1
|
Sardu ML, Poggesi I. Pharmacokinetics of intranasal drugs, still a missed opportunity? Xenobiotica 2024; 54:424-438. [PMID: 38687903 DOI: 10.1080/00498254.2024.2349046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The intranasal (IN) route of administration is important for topical drugs and drugs intended to act systemically. More recently, direct nose-to-brain input was considered to bypass the blood-brain barrier.Processes related to IN absorption and nose-to-brain distribution are complex and depend, sometimes in contrasting ways, on chemico-physical and structural parameters of the compounds, and on formulation options.Due to the intricacies of these processes and despite the large number of articles published on many different IN compounds, it appears that absorption after IN dosing is not yet fully understood. In particular, at variance of the understanding and modelling approaches that are available for predicting the pharmacokinetics (PK) following oral administration of xenobiotics, it appears that there is not a similar understanding of the chemico-physical and structural determinants influencing drug absorption and disposition of compounds after IN administration, which represents a missed opportunity for this research field. This is even more true regarding the understanding of the direct nose-to-brain input. Due to this, IN administrations may represent an interesting and open research field for scientists aiming to develop PK property predictions tools, mechanistic PK models describing rate and extent of IN absorption, and translational tools to anticipate the clinical PK following IN dosing based on in vitro and in vivo non clinical experiments.This review intends to provide: i) some basic knowledge related to the physiology of PK after IN dosing, ii) a non-exhaustive list of preclinical and clinical examples related to compounds explored for the potential nose-to-blood and nose-to-brain passage, and iii) the identification of some areas requiring improvements, the understanding of which may facilitate the development of IN drug candidates.
Collapse
Affiliation(s)
| | - Italo Poggesi
- Clinical Pharmacology, Modeling and Simulation, GSK, Verona, Italy
| |
Collapse
|
2
|
Yang Y, Wang C, Chen Y, Wang X, Jiao Z, Wang Z. External evaluation and systematic review of population pharmacokinetic models for high-dose methotrexate in cancer patients. Eur J Pharm Sci 2023; 186:106416. [PMID: 37119861 DOI: 10.1016/j.ejps.2023.106416] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 05/01/2023]
Abstract
Several population pharmacokinetic (PPK) models have been established to optimize the therapeutic regimen and reduce the toxicity of high-dose methotrexate (HDMTX) in patients with cancer. However, their predictive performance when extrapolated to different clinical centers was unknown. In this study, we aimed to externally evaluate the predictive ability of HDMTX PPK models and determine the potential influencing factors. We searched the literature and determined the predictive performance of the selected models using methotrexate concentrations in 721 samples from 60 patients in the First Affiliated Hospital of the Navy Medical University. Prediction-based diagnostics and simulation-based normalized prediction distribution errors (NPDE) were used to evaluate the predictive performance of the models. The influence of prior information was also assessed using Bayesian forecasting, and the potential factors affecting model predictability were investigated. Thirty models extracted from published PPK studies were assessed. Prediction-based diagnostics showed that the number of compartments potentially influenced model transferability, and simulation-based NPDE indicated model misspecification. Bayesian forecasting significantly improved the predictive performance of the models. Various factors, including bioassays, covariates, and population diagnosis, influence model extrapolation. The published models were unsatisfactory for all prediction-based diagnostics, except for the 24 h methotrexate concentration monitoring and simulation-based diagnostics, making them inappropriate for direct extrapolation. Moreover, Bayesian forecasting combined therapeutic drug monitoring could improve the predictive performance of the models.
Collapse
Affiliation(s)
- Yunyun Yang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Pharmacy, Shanghai Changhai Hospital, First Affiliated Hospital of Navy Medical University, Shanghai 200433, China
| | - Chenyu Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yueting Chen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xuebin Wang
- Department of Pharmacy, Shanghai Changhai Hospital, First Affiliated Hospital of Navy Medical University, Shanghai 200433, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Zhuo Wang
- Department of Pharmacy, Shanghai Changhai Hospital, First Affiliated Hospital of Navy Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Chen EP, Bondi RW, Michalski PJ. Model-based Target Pharmacology Assessment (mTPA): An Approach Using PBPK/PD Modeling and Machine Learning to Design Medicinal Chemistry and DMPK Strategies in Early Drug Discovery. J Med Chem 2021; 64:3185-3196. [PMID: 33719432 DOI: 10.1021/acs.jmedchem.0c02033] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The optimal pharmacokinetic (PK) required for a drug candidate to elicit efficacy is highly dependent on the targeted pharmacology, a relationship that is often not well characterized during early phases of drug discovery. Generic assumptions around PK and potency risk misguiding screening and compound design toward nonoptimal absorption, distribution, metabolism, and excretion (ADME) or molecular properties and ultimately may increase attrition as well as hit-to-lead and lead optimization timelines. The present work introduces model-based target pharmacology assessment (mTPA), a computational approach combining physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling, sensitivity analysis, and machine learning (ML) to elucidate the optimal combination of PK, potency, and ADME specific for the targeted pharmacology. Examples using frequently encountered PK/PD relationships are presented to illustrate its application, and the utility and benefits of deploying such an approach to guide early discovery efforts are discussed.
Collapse
Affiliation(s)
- Emile P Chen
- Systems Modeling and Translational Biology, Computational Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Robert W Bondi
- Systems Modeling and Translational Biology, Computational Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Paul J Michalski
- Systems Modeling and Translational Biology, Computational Sciences, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
4
|
Chan Kwong AHXP, Calvier EAM, Fabre D, Gattacceca F, Khier S. Prior information for population pharmacokinetic and pharmacokinetic/pharmacodynamic analysis: overview and guidance with a focus on the NONMEM PRIOR subroutine. J Pharmacokinet Pharmacodyn 2020; 47:431-446. [PMID: 32535847 PMCID: PMC7520416 DOI: 10.1007/s10928-020-09695-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Abstract Population pharmacokinetic analysis is used to estimate pharmacokinetic parameters and their variability from concentration data. Due to data sparseness issues, available datasets often do not allow the estimation of all parameters of the suitable model. The PRIOR subroutine in NONMEM supports the estimation of some or all parameters with values from previous models, as an alternative to fixing them or adding data to the dataset. From a literature review, the best practices were compiled to provide a practical guidance for the use of the PRIOR subroutine in NONMEM. Thirty-three articles reported the use of the PRIOR subroutine in NONMEM, mostly in special populations. This approach allowed fast, stable and satisfying modelling. The guidance provides general advice on how to select the most appropriate reference model when there are several previous models available, and to implement and weight the selected parameter values in the PRIOR function. On the model built with PRIOR, the similarity of estimates with the ones of the reference model and the sensitivity of the model to the PRIOR values should be checked. Covariates could be implemented a priori (from the reference model) or a posteriori, only on parameters estimated without prior (search for new covariates). Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s10928-020-09695-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna H-X P Chan Kwong
- Pharmacokinetic and Modeling Department, School of Pharmacy, Montpellier University, Montpellier, France.
- Probabilities and Statistics Department, Institut Montpelliérain Alexander Grothendieck (IMAG), UMR 5149, CNRS, Montpellier University, Montpellier, France.
- SMARTc group, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France.
- Pharmacokinetics-Dynamics and Metabolism (PKDM), Sanofi R&D, Translational Medicine and Early Development, Montpellier, France.
| | - Elisa A M Calvier
- Pharmacokinetics-Dynamics and Metabolism (PKDM), Sanofi R&D, Translational Medicine and Early Development, Montpellier, France
| | - David Fabre
- Pharmacokinetics-Dynamics and Metabolism (PKDM), Sanofi R&D, Translational Medicine and Early Development, Montpellier, France
| | - Florence Gattacceca
- SMARTc group, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Sonia Khier
- Pharmacokinetic and Modeling Department, School of Pharmacy, Montpellier University, Montpellier, France
- Probabilities and Statistics Department, Institut Montpelliérain Alexander Grothendieck (IMAG), UMR 5149, CNRS, Montpellier University, Montpellier, France
| |
Collapse
|
5
|
van Esdonk MJ, Burggraaf J, Dehez M, van der Graaf PH, Stevens J. Quantification of the endogenous growth hormone and prolactin lowering effects of a somatostatin-dopamine chimera using population PK/PD modeling. J Pharmacokinet Pharmacodyn 2020; 47:229-239. [PMID: 32248329 PMCID: PMC7289785 DOI: 10.1007/s10928-020-09683-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/26/2020] [Indexed: 12/02/2022]
Abstract
A phase 1 clinical trial in healthy male volunteers was conducted with a somatostatin-dopamine chimera (BIM23B065), from which information could be obtained on the concentration-effect relationship of the inhibition of pulsatile endogenous growth hormone and prolactin secretion. Endogenous growth hormone profiles were analyzed using a two-step deconvolution-analysis-informed population pharmacodynamic modeling approach, which was developed for the analyses of pulsatile profiles. Prolactin concentrations were modelled using a population pool model with a circadian component on the prolactin release. During treatment with BIM23B065, growth hormone secretion was significantly reduced (maximal effect [EMAX] = − 64.8%) with significant reductions in the pulse frequency in two out of three multiple ascending dose cohorts. A circadian component in prolactin secretion was identified, modelled using a combination of two cosine functions with 24 h and 12 h periods. Dosing of BIM23B065 strongly inhibited (EMAX = − 91%) the prolactin release and demonstrated further reduction of prolactin secretion after multiple days of dosing. This study quantified the concentration-effect relationship of BIM23B065 on the release of two pituitary hormones, providing proof of pharmacology of the chimeric actions of BIM23B065.
Collapse
Affiliation(s)
- Michiel J van Esdonk
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands. .,Centre for Human Drug Research, Leiden, The Netherlands.
| | - Jacobus Burggraaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Certara QSP, Canterbury, UK
| | - Jasper Stevens
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
6
|
van den Brink WJ, van den Berg D, Bonsel FEM, Hartman R, Wong Y, van der Graaf PH, de Lange ECM. Fingerprints of CNS drug effects: a plasma neuroendocrine reflection of D 2 receptor activation using multi-biomarker pharmacokinetic/pharmacodynamic modelling. Br J Pharmacol 2018; 175:3832-3843. [PMID: 30051461 PMCID: PMC6135786 DOI: 10.1111/bph.14452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 07/06/2018] [Accepted: 07/11/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Because biological systems behave as networks, multi-biomarker approaches increasingly replace single biomarker approaches in drug development. To improve the mechanistic insights into CNS drug effects, a plasma neuroendocrine fingerprint was identified using multi-biomarker pharmacokinetic/pharmacodynamic (PK/PD) modelling. Short- and long-term D2 receptor activation was evaluated using quinpirole as a paradigm compound. EXPERIMENTAL APPROACH Rats received 0, 0.17 or 0.86 mg·kg-1 of the D2 agonist quinpirole i.v. Quinpirole concentrations in plasma and brain extracellular fluid (brainECF ), as well as plasma concentrations of 13 hormones and neuropeptides, were measured. Experiments were performed at day 1 and repeated after 7-day s.c. drug administration. PK/PD modelling was applied to identify the in vivo concentration-effect relations and neuroendocrine dynamics. KEY RESULTS The quinpirole pharmacokinetics were adequately described by a two-compartment model with an unbound brainECF -to-plasma concentration ratio of 5. The release of adenocorticotropic hormone (ACTH), growth hormone, prolactin and thyroid-stimulating hormone (TSH) from the pituitary was influenced. Except for ACTH, D2 receptor expression levels on the pituitary hormone-releasing cells predicted the concentration-effect relationship differences. Baseline levels (ACTH, prolactin, TSH), hormone release (ACTH) and potency (TSH) changed with treatment duration. CONCLUSIONS AND IMPLICATIONS The integrated multi-biomarker PK/PD approach revealed a fingerprint reflecting D2 receptor activation. This forms the conceptual basis for in vivo evaluation of on- and off-target CNS drug effects. The effect of treatment duration is highly relevant given the long-term use of D2 agonists in clinical practice. Further development towards quantitative systems pharmacology models will eventually facilitate mechanistic drug development.
Collapse
Affiliation(s)
- Willem J van den Brink
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Dirk‐Jan van den Berg
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Floor E M Bonsel
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Robin Hartman
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Yin‐Cheong Wong
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
- Certara QSP, Canterbury Innovation HouseCanterburyUK
| | - Elizabeth C M de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
7
|
Min J, Chen H, Gong Z, Liu X, Wu T, Li W, Fang J, Huang T, Zhang Y, Zhao W, Zhu C, Wang Q, Mi S, Wang N. Pharmacokinetic and Pharmacodynamic Properties of Rosmarinic Acid in Rat Cholestatic Liver Injury. Molecules 2018; 23:E2287. [PMID: 30205454 PMCID: PMC6225135 DOI: 10.3390/molecules23092287] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/18/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
The objective of this study was to evaluate the hepatoprotective and metabolic effects of rosmarinic acid (RA) in rats. RA [100 mg/kg body weight (BW)] was intragastrically (i.g.) administered to Sprague-Dawley (SD) rats once a day for seven consecutive days. The rats were then i.g. administered α-naphthylisothiocyanate (ANIT) (80 mg/kg once on the 5th day) to induce acute intrahepatic cholestasis after the last administration of RA. Blood samples were collected at different time points (0.083 h, 0.17 h, 0.33 h, 0.5 h, 0.75 h, 1 h, 1.5 h, 3 h, 4 h, 6 h, 8 h, 12 h, 20 h) after administration, and the levels of RA were estimated by HPLC. Plasma and bile biochemical analysis, bile flow rate, and liver histopathology were measured to evaluate the hepatoprotective effect of RA. The PK-PD curves showed obviously clockwise (AST and ALT) or anticlockwise (TBA, TBIL). Pretreatment with RA at different doses significantly restrained ANIT-induced pathological changes in bile rate, TBA, TBIL, ALT, AST (p < 0.05 or p < 0.01). The relationship between RA concentration and its hepatoprotective effects on acute cholestasis responses was assessed by PK-PD modeling.
Collapse
Affiliation(s)
- Jianbin Min
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Hao Chen
- College of Food and Drug, Anhui Science and Technology of University, Fengyang 233100, Anhui, China.
| | - Zipeng Gong
- Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Beijing Road, Guiyang 550004, China.
| | - Xian Liu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Tian Wu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Weirong Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Jiansong Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Tianlai Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Yingfeng Zhang
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Wei Zhao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Chenchen Zhu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Suiqing Mi
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Ningsheng Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| |
Collapse
|
8
|
de Witte WEA, Rottschäfer V, Danhof M, van der Graaf PH, Peletier LA, de Lange ECM. Modelling the delay between pharmacokinetics and EEG effects of morphine in rats: binding kinetic versus effect compartment models. J Pharmacokinet Pharmacodyn 2018; 45:621-635. [PMID: 29777407 PMCID: PMC6061075 DOI: 10.1007/s10928-018-9593-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 05/02/2018] [Indexed: 01/10/2023]
Abstract
Drug–target binding kinetics (as determined by association and dissociation rate constants, kon and koff) can be an important determinant of the kinetics of drug action. However, the effect compartment model is used most frequently instead of a target binding model to describe hysteresis. Here we investigate when the drug–target binding model should be used in lieu of the effect compartment model. The utility of the effect compartment (EC), the target binding kinetics (TB) and the combined effect compartment–target binding kinetics (EC–TB) model were tested on either plasma (ECPL, TBPL and EC–TBPL) or brain extracellular fluid (ECF) (ECECF, TBECF and EC–TBECF) morphine concentrations and EEG amplitude in rats. It was also analyzed when a significant shift in the time to maximal target occupancy (TmaxTO) with increasing dose, the discriminating feature between the TB and EC model, occurs in the TB model. All TB models assumed a linear relationship between target occupancy and drug effect on the EEG amplitude. All three model types performed similarly in describing the morphine pharmacodynamics data, although the EC model provided the best statistical result. The analysis of the shift in TmaxTO (∆TmaxTO) as a result of increasing dose revealed that ∆TmaxTO is decreasing towards zero if the koff is much smaller than the elimination rate constant or if the target concentration is larger than the initial morphine concentration. The results for the morphine PKPD modelling and the analysis of ∆TmaxTO indicate that the EC and TB models do not necessarily lead to different drug effect versus time curves for different doses if a delay between drug concentrations and drug effect (hysteresis) is described. Drawing mechanistic conclusions from successfully fitting one of these two models should therefore be avoided. Since the TB model can be informed by in vitro measurements of kon and koff, a target binding model should be considered more often for mechanistic modelling purposes.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, 2333 CA, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands
- Certara Quantitative Systems Pharmacology, Canterbury Innovation Centre, Canterbury, CT2 7FG, UK
| | | | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
9
|
van den Brink WJ, Hankemeier T, van der Graaf PH, de Lange ECM. Bundling arrows: improving translational CNS drug development by integrated PK/PD-metabolomics. Expert Opin Drug Discov 2018. [DOI: 10.1080/17460441.2018.1446935] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- W. J. van den Brink
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - T. Hankemeier
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - P. H. van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Certara QSP, Canterbury Innovation House, Canterbury, United Kingdom
| | - E. C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
10
|
van den Brink WJ, Palic S, Köhler I, de Lange ECM. Access to the CNS: Biomarker Strategies for Dopaminergic Treatments. Pharm Res 2018; 35:64. [PMID: 29450650 PMCID: PMC5814527 DOI: 10.1007/s11095-017-2333-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/18/2017] [Indexed: 12/26/2022]
Abstract
Despite substantial research carried out over the last decades, it remains difficult to understand the wide range of pharmacological effects of dopaminergic agents. The dopaminergic system is involved in several neurological disorders, such as Parkinson's disease and schizophrenia. This complex system features multiple pathways implicated in emotion and cognition, psychomotor functions and endocrine control through activation of G protein-coupled dopamine receptors. This review focuses on the system-wide effects of dopaminergic agents on the multiple biochemical and endocrine pathways, in particular the biomarkers (i.e., indicators of a pharmacological process) that reflect these effects. Dopaminergic treatments developed over the last decades were found to be associated with numerous biochemical pathways in the brain, including the norepinephrine and the kynurenine pathway. Additionally, they have shown to affect peripheral systems, for example the hypothalamus-pituitary-adrenal (HPA) axis. Dopaminergic agents thus have a complex and broad pharmacological profile, rendering drug development challenging. Considering the complex system-wide pharmacological profile of dopaminergic agents, this review underlines the needs for systems pharmacology studies that include: i) proteomics and metabolomics analysis; ii) longitudinal data evaluation and mathematical modeling; iii) pharmacokinetics-based interpretation of drug effects; iv) simultaneous biomarker evaluation in the brain, the cerebrospinal fluid (CSF) and plasma; and v) specific attention to condition-dependent (e.g., disease) pharmacology. Such approach is considered essential to increase our understanding of central nervous system (CNS) drug effects and substantially improve CNS drug development.
Collapse
Affiliation(s)
- Willem Johan van den Brink
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Semra Palic
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Isabelle Köhler
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth Cunera Maria de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
11
|
Taneja A, Vermeulen A, Huntjens DRH, Danhof M, De Lange ECM, Proost JH. Modeling of prolactin response following dopamine D 2 receptor antagonists in rats: can it be translated to clinical dosing? Pharmacol Res Perspect 2017; 5. [PMID: 29226628 PMCID: PMC5723698 DOI: 10.1002/prp2.364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 01/23/2023] Open
Abstract
Prolactin release is a side effect of antipsychotic therapy with dopamine antagonists, observed in rats as well as humans. We examined whether two semimechanistic models could describe prolactin response in rats and subsequently be translated to predict pituitary dopamine D2 receptor occupancy and plasma prolactin concentrations in humans following administration of paliperidone or remoxipride. Data on male Wistar rats receiving single or multiple doses of risperidone, paliperidone, or remoxipride was described by two semimechanistic models, the precursor pool model and the agonist–antagonist interaction model. Using interspecies scaling approaches, human D2 receptor occupancy and plasma prolactin concentrations were predicted for a range of clinical paliperidone and remoxipride doses. The predictions were compared with corresponding observations described in literature as well as with predictions from published models developed on human data. The pool model could predict D2 receptor occupancy and prolactin response in humans following single doses of paliperidone and remoxipride. Tolerance of prolactin release was predicted following multiple doses. The interaction model underpredicted both D2 receptor occupancy and prolactin response. Prolactin elevation may be deployed as a suitable biomarker for interspecies translation and can inform the clinical safe and effective dose range of antipsychotic drugs. While the pool model was more predictive than the interaction model, it overpredicted tolerance on multiple dosing. Shortcomings of the translations reflect the need for better mechanistic models.
Collapse
Affiliation(s)
- Amit Taneja
- Division of Pharmacokinetics, Toxicology and TargetingGroningen Research Institute of PharmacyUniversity of GroningenAntonius Deusinglaan 19713 AVGroningenThe Netherlands
| | - An Vermeulen
- Division of Janssen Pharmaceutica NVClinical Pharmacology and PharmacometricsJanssen Research and DevelopmentBeerseBelgium
| | - Dymphy R. H. Huntjens
- Division of Janssen Pharmaceutica NVClinical Pharmacology and PharmacometricsJanssen Research and DevelopmentBeerseBelgium
| | - Meindert Danhof
- Department of PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Elizabeth C. M. De Lange
- Department of PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Johannes H. Proost
- Division of Pharmacokinetics, Toxicology and TargetingGroningen Research Institute of PharmacyUniversity of GroningenAntonius Deusinglaan 19713 AVGroningenThe Netherlands
| |
Collapse
|
12
|
Multivariate pharmacokinetic/pharmacodynamic (PKPD) analysis with metabolomics shows multiple effects of remoxipride in rats. Eur J Pharm Sci 2017; 109:431-440. [DOI: 10.1016/j.ejps.2017.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/12/2023]
|
13
|
de Lange ECM, van den Brink W, Yamamoto Y, de Witte WEA, Wong YC. Novel CNS drug discovery and development approach: model-based integration to predict neuro-pharmacokinetics and pharmacodynamics. Expert Opin Drug Discov 2017; 12:1207-1218. [PMID: 28933618 DOI: 10.1080/17460441.2017.1380623] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION CNS drug development has been hampered by inadequate consideration of CNS pharmacokinetic (PK), pharmacodynamics (PD) and disease complexity (reductionist approach). Improvement is required via integrative model-based approaches. Areas covered: The authors summarize factors that have played a role in the high attrition rate of CNS compounds. Recent advances in CNS research and drug discovery are presented, especially with regard to assessment of relevant neuro-PK parameters. Suggestions for further improvements are also discussed. Expert opinion: Understanding time- and condition dependent interrelationships between neuro-PK and neuro-PD processes is key to predictions in different conditions. As a first screen, it is suggested to use in silico/in vitro derived molecular properties of candidate compounds and predict concentration-time profiles of compounds in multiple compartments of the human CNS, using time-course based physiology-based (PB) PK models. Then, for selected compounds, one can include in vitro drug-target binding kinetics to predict target occupancy (TO)-time profiles in humans. This will improve neuro-PD prediction. Furthermore, a pharmaco-omics approach is suggested, providing multilevel and paralleled data on systems processes from individuals in a systems-wide manner. Thus, clinical trials will be better informed, using fewer animals, while also, needing fewer individuals and samples per individual for proof of concept in humans.
Collapse
Affiliation(s)
- Elizabeth C M de Lange
- a Leiden Academic Center of Drug Research, Translational Pharmacology , Leiden University , Leiden , The Netherlands
| | - Willem van den Brink
- a Leiden Academic Center of Drug Research, Translational Pharmacology , Leiden University , Leiden , The Netherlands
| | - Yumi Yamamoto
- a Leiden Academic Center of Drug Research, Translational Pharmacology , Leiden University , Leiden , The Netherlands
| | - Wilhelmus E A de Witte
- a Leiden Academic Center of Drug Research, Translational Pharmacology , Leiden University , Leiden , The Netherlands
| | - Yin Cheong Wong
- a Leiden Academic Center of Drug Research, Translational Pharmacology , Leiden University , Leiden , The Netherlands
| |
Collapse
|
14
|
Peletier LA, Gabrielsson J. Impact of mathematical pharmacology on practice and theory: four case studies. J Pharmacokinet Pharmacodyn 2017; 45:3-21. [PMID: 28884259 PMCID: PMC5847232 DOI: 10.1007/s10928-017-9539-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/18/2017] [Indexed: 11/25/2022]
Abstract
Drug-discovery has become a complex discipline in which the amount of knowledge about human biology, physiology, and biochemistry have increased. In order to harness this complex body of knowledge mathematics can play a critical role, and has actually already been doing so. We demonstrate through four case studies, taken from previously published data and analyses, what we can gain from mathematical/analytical techniques when nonlinear concentration-time courses have to be transformed into their equilibrium concentration-response (target or complex) relationships and new structures of drug potency have to be deciphered; when pattern recognition needs to be carried out for an unconventional response-time dataset; when what-if? predictions beyond the observational concentration-time range need to be made; or when the behaviour of a semi-mechanistic model needs to be elucidated or challenged. These four examples are typical situations when standard approaches known to the general community of pharmacokineticists prove to be inadequate.
Collapse
Affiliation(s)
| | - Johan Gabrielsson
- Division of Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 750 07 Uppsala, Sweden
| |
Collapse
|
15
|
Lott D, Krause A, Seemayer CA, Strasser DS, Dingemanse J, Lehr T. Modeling the Effect of the Selective S1P1 Receptor Modulator Ponesimod on Subsets of Blood Lymphocytes. Pharm Res 2016; 34:599-609. [DOI: 10.1007/s11095-016-2087-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/08/2016] [Indexed: 01/21/2023]
|
16
|
Yamamoto Y, Välitalo PA, van den Berg DJ, Hartman R, van den Brink W, Wong YC, Huntjens DR, Proost JH, Vermeulen A, Krauwinkel W, Bakshi S, Aranzana-Climent V, Marchand S, Dahyot-Fizelier C, Couet W, Danhof M, van Hasselt JGC, de Lange ECM. A Generic Multi-Compartmental CNS Distribution Model Structure for 9 Drugs Allows Prediction of Human Brain Target Site Concentrations. Pharm Res 2016; 34:333-351. [PMID: 27864744 PMCID: PMC5236087 DOI: 10.1007/s11095-016-2065-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022]
Abstract
Purpose Predicting target site drug concentration in the brain is of key importance for the successful development of drugs acting on the central nervous system. We propose a generic mathematical model to describe the pharmacokinetics in brain compartments, and apply this model to predict human brain disposition. Methods A mathematical model consisting of several physiological brain compartments in the rat was developed using rich concentration-time profiles from nine structurally diverse drugs in plasma, brain extracellular fluid, and two cerebrospinal fluid compartments. The effect of active drug transporters was also accounted for. Subsequently, the model was translated to predict human concentration-time profiles for acetaminophen and morphine, by scaling or replacing system- and drug-specific parameters in the model. Results A common model structure was identified that adequately described the rat pharmacokinetic profiles for each of the nine drugs across brain compartments, with good precision of structural model parameters (relative standard error <37.5%). The model predicted the human concentration-time profiles in different brain compartments well (symmetric mean absolute percentage error <90%). Conclusions A multi-compartmental brain pharmacokinetic model was developed and its structure could adequately describe data across nine different drugs. The model could be successfully translated to predict human brain concentrations. Electronic supplementary material The online version of this article (doi:10.1007/s11095-016-2065-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Pyry A Välitalo
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Dirk-Jan van den Berg
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Robin Hartman
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Willem van den Brink
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Yin Cheong Wong
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Dymphy R Huntjens
- Quantitative Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Johannes H Proost
- Division of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands
| | - An Vermeulen
- Quantitative Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Walter Krauwinkel
- Department of Clinical Pharmacology & Exploratory Development, Astellas Pharma BV, Leiden, The Netherlands
| | - Suruchi Bakshi
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Sandrine Marchand
- Department of Medicine and Pharmacy, University of Poitiers, Poitiers, France
| | - Claire Dahyot-Fizelier
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Center of Poitiers, Poitiers, France
| | - William Couet
- Department of Medicine and Pharmacy, University of Poitiers, Poitiers, France
| | - Meindert Danhof
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Johan G C van Hasselt
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
- Leiden University Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, The Netherlands.
| |
Collapse
|
17
|
Shimizu S, den Hoedt SM, Mangas-Sanjuan V, Cristea S, Geuer JK, van den Berg DJ, Hartman R, Bellanti F, de Lange ECM. Target-Site Investigation for the Plasma Prolactin Response: Mechanism-Based Pharmacokinetic-Pharmacodynamic Analysis of Risperidone and Paliperidone in the Rat. Drug Metab Dispos 2016; 45:152-159. [PMID: 27836941 DOI: 10.1124/dmd.116.072306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022] Open
Abstract
To understand the drivers in the biological system response to dopamine D2 receptor antagonists, a mechanistic semiphysiologically based (PB) pharmacokinetic-pharmacodymanic (PKPD) model was developed to describe prolactin responses to risperidone (RIS) and its active metabolite paliperidone (PAL). We performed a microdialysis study in rats to obtain detailed plasma, brain extracellular fluid (ECF), and cerebrospinal fluid (CSF) concentrations of PAL and RIS. To assess the impact of P-glycoprotein (P-gp) functioning on brain distribution, we performed experiments in the absence or presence of the P-gp inhibitor tariquidar (TQD). PK and PKPD modeling was performed by nonlinear mixed-effect modeling. Plasma, brain ECF, and CSF PK values of RIS and PAL were well described by a 12-compartmental semi-PBPK model, including metabolic conversion of RIS to PAL. P-gp efflux functionality was identified on brain ECF for RIS and PAL and on CSF only for PAL. In the PKPD analysis, the plasma drug concentrations were more relevant than brain ECF or CSF concentrations to explain the prolactin response; the estimated EC50 was in accordance with reports in the literature for both RIS and PAL. We conclude that for RIS and PAL, the plasma concentrations better explain the prolactin response than do brain ECF or CSF concentrations. This research shows that PKPD modeling is of high value to delineate the target site of drugs.
Collapse
Affiliation(s)
- Shinji Shimizu
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Sandra M den Hoedt
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Victor Mangas-Sanjuan
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Sinziana Cristea
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Jana K Geuer
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Dirk-Jan van den Berg
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Robin Hartman
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Francisco Bellanti
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, The Netherlands
| |
Collapse
|
18
|
van den Brink WJ, Wong YC, Gülave B, van der Graaf PH, de Lange ECM. Revealing the Neuroendocrine Response After Remoxipride Treatment Using Multi-Biomarker Discovery and Quantifying It by PK/PD Modeling. AAPS JOURNAL 2016; 19:274-285. [PMID: 27785749 DOI: 10.1208/s12248-016-0002-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/03/2016] [Indexed: 01/10/2023]
Abstract
To reveal unknown and potentially important mechanisms of drug action, multi-biomarker discovery approaches are increasingly used. Time-course relationships between drug action and multi-biomarker profiles, however, are typically missing, while such relationships will provide increased insight in the underlying body processes. The aim of this study was to investigate the effect of the dopamine D2 antagonist remoxipride on the neuroendocrine system. Different doses of remoxipride (0, 0.7, 5.2, or 14 mg/kg) were administered to rats by intravenous infusion. Serial brain extracellular fluid (brainECF) and plasma samples were collected and analyzed for remoxipride pharmacokinetics (PK). Plasma samples were analyzed for concentrations of the eight pituitary-related hormones as a function of time. A Mann-Whitney test was used to identify the responding hormones, which were further analyzed by pharmacokinetic/pharmacodynamic (PK/PD) modeling. A three-compartment PK model adequately described remoxipride PK in plasma and brainECF. Not only plasma PRL, but also adrenocorticotrophic hormone (ACTH) concentrations were increased, the latter especially at higher concentrations of remoxipride. Brain-derived neurotropic factor (BDNF), follicle stimulating hormone (FSH), growth hormone (GH), luteinizing hormone (LH), and thyroid stimulating hormones (TSH) did not respond to remoxipride at the tested doses, while oxytocin (OXT) measurements were below limit of quantification. Precursor pool models were linked to brainECF remoxipride PK by Emax drug effect models, which could accurately describe the PRL and ACTH responses. To conclude, this study shows how a multi-biomarker identification approach combined with PK/PD modeling can reveal and quantify a neuroendocrine multi-biomarker response for single drug action.
Collapse
Affiliation(s)
- Willem J van den Brink
- Systems Pharmacology, Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands
| | - Yin C Wong
- Systems Pharmacology, Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands
| | - Berfin Gülave
- Systems Pharmacology, Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands
| | - Piet H van der Graaf
- Systems Pharmacology, Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands.,Certara QSP, Canterbury Innovation House, Canterbury, UK
| | - Elizatbeth C M de Lange
- Systems Pharmacology, Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands.
| |
Collapse
|
19
|
Intracerebral microdialysis in blood-brain barrier drug research with focus on nanodelivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 20:13-18. [PMID: 27986218 DOI: 10.1016/j.ddtec.2016.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/13/2016] [Indexed: 01/09/2023]
Abstract
Microdialysis has contributed significantly to advance the understanding of BBB transport of drugs and to reveal key aspects of BBB transport, including quantifying active efflux and active uptake. Microdialysis studies on pharmacokinetic-pharmacodynamic relationships have given in-depth understanding of the processes involved. Recently, nanodelivery to the brain has been investigated with microdialysis, contributing to nanodelivery science by giving quantitative information on the possible success of different delivery vehicles and how they are involved in BBB transport.
Collapse
|
20
|
Taneja A, Vermeulen A, Huntjens DRH, Danhof M, De Lange ECM, Proost JH. Summary data of potency and parameter information from semi-mechanistic PKPD modeling of prolactin release following administration of the dopamine D2 receptor antagonists risperidone, paliperidone and remoxipride in rats. Data Brief 2016; 8:1433-7. [PMID: 27617278 PMCID: PMC5007417 DOI: 10.1016/j.dib.2016.07.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/23/2016] [Accepted: 07/28/2016] [Indexed: 11/25/2022] Open
Abstract
We provide the reader with relevant data related to our recently published paper, comparing two mathematical models to describe prolactin turnover in rats following one or two doses of the dopamine D2 receptor antagonists risperidone, paliperidone and remoxipride, "A comparison of two semi-mechanistic models for prolactin release and prediction of receptor occupancy following administration of dopamine D2 receptor antagonists in rats" (Taneja et al., 2016) [1]. All information is tabulated. Summary level data on the in vitro potencies and the physicochemical properties is presented in Table 1. Model parameters required to explore the precursor pool model are presented in Table 2. In Table 3, estimated parameter comparisons for both models are presented, when separate potencies are estimated for risperidone and paliperidone, as compared to a common potency for both drugs. In Table 4, parameter estimates are compared when the drug effect is parameterized in terms of drug concentration or receptor occupancy.
Collapse
Affiliation(s)
- Amit Taneja
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - An Vermeulen
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Dymphy R H Huntjens
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Meindert Danhof
- Department of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Elizabeth C M De Lange
- Department of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Johannes H Proost
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
21
|
Bakshi S, de Lange EC, van der Graaf PH, Danhof M, Peletier LA. Understanding the Behavior of Systems Pharmacology Models Using Mathematical Analysis of Differential Equations: Prolactin Modeling as a Case Study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:339-51. [PMID: 27405001 PMCID: PMC4961077 DOI: 10.1002/psp4.12098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/21/2016] [Accepted: 05/19/2016] [Indexed: 01/20/2023]
Abstract
In this tutorial, we introduce basic concepts in dynamical systems analysis, such as phase‐planes, stability, and bifurcation theory, useful for dissecting the behavior of complex and nonlinear models. A precursor‐pool model with positive feedback is used to demonstrate the power of mathematical analysis. This model is nonlinear and exhibits multiple steady states, the stability of which is analyzed. The analysis offers insight into model behavior and suggests useful parameter regions, which simulations alone could not.
Collapse
Affiliation(s)
- S Bakshi
- Systems Pharmacology, Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | - E C de Lange
- Systems Pharmacology, Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | - P H van der Graaf
- Systems Pharmacology, Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands.,Certara QSP, Canterbury Innovation House, Canterbury, United Kingdom
| | - M Danhof
- Systems Pharmacology, Division of Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | - L A Peletier
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
22
|
Taneja A, Vermeulen A, Huntjens DRH, Danhof M, De Lange ECM, Proost JH. A comparison of two semi-mechanistic models for prolactin release and prediction of receptor occupancy following administration of dopamine D2 receptor antagonists in rats. Eur J Pharmacol 2016; 789:202-214. [PMID: 27395799 DOI: 10.1016/j.ejphar.2016.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 01/11/2023]
Abstract
We compared the model performance of two semi-mechanistic pharmacokinetic-pharmacodynamic models, the precursor pool model and the agonist-antagonist interaction model, to describe prolactin response following the administration of the dopamine D2 receptor antagonists risperidone, paliperidone or remoxipride in rats. The time course of pituitary dopamine D2 receptor occupancy was also predicted. Male Wistar rats received a single dose (risperidone, paliperidone, remoxipride) or two consecutive doses (remoxipride). Population modeling was applied to fit the pool and interaction models to the prolactin data. The pool model was modified to predict the time course of pituitary D2 receptor occupancy. Unbound plasma concentrations of the D2 receptor antagonists were considered the drivers of the prolactin response. Both models were used to predict prolactin release following multiple doses of paliperidone. Both models described the data well and model performance was comparable. Estimated unbound EC50 for risperidone and paliperidone was 35.1nM (relative standard error 51%) and for remoxipride it was 94.8nM (31%). KI values for these compounds were 11.1nM (21%) and 113nM (27%), respectively. Estimated pituitary D2 receptor occupancies for risperidone and remoxipride were comparable to literature findings. The interaction model better predicted prolactin profiles following multiple paliperidone doses, while the pool model predicted tolerance better. The performance of both models in describing the prolactin profiles was comparable. The pool model could additionally describe the time course of pituitary D2 receptor occupancy. Prolactin response following multiple paliperidone doses was better predicted by the interaction model.
Collapse
Affiliation(s)
- Amit Taneja
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - An Vermeulen
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Dymphy R H Huntjens
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Meindert Danhof
- Department of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Elizabeth C M De Lange
- Department of Pharmacology, Leiden Academic Center for Drug Research, Leiden University, The Netherlands
| | - Johannes H Proost
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
23
|
Danhof M. Systems pharmacology - Towards the modeling of network interactions. Eur J Pharm Sci 2016; 94:4-14. [PMID: 27131606 DOI: 10.1016/j.ejps.2016.04.027] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/21/2016] [Accepted: 04/24/2016] [Indexed: 12/13/2022]
Abstract
Mechanism-based pharmacokinetic and pharmacodynamics (PKPD) and disease system (DS) models have been introduced in drug discovery and development research, to predict in a quantitative manner the effect of drug treatment in vivo in health and disease. This requires consideration of several fundamental properties of biological systems behavior including: hysteresis, non-linearity, variability, interdependency, convergence, resilience, and multi-stationarity. Classical physiology-based PKPD models consider linear transduction pathways, connecting processes on the causal path between drug administration and effect, as the basis of drug action. Depending on the drug and its biological target, such models may contain expressions to characterize i) the disposition and the target site distribution kinetics of the drug under investigation, ii) the kinetics of target binding and activation and iii) the kinetics of transduction. When connected to physiology-based DS models, PKPD models can characterize the effect on disease progression in a mechanistic manner. These models have been found useful to characterize hysteresis and non-linearity, yet they fail to explain the effects of the other fundamental properties of biological systems behavior. Recently systems pharmacology has been introduced as novel approach to predict in vivo drug effects, in which biological networks rather than single transduction pathways are considered as the basis of drug action and disease progression. These models contain expressions to characterize the functional interactions within a biological network. Such interactions are relevant when drugs act at multiple targets in the network or when homeostatic feedback mechanisms are operative. As a result systems pharmacology models are particularly useful to describe complex patterns of drug action (i.e. synergy, oscillatory behavior) and disease progression (i.e. episodic disorders). In this contribution it is shown how physiology-based PKPD and disease models can be extended to account for internal systems interactions. It is demonstrated how SP models can be used to predict the effects of multi-target interactions and of homeostatic feedback on the pharmacological response. In addition it is shown how DS models may be used to distinguish symptomatic from disease modifying effects and to predict the long term effects on disease progression, from short term biomarker responses. It is concluded that incorporation of expressions to describe the interactions in biological network analysis opens new avenues to the understanding of the effects of drug treatment on the fundamental aspects of biological systems behavior.
Collapse
Affiliation(s)
- Meindert Danhof
- Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
24
|
Danhof M. Kinetics of drug action in disease states: towards physiology-based pharmacodynamic (PBPD) models. J Pharmacokinet Pharmacodyn 2015; 42:447-62. [PMID: 26319673 PMCID: PMC4582079 DOI: 10.1007/s10928-015-9437-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/17/2015] [Indexed: 11/26/2022]
Abstract
Gerhard Levy started his investigations on the "Kinetics of Drug Action in Disease States" in the fall of 1980. The objective of his research was to study inter-individual variation in pharmacodynamics. To this end, theoretical concepts and experimental approaches were introduced, which enabled assessment of the changes in pharmacodynamics per se, while excluding or accounting for the cofounding effects of concomitant changes in pharmacokinetics. These concepts were applied in several studies. The results, which were published in 45 papers in the years 1984-1994, showed considerable variation in pharmacodynamics. These initial studies on kinetics of drug action in disease states triggered further experimental research on the relations between pharmacokinetics and pharmacodynamics. Together with the concepts in Levy's earlier publications "Kinetics of Pharmacologic Effects" (Clin Pharmacol Ther 7(3): 362-372, 1966) and "Kinetics of pharmacologic effects in man: the anticoagulant action of warfarin" (Clin Pharmacol Ther 10(1): 22-35, 1969), they form a significant impulse to the development of physiology-based pharmacodynamic (PBPD) modeling as novel discipline in the pharmaceutical sciences. This paper reviews Levy's research on the "Kinetics of Drug Action in Disease States". Next it addresses the significance of his research for the evolution of PBPD modeling as a scientific discipline. PBPD models contain specific expressions to characterize in a strictly quantitative manner processes on the causal path between exposure (in terms of concentration at the target site) and the drug effect (in terms of the change in biological function). Pertinent processes on the causal path are: (1) target site distribution, (2) target binding and activation and (3) transduction and homeostatic feedback.
Collapse
Affiliation(s)
- Meindert Danhof
- Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands.
| |
Collapse
|
25
|
de Lange ECM, Hammarlund-Udenaes M. Translational aspects of blood-brain barrier transport and central nervous system effects of drugs: From discovery to patients. Clin Pharmacol Ther 2015; 97:380-94. [DOI: 10.1002/cpt.76] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/06/2015] [Accepted: 01/06/2015] [Indexed: 02/06/2023]
Affiliation(s)
- ECM de Lange
- Leiden Academic Centre for Drug Research; Division of Pharmacology; Leiden University, Gorlaeus Laboratories; Leiden The Netherlands
| | | |
Collapse
|
26
|
Ruigrok MJR, de Lange ECM. Emerging Insights for Translational Pharmacokinetic and Pharmacokinetic-Pharmacodynamic Studies: Towards Prediction of Nose-to-Brain Transport in Humans. AAPS JOURNAL 2015; 17:493-505. [PMID: 25693488 PMCID: PMC4406961 DOI: 10.1208/s12248-015-9724-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/27/2015] [Indexed: 01/03/2023]
Abstract
To investigate the potential added value of intranasal drug administration, preclinical studies to date have typically used the area under the curve (AUC) in brain tissue or cerebrospinal fluid (CSF) compared to plasma following intranasal and intravenous administration to calculate measures of extent like drug targeting efficiencies (%DTE) and nose-to-brain transport percentages (%DTP). However, CSF does not necessarily provide direct information on the target site concentrations, while total brain concentrations are not specific to that end either as non-specific binding is not explicitly considered. Moreover, to predict nose-to-brain transport in humans, the use of descriptive analysis of preclinical data does not suffice. Therefore, nose-to-brain research should be performed translationally and focus on preclinical studies to obtain specific information on absorption from the nose, and distinguish between the different transport routes to the brain (absorption directly from the nose to the brain, absorption from the nose into the systemic circulation, and distribution between the systemic circulation and the brain), in terms of extent as well as rate. This can be accomplished by the use of unbound concentrations obtained from plasma and brain, with subsequent advanced mathematical modeling. To that end, brain extracellular fluid (ECF) is a preferred sampling site as it represents most closely the site of action for many targets. Furthermore, differences in nose characteristics between preclinical species and humans should be considered. Finally, pharmacodynamic measurements that can be obtained in both animals and humans should be included to further improve the prediction of the pharmacokinetic-pharmacodynamic relationship of intranasally administered CNS drugs in humans.
Collapse
Affiliation(s)
- Mitchel J R Ruigrok
- Division of Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | | |
Collapse
|
27
|
Gomeni R. Use of predictive models in CNS diseases. Curr Opin Pharmacol 2014; 14:23-9. [DOI: 10.1016/j.coph.2013.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/15/2013] [Accepted: 10/24/2013] [Indexed: 11/28/2022]
|
28
|
|
29
|
|
30
|
PKPD Aspects of Brain Drug Delivery in a Translational Perspective. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
31
|
Bueters T, Ploeger BA, Visser SA. The virtue of translational PKPD modeling in drug discovery: selecting the right clinical candidate while sparing animal lives. Drug Discov Today 2013; 18:853-62. [DOI: 10.1016/j.drudis.2013.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/17/2013] [Accepted: 05/01/2013] [Indexed: 10/26/2022]
|
32
|
White paper: landscape on technical and conceptual requirements and competence framework in drug/disease modeling and simulation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2013; 2:e40. [PMID: 23887723 PMCID: PMC3674326 DOI: 10.1038/psp.2013.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/26/2013] [Indexed: 12/19/2022]
Abstract
Pharmaceutical sciences experts and regulators acknowledge that pharmaceutical development as well as drug usage requires more than scientific advancements to cope with current attrition rates/therapeutic failures. Drug disease modeling and simulation (DDM&S) creates a paradigm to enable an integrated and higher-level understanding of drugs, (diseased)systems, and their interactions (systems pharmacology) through mathematical/statistical models (pharmacometrics)1—hence facilitating decision making during drug development and therapeutic usage of medicines. To identify gaps and challenges in DDM&S, an inventory of skills and competencies currently available in academia, industry, and clinical practice was obtained through survey. The survey outcomes revealed benefits, weaknesses, and hurdles for the implementation of DDM&S. In addition, the survey indicated that no consensus exists about the knowledge, skills, and attributes required to perform DDM&S activities effectively. Hence, a landscape of technical and conceptual requirements for DDM&S was identified and serves as a basis for developing a framework of competencies to guide future education and training in DDM&S.
Collapse
|
33
|
de Lange EC. The mastermind approach to CNS drug therapy: translational prediction of human brain distribution, target site kinetics, and therapeutic effects. Fluids Barriers CNS 2013; 10:12. [PMID: 23432852 PMCID: PMC3602026 DOI: 10.1186/2045-8118-10-12] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/01/2013] [Indexed: 01/11/2023] Open
Abstract
Despite enormous advances in CNS research, CNS disorders remain the world's leading cause of disability. This accounts for more hospitalizations and prolonged care than almost all other diseases combined, and indicates a high unmet need for good CNS drugs and drug therapies.Following dosing, not only the chemical properties of the drug and blood-brain barrier (BBB) transport, but also many other processes will ultimately determine brain target site kinetics and consequently the CNS effects. The rate and extent of all these processes are regulated dynamically, and thus condition dependent. Therefore, heterogenious conditions such as species, gender, genetic background, tissue, age, diet, disease, drug treatment etc., result in considerable inter-individual and intra-individual variation, often encountered in CNS drug therapy.For effective therapy, drugs should access the CNS "at the right place, at the right time, and at the right concentration". To improve CNS therapies and drug development, details of inter-species and inter-condition variations are needed to enable target site pharmacokinetics and associated CNS effects to be translated between species and between disease states. Specifically, such studies need to include information about unbound drug concentrations which drive the effects. To date the only technique that can obtain unbound drug concentrations in brain is microdialysis. This (minimally) invasive technique cannot be readily applied to humans, and we need to rely on translational approaches to predict human brain distribution, target site kinetics, and therapeutic effects of CNS drugs.In this review the term "Mastermind approach" is introduced, for strategic and systematic CNS drug research using advanced preclinical experimental designs and mathematical modeling. In this way, knowledge can be obtained about the contributions and variability of individual processes on the causal path between drug dosing and CNS effect in animals that can be translated to the human situation. On the basis of a few advanced preclinical microdialysis based investigations it will be shown that the "Mastermind approach" has a high potential for the prediction of human CNS drug effects.
Collapse
Affiliation(s)
- Elizabeth Cm de Lange
- Division of Pharmacology, Leiden-Academic Center for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|