentage of children had low trough concentrations. Our population pharmacokinetic model could be used for therapeutic drug monitoring of levetiracetam in children.
Collapse
Affiliation(s)
- Manon Tauzin
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,Réanimation néonatale et néonatologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean-Marc Tréluyer
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France.,Unité de recherche Clinique, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes, Paris, France
| | - Rima Nabbout
- Centre de référence épilepsies rares, Service de Neurologie pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Nicole Chemaly
- Centre de référence épilepsies rares, Service de Neurologie pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Thierry Billette de Villemeur
- Sorbonne Université, UPMC, GRC ConCer-LD and AP-HP, Hôpital Trousseau, Service de Neuropédiatrie-Pathologie du développement, Centre de référence des déficits intellectuels de causes rares, Paris, France
| | - Isabelle Desguerre
- Centre de référence épilepsies rares, Service de Neurologie pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Gabrielle Lui
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Ines Gana
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
| | - Sana Boujaafar
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,Unité de recherche Clinique, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes, Paris, France
| | - Yi Zheng
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
| | - Sihem Benaboud
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Naim Bouazza
- EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Camille Chenevier-Gobeaux
- Service de Diagnostic Biologique Automatisé, Hôpital Cochin, Hôpitaux Universitaires Paris Centre (HUPC), Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Cécile Freihuber
- Sorbonne Université, UPMC, GRC ConCer-LD and AP-HP, Hôpital Trousseau, Service de Neuropédiatrie-Pathologie du développement, Centre de référence des déficits intellectuels de causes rares, Paris, France
| | - Déborah Hirt
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France.,EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France.,Inserm 1018 CESP, Hôpital Bicêtre, Le Kremlin-Bicêtre, Paris, France
| |
Collapse
|
7
|
Gonzalez D, Sinha J. Pediatric Drug-Drug Interaction Evaluation: Drug, Patient Population, and Methodological Considerations. J Clin Pharmacol 2021; 61 Suppl 1:S175-S187. [PMID: 34185913 PMCID: PMC8500325 DOI: 10.1002/jcph.1881] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/18/2021] [Indexed: 12/27/2022]
Abstract
Hospitalized pediatric patients and those with complex or chronic conditions treated on an outpatient basis are commonly prescribed multiple drugs, resulting in increased risk for drug-drug interactions (DDIs). Although dedicated DDI evaluations are routinely performed in healthy adult volunteers during drug development, they are rarely performed in pediatric patients because of ethical, logistical, and methodological challenges. In the absence of pediatric DDI evaluations, adult DDI data are often extrapolated to pediatric patients. However, the magnitude of a DDI in pediatric patients may differ from adults because of age-dependent physiological changes that can impact drug disposition or response and because of other factors related to the drug (eg, dose, formulation) and the patient population (eg, disease state, obesity). Therefore, the DDI magnitude needs to be assessed in children separately from adults, although a lack of clinical DDI data in pediatric populations makes this evaluation challenging. As a result, pediatric DDI assessment relies on the predictive performance of the pharmacometric approaches used, such as population and physiologically based pharmacokinetic modeling. Therefore, careful consideration needs to be given to adequately account for the age-dependent physiological changes in these models to build high confidence for such untested DDI scenarios. This review article summarizes the key considerations related to the drug, patient population, and methodology, and how they can impact DDI evaluation in the pediatric population.
Collapse
Affiliation(s)
- Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jaydeep Sinha
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Carona A, Bicker J, Silva R, Fonseca C, Falcão A, Fortuna A. Pharmacology of lacosamide: From its molecular mechanisms and pharmacokinetics to future therapeutic applications. Life Sci 2021; 275:119342. [PMID: 33713668 DOI: 10.1016/j.lfs.2021.119342] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 01/14/2023]
Abstract
Epilepsy is one of the most common brain disorders, affecting more than 50 million people worldwide. Although its treatment is currently symptomatic, the last generation of anti-seizure drugs is characterized by better pharmacokinetic profiles, efficacy, tolerability and safety. Lacosamide is a third-generation anti-seizure drug that stands out due to its good efficacy and safety profile. It is used with effectiveness in the treatment of partial-onset seizures with or without secondary generalization, primary generalized tonic-clonic seizures and off-label in status epilepticus. Despite scarcely performed until today, therapeutic drug monitoring of lacosamide is proving to be advantageous by allowing the control of inter and intra-individual variability and promoting a successful personalized therapy, particularly in special populations. Herein, the pharmacology, pharmacokinetics, and clinical data of lacosamide were reviewed, giving special emphasis to the latest molecular investigations underlying its mechanism of action and therapeutic applications in pathologies besides epilepsy. In addition, the pharmacokinetic characteristics of lacosamide were updated, as well as current literature concerning the high pharmacokinetic variability observed in special patient populations and that must be considered during treatment individualization.
Collapse
Affiliation(s)
- Andreia Carona
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Rui Silva
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Carla Fonseca
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
9
|
Tauzin M, Tréluyer JM, Nabbout R, Billette de Villemeur T, Desguerre I, Aboura R, Gana I, Zheng Y, Benaboud S, Bouazza N, Chenevier-Gobeaux C, Freihuber C, Hirt D. Dosing Recommendations for Lamotrigine in Children: Evaluation Based on Previous and New Population Pharmacokinetic Models. J Clin Pharmacol 2020; 61:677-687. [PMID: 33244764 DOI: 10.1002/jcph.1791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022]
Abstract
Lamotrigine is a broad-spectrum antiepileptic drug with high interindividual variability in serum concentrations in children. The aims of this study were to evaluate the predictive performance of pediatric population pharmacokinetic (PPK) models published on lamotrigine, to build a new model with our monitoring data and to evaluate the current recommended doses. A validation cohort included patients treated with lamotrigine who had a serum level assayed during therapeutic drug monitoring (TDM). PPK models published in the literature were first applied to the validation cohort. We assessed their predictive performance using mean prediction errors, root mean squared errors, and visual predictive checks. A new model was then built using the data. Dose simulations were performed to evaluate the doses recommended. We included 270 lamotrigine concentrations ranging from 0.5 to 17.9 mg/L from 175 patients. The median (range) age and weight were 11.8 years (0.8-18 years) and 32.7 kg (8-110 kg). We tested 6 PPK models; most had acceptable bias and precision but underestimated the variability of the cohort. We built a 1-compartment model with first-order absorption and elimination, allometric scaling, and effects of inhibitor and inducer comedications. In our cohort, 22.6% of trough concentrations were below 2.5 mg/L. In conclusion, we proposed a PPK model that can be used for TDM of lamotrigine in children. In our population, a high percentage of children had low trough concentrations of lamotrigine. As the intervals of recommended doses are large, we suggest aiming at the higher range of doses to reach the target concentration.
Collapse
Affiliation(s)
- Manon Tauzin
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
- Réanimation néonatale et néonatologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean-Marc Tréluyer
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
- EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
- Unité de recherche Clinique, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes, Paris, France
| | - Rima Nabbout
- Centre de référence épilepsies rares, Service de Neurologie pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Thierry Billette de Villemeur
- Sorbonne Université, UPMC, GRC ConCer-LD and AP-HP, Hôpital Trousseau, Service de Neuropédiatrie - Pathologie du développement, Centre de référence des déficits intellectuels de causes rares, Paris, France
| | - Isabelle Desguerre
- Centre de référence épilepsies rares, Service de Neurologie pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Radia Aboura
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
| | - Ines Gana
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
| | - Yi Zheng
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
| | - Sihem Benaboud
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
- EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Naim Bouazza
- EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Camille Chenevier-Gobeaux
- Service de Diagnostic Biologique Automatisé, Hôpital Cochin, Hôpitaux Universitaires Paris Centre (HUPC), Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Cécile Freihuber
- Sorbonne Université, UPMC, GRC ConCer-LD and AP-HP, Hôpital Trousseau, Service de Neuropédiatrie - Pathologie du développement, Centre de référence des déficits intellectuels de causes rares, Paris, France
| | - Déborah Hirt
- Service de Pharmacologie Clinique, Hôpital Cochin, APHP, Paris, France
- EA 7323, Université Paris Descartes Sorbonne Paris Cité, Paris, France
- Inserm 1018 CESP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
10
|
Krishnaswami S, Austin D, Della Pasqua O, Gastonguay MR, Gobburu J, van der Graaf PH, Ouellet D, Tannenbaum S, Visser SAG. MID3: Mission Impossible or Model-Informed Drug Discovery and Development? Point-Counterpoint Discussions on Key Challenges. Clin Pharmacol Ther 2020; 107:762-772. [PMID: 31955417 PMCID: PMC7158219 DOI: 10.1002/cpt.1788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/03/2020] [Indexed: 11/12/2022]
|
11
|
van Dijkman SC, de Jager NCB, Rauwé WM, Danhof M, Della Pasqua O. Effect of Age-Related Factors on the Pharmacokinetics of Lamotrigine and Potential Implications for Maintenance Dose Optimisation in Future Clinical Trials. Clin Pharmacokinet 2019; 57:1039-1053. [PMID: 29363050 DOI: 10.1007/s40262-017-0614-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND AIMS In this study, we evaluate the performance of allometric concepts to predict the implications of age and size on the pharmacokinetics of lamotrigine, and assess the dose rationale across different age groups from 0.2 to 91 years. METHODS An allometrically scaled pharmacokinetic model was developed using adolescent and adult data, taking into account the effect of comedications. Model parameters were then used to extrapolate lamotrigine pharmacokinetics to older adults (> 65 years), children (4-12 years) and infants and toddlers (0.2-2.0 years). In addition, simulations were performed to identify the implication of different doses and dosing regimens for each population, so as to ensure steady-state concentrations within a predefined reference range. RESULTS The pharmacokinetics of lamotrigine was best described using a one-compartment model with first-order absorption and elimination. Carbamazepine, phenytoin, and valproic acid changed systemic clearance (CL) by + 76.5, + 129, and - 47.4%, respectively. Allometric principles allowed accurate extrapolation of disposition parameters to older adults and children older than 4 years of age. A maturation function was required to describe changes in exposure in younger patients. Compared with adults, a child aged 1.7 years has a 31.5% higher CL, after correcting for body weight. Patients > 65 years of age showed a decrease in CL of approximately 15%. CONCLUSION Population pharmacokinetic models are usually limited to a subgroup of patients, which may mask the identification of factors contributing to interindividual variability. The availability of an integrated model including the whole patient population provides insight into the role of age-related changes in the disposition of lamotrigine, and potential implications for maintenance dose optimisation in any future trials. TRIAL REGISTRATION According to GlaxoSmithKline's Clinical Trial Register, data from the GlaxoSmithKline studies LAM100034 and LEP103944, corresponding to ClinicalTrials.gov identifiers NCT00113165 and NCT00264615, used in this work, have been used in previous publications (doi: https://doi.org/10.1212/01.wnl.0000277698.33743.8b , https://doi.org/10.1111/j.1528-1167.2007.01274.x ).
Collapse
Affiliation(s)
- Sven C van Dijkman
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Nico C B de Jager
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Willem M Rauwé
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Oscar Della Pasqua
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333CC, Leiden, The Netherlands. .,Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Uxbridge, UB11 1BT, UK. .,Clinical Pharmacology and Therapeutics Group, University College London, BMA House (North Entrance), Tavistock Square, London, WC1H 9JP, UK.
| |
Collapse
|
12
|
Wang R, Cui Y, Hu F, Liu W, Du Q, Zhang Y, Zha J, Huang T, Fizir M, He H. Selective recognition and enrichment of carbamazepine in biological samples by magnetic imprinted polymer based on reversible addition-fragmentation chain transfer polymerization. J Chromatogr A 2019; 1591:62-70. [PMID: 30712819 DOI: 10.1016/j.chroma.2019.01.057] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/14/2019] [Accepted: 01/18/2019] [Indexed: 01/01/2023]
Abstract
A well-defined molecularly imprinted polymer (Fe3O4@CS@MIP) was synthesized via reversible addition-fragmentation chain transfer polymerization for magnetic solid-phase extraction coupled with high-performance liquid chromatography-diode array detector to detect carbamazepine (CBZ) in biological samples. The composition of Fe3O4@CS@MIP was selected by a two-step screening method. 4-vinyl pyridine, divinylbenzene and dimethylformamide were chosen as the functional monomer, cross-linker and porogen, respectively. The imprinted layer was coated on the surface of the chain transfer agent-modified magnetic chitosan nanoparticles. The prepared Fe3O4@CS@MIP was characterized by scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, Brunauer-Emmett-Teller measurement and vibrating sample magnetometer. The results indicated that Fe3O4@CS@MIP had a large surface area (265.8 m2/g), high saturation magnetization (19.88 emu/g) and uniform structure. Besides, the binding property of the Fe3O4@CS@MIP was studied in detail. The Fe3O4@CS@MIP showed high imprinting factor (IF = 4.83) and desirable adsorption capacity (323.10 μmol/g) to CBZ. Under the optimum conditions, the developed method exhibited excellent linearity (R2>0.999) in the range of 0.01-0.5 mg/L and 1.0-30.0 mg/L, and the limits of detection were 1.0 μg/L and 9.6 μg/L for the urine and serum samples, respectively. Good recoveries (88.22%-101.18%) were obtained with relative standard deviations less than 4.83%. This work provided a practical approach for the selective extraction and detection of CBZ in real samples.
Collapse
Affiliation(s)
- Ruya Wang
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Yanru Cui
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Fan Hu
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Wei Liu
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiuzheng Du
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Yan Zhang
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun Zha
- Vocational and Technical College of Guizhou Minzu University, Guiyang, Guizhou, 550025, China
| | - Tao Huang
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Meriem Fizir
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| | - Hua He
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing, 211198, China; Vocational and Technical College of Guizhou Minzu University, Guiyang, Guizhou, 550025, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
13
|
Tauzin M, Tréluyer JM, Nabbout R, Billette de Villemeur T, Desguerre I, Aboura R, Gana I, Zheng Y, Benaboud S, Bouazza N, Chenevier-Gobeaux C, Freihuber C, Hirt D. Simulations of Valproate Doses Based on an External Evaluation of Pediatric Population Pharmacokinetic Models. J Clin Pharmacol 2018; 59:406-417. [DOI: 10.1002/jcph.1333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/02/2018] [Indexed: 01/28/2023]
Affiliation(s)
- Manon Tauzin
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
| | - Jean-Marc Tréluyer
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
- EA 7323; Université Paris Descartes Sorbonne Paris Cité; Paris France
- Unité de recherche Clinique; Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes; Paris France
| | - Rima Nabbout
- Centre de référence épilepsies rares; Service de Neurologie pédiatrique; Hôpital Necker Enfants Malades; APHP; Paris France
| | - Thierry Billette de Villemeur
- Sorbonne Université; UPMC; GRC ConCer-LD and AP-HP; Hôpital Trousseau, Service de Neuropédiatrie - Pathologie du développement, Centre de référence des déficits intellectuels de causes rares; Inserm U 1141 Paris France
| | - Isabelle Desguerre
- Centre de référence épilepsies rares; Service de Neurologie pédiatrique; Hôpital Necker Enfants Malades; APHP; Paris France
| | - Radia Aboura
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
| | - Ines Gana
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
| | - Yi Zheng
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
| | - Sihem Benaboud
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
- EA 7323; Université Paris Descartes Sorbonne Paris Cité; Paris France
| | - Naim Bouazza
- Unité de recherche Clinique; Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes; Paris France
| | - Camille Chenevier-Gobeaux
- Service de Diagnostic Biologique Automatisé; Hôpital Cochin; Hôpitaux Universitaires Paris Centre (HUPC); Assistance Publique des Hôpitaux de Paris (APHP); Paris France
| | - Cécile Freihuber
- Sorbonne Université; UPMC; GRC ConCer-LD and AP-HP; Hôpital Trousseau, Service de Neuropédiatrie - Pathologie du développement, Centre de référence des déficits intellectuels de causes rares; Inserm U 1141 Paris France
| | - Déborah Hirt
- Service de Pharmacologie Clinique; Hôpital Cochin; APHP; Paris France
- EA 7323; Université Paris Descartes Sorbonne Paris Cité; Paris France
- Unité de recherche Clinique; Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Descartes; Paris France
| |
Collapse
|
14
|
Liang J, Zhang Z, Zhao H, Wan S, Zhai X, Zhou J, Liang R, Deng Q, Wu Y, Lin G. Simple and rapid monitoring of doxorubicin using streptavidin-modified microparticle-based time-resolved fluorescence immunoassay. RSC Adv 2018; 8:15621-15631. [PMID: 35539486 PMCID: PMC9080157 DOI: 10.1039/c8ra01807c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
Developing a simple analytical method suitable for therapeutic drug monitoring in a clinical setting is key to establishing guidelines on accurate dose administration and the advancement of precision medicine. We devised a simple rapid analytical method through the combination of streptavidin-modified microparticles and a time-resolved fluorescence immunoassay for therapeutic drug monitoring. The analytical performance of this method was investigated and validated using clinical samples. By determination of doxorubicin concentration, the proposed assay has shown a satisfactory linear range of detection (3.8-3000 ng mL-1) with a limit of detection of 3.8 ng mL-1 and an IC50 of 903.9 ng mL-1. The intra and inter-assay coefficients of variation were 4.12-5.72% and 5.48-6.91%, respectively, and the recovery was acceptable. The applicability of the proposed assay was assessed by comparing the determined results with those measured by LC-MS/MS, presenting a satisfactory correlation (R 2 = 0.9868). The proposed assay, which shows satisfactory analytical performance, has great potential for application in the field of TDM in the future.
Collapse
Affiliation(s)
- Junyu Liang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Zhigao Zhang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Hui Zhao
- Department of Plastic and Aesthetic Surgery, Third Affiliated Hospital, Sun Yat-Sen University Guangzhou China
| | - Shanhe Wan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University Guangzhou China
| | - Xiangming Zhai
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Jianwei Zhou
- Guangzhou Darui Biotechnology Co. LTD Guangzhou China
| | - Rongliang Liang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Qiaoting Deng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Yingsong Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789355
| | - Guanfeng Lin
- Experimental Center of Teaching and Scientific Research, School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou China +86-20-37247604 +86-20-62789356
| |
Collapse
|
15
|
van Dijkman SC, Voskuyl RA, de Lange EC. Biomarkers in epilepsy-A modelling perspective. Eur J Pharm Sci 2017; 109S:S47-S52. [PMID: 28528284 DOI: 10.1016/j.ejps.2017.05.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 05/15/2017] [Indexed: 10/19/2022]
Abstract
Biomarkers can be categorised from type 0 (genotype or phenotype), through 6 (clinical scales), each level representing a part of the processes involved in the biological system and drug treatment. This classification facilitates the identification and connection of information required to fully (mathematically) model a disease and its treatment using integrated information from biomarkers. Two recent reviews thoroughly discussed the current status and development of biomarkers for epilepsy, but a path towards the integration of such biomarkers for the personalisation of anti-epileptic drug treatment is lacking. Here we aim to 1) briefly categorise the available epilepsy biomarkers and identify gaps, and 2) provide a modelling perspective on approaches to fill such gaps. There is mainly a lack of biomarker types 2 (target occupancy) and 3 (target activation). Current literature typically focuses on qualitative biomarkers for diagnosis and prediction of treatment response or failure, leaving a need for biomarkers that help to quantitatively understand the overall system to explain and predict differences in disease and treatment outcome. Due to the complexity of epilepsy, filling the biomarker gaps will require collaboration and expertise from the fields of systems biology and systems pharmacology.
Collapse
Affiliation(s)
- Sven C van Dijkman
- Division of Pharmacology, Leiden Academic Centre for Drug Research, The Netherlands.
| | - Rob A Voskuyl
- Division of Pharmacology, Leiden Academic Centre for Drug Research, The Netherlands
| | - Elizabeth C de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research, The Netherlands
| |
Collapse
|
16
|
van Dijkman SC, Rauwé WM, Danhof M, Della Pasqua O. Pharmacokinetic interactions and dosing rationale for antiepileptic drugs in adults and children. Br J Clin Pharmacol 2017; 84:97-111. [PMID: 28815754 DOI: 10.1111/bcp.13400] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 07/19/2017] [Accepted: 07/30/2017] [Indexed: 01/31/2023] Open
Abstract
AIMS Population pharmacokinetic modelling has been widely used across many therapeutic areas to identify sources of variability, which are incorporated into models as covariate factors. Despite numerous publications on pharmacokinetic drug-drug interactions (DDIs) between antiepileptic drugs (AEDs), such data are not used to support the dose rationale for polytherapy in the treatment of epileptic seizures. Here we assess the impact of DDIs on plasma concentrations and evaluate the need for AED dose adjustment. METHODS Models describing the pharmacokinetics of carbamazepine, clobazam, clonazepam, lamotrigine, levetiracetam, oxcarbazepine, phenobarbital, phenytoin, topiramate, valproic acid and zonisamide in adult and paediatric patients were collected from the published literature and implemented in NONMEM v7.2. Taking current clinical practice into account, we explore simulation scenarios to characterize AED exposure in virtual patients receiving mono- and polytherapy. Steady-state, maximum and minimum concentrations were selected as parameters of interest for this analysis. RESULTS Our simulations show that DDIs can cause major changes in AED concentrations both in adults and children. When more than one AED is used, even larger changes are observed in the concentrations of the primary drug, leading to significant differences in steady-state concentration between mono- and polytherapy for most AEDs. These results suggest that currently recommended dosing algorithms and titration procedures do not ensure attainment of appropriate therapeutic concentrations. CONCLUSIONS The effect of DDIs on AED exposure cannot be overlooked. Clinical guidelines must consider such covariate effects and ensure appropriate dosing recommendations for adult and paediatric patients who require combination therapy.
Collapse
Affiliation(s)
- Sven C van Dijkman
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Willem M Rauwé
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, UK.,Clinical Pharmacology & Therapeutics Group, University College London, London, UK
| |
Collapse
|