Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Pharmacokinet Pharmacodyn ; 50(3): 229-241, 2023 06.
Article in English | MEDLINE | ID: mdl-36877385

ABSTRACT

Monoclonal antibodies, endogenous IgG, and serum albumin bind to FcRn in the endosome for salvaging and recycling after pinocytotic uptake, which prolongs their half-life. This mechanism has been broadly recognized and is incorporated in currently available PBPK models. Newer types of large molecules have been designed and developed, which also bind to FcRn in the plasma space for various mechanistic reasons. To incorporate FcRn binding affinity in PBPK models, binding in the plasma space and subsequent internalisation into the endosome needs to be explicitly represented. This study investigates the large molecules model in PK-Sim® and its applicability to molecules with FcRn binding affinity in plasma. With this purpose, simulations of biologicals with and without plasma binding to FcRn were performed with the large molecule model in PK-Sim®. Subsequently, this model was extended to ensure a more mechanistic description of the internalisation of FcRn and the FcRn-drug complexes. Finally, the newly developed model was used in simulations to explore the sensitivity for FcRn binding in the plasma space, and it was fitted to an in vivo dataset of wild-type IgG and FcRn inhibitor plasma concentrations in Tg32 mice. The extended model demonstrated a strongly increased sensitivity of the terminal half-life towards the plasma FcRn binding affinity and could successfully fit the in vivo dataset in Tg32 mice with meaningful parameter estimates.


Subject(s)
Antibodies, Monoclonal , Receptors, Fc , Mice , Animals , Receptors, Fc/metabolism , Antibodies, Monoclonal/metabolism , Endosomes/metabolism , Immunoglobulin G/metabolism
3.
J Pharmacokinet Pharmacodyn ; 45(5): 763, 2018 10.
Article in English | MEDLINE | ID: mdl-30171443

ABSTRACT

The original version of this article was published open access. Unfortunately, due to a technical issue, the copyright holder name in the online version (HTML and XML) is incorrectly published as "Springer Science+Business Media, LLC, part of Springer Nature 2018". Instead, it should be "The Author(s) 2018".

4.
Br J Pharmacol ; 175(21): 4121-4136, 2018 11.
Article in English | MEDLINE | ID: mdl-30051456

ABSTRACT

BACKGROUND AND PURPOSE: Target binding kinetics influence the time course of the drug effect (pharmacodynamics) both (i) directly, by affecting the time course of target occupancy, driven by the pharmacokinetics of the drug, competition with endogenous ligands and target turnover, and (ii) indirectly, by affecting signal transduction and homeostatic feedback. For dopamine D2 receptor antagonists, it has been hypothesized that fast receptor binding kinetics cause fewer side effects, because part of the dynamics of the dopaminergic system is preserved by displacement of these antagonists. EXPERIMENTAL APPROACH: Target binding kinetics of D2 receptor antagonists and signal transduction after dopamine and D2 receptor antagonist exposure were measured in vitro. These data were integrated by mechanistic modelling, taking into account competitive binding of endogenous dopamine and the antagonist, the turnover of the second messenger cAMP and negative feedback by PDE turnover. KEY RESULTS: The proposed signal transduction model successfully described the cellular cAMP response for 17 D2 receptor antagonists with widely different binding kinetics. Simulation of the response to fluctuating dopamine concentrations revealed that a significant effect of the target binding kinetics on the dynamics of the signalling only occurs at endogenous dopamine concentration fluctuations with frequencies below 1 min-1 . CONCLUSIONS AND IMPLICATIONS: Signal transduction and feedback are important determinants of the time course of drug effects. The effect of the D2 receptor antagonist dissociation rate constant (koff ) is limited to the maximal rate of fluctuations in dopamine signalling as determined by the dopamine koff and the cAMP turnover.


Subject(s)
Dopamine Antagonists/pharmacology , Dopamine/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Binding Sites/drug effects , CHO Cells , Cricetulus , Kinetics , Models, Biological , Signal Transduction/drug effects
5.
J Pharmacokinet Pharmacodyn ; 45(4): 621-635, 2018 08.
Article in English | MEDLINE | ID: mdl-29777407

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.


Subject(s)
Morphine/pharmacokinetics , Animals , Brain/metabolism , Electroencephalography/methods , Extracellular Fluid/metabolism , Kinetics , Male , Models, Biological , Rats , Rats, Wistar
6.
AAPS J ; 20(1): 11, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29204742

ABSTRACT

Selectivity is an important attribute of effective and safe drugs, and prediction of in vivo target and tissue selectivity would likely improve drug development success rates. However, a lack of understanding of the underlying (pharmacological) mechanisms and availability of directly applicable predictive methods complicates the prediction of selectivity. We explore the value of combining physiologically based pharmacokinetic (PBPK) modeling with quantitative structure-activity relationship (QSAR) modeling to predict the influence of the target dissociation constant (K D) and the target dissociation rate constant on target and tissue selectivity. The K D values of CB1 ligands in the ChEMBL database are predicted by QSAR random forest (RF) modeling for the CB1 receptor and known off-targets (TRPV1, mGlu5, 5-HT1a). Of these CB1 ligands, rimonabant, CP-55940, and Δ8-tetrahydrocanabinol, one of the active ingredients of cannabis, were selected for simulations of target occupancy for CB1, TRPV1, mGlu5, and 5-HT1a in three brain regions, to illustrate the principles of the combined PBPK-QSAR modeling. Our combined PBPK and target binding modeling demonstrated that the optimal values of the K D and k off for target and tissue selectivity were dependent on target concentration and tissue distribution kinetics. Interestingly, if the target concentration is high and the perfusion of the target site is low, the optimal K D value is often not the lowest K D value, suggesting that optimization towards high drug-target affinity can decrease the benefit-risk ratio. The presented integrative structure-pharmacokinetic-pharmacodynamic modeling provides an improved understanding of tissue and target selectivity.


Subject(s)
Models, Biological , Pharmacokinetics , Quantitative Structure-Activity Relationship , Humans , Organ Specificity , Receptor, Cannabinoid, CB1/metabolism
7.
Expert Opin Drug Discov ; 12(12): 1207-1218, 2017 12.
Article in English | MEDLINE | ID: mdl-28933618

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.


Subject(s)
Central Nervous System Agents/administration & dosage , Drug Design , Models, Biological , Animals , Central Nervous System Agents/pharmacokinetics , Central Nervous System Agents/pharmacology , Computer Simulation , Drug Discovery/methods , Humans , Molecular Targeted Therapy , Time Factors , Tissue Distribution
8.
Sci Rep ; 7(1): 52, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28246398

ABSTRACT

CC Chemokine Receptor 2 (CCR2) and its endogenous ligand CCL2 are involved in a number of diseases, including atherosclerosis. Several CCR2 antagonists have been developed as potential therapeutic agents, however their in vivo clinical efficacy was limited. In this report, we aimed to determine whether 15a, an antagonist with a long residence time on the human CCR2, is effective in inhibiting the development of atherosclerosis in a mouse disease model. First, radioligand binding assays were performed to determine affinity and binding kinetics of 15a on murine CCR2. To assess the in vivo efficacy, western-type diet fed apoE-/- mice were treated daily with 15a or vehicle as control. Treatment with 15a reduced the amount of circulating CCR2+ monocytes and the size of the atherosclerotic plaques in both the carotid artery and the aortic root. We then showed that the long pharmacokinetic half-life of 15a combined with the high drug concentrations ensured prolonged CCR2 occupancy. These data render 15a a promising compound for drug development and confirms high receptor occupancy as a key parameter when targeting chemokine receptors.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Cyclopentanes/pharmacology , Isoquinolines/pharmacology , Receptors, CCR2/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/pathology , CHO Cells , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cricetulus , Cyclopentanes/pharmacokinetics , Diet, Atherogenic , Disease Models, Animal , Isoquinolines/pharmacokinetics , Male , Mice , Monocytes/metabolism , Monocytes/pathology , Receptors, CCR2/metabolism
9.
Trends Pharmacol Sci ; 37(10): 831-842, 2016 10.
Article in English | MEDLINE | ID: mdl-27394919

ABSTRACT

It is generally accepted that, in conjunction with pharmacokinetics, the first-order rate constant of target dissociation is a major determinant of the time course and duration of in vivo target occupancy. Here we show that the second-order rate constant of target association can be equally important. On the basis of the commonly used mathematical models for drug-target binding, it is shown that a high target association rate constant can increase the (local) concentration of the drug, which decreases the rate of decline of target occupancy. The increased drug concentration can also lead to increased off-target binding and decreased selectivity. Therefore, the kinetics of both target association and dissociation need to be taken into account in the selection of drug candidates with optimal pharmacodynamic properties.


Subject(s)
Drug Design , Models, Theoretical , Pharmacokinetics , Drug Delivery Systems , Drug Discovery/methods , Humans , Pharmaceutical Preparations/metabolism
10.
Expert Opin Drug Discov ; 11(1): 45-63, 2016.
Article in English | MEDLINE | ID: mdl-26484747

ABSTRACT

INTRODUCTION: Drug-target binding kinetics are major determinants of the time course of drug action for several drugs, as clearly described for the irreversible binders omeprazole and aspirin. This supports the increasing interest to incorporate newly developed high-throughput assays for drug-target binding kinetics in drug discovery. A meaningful application of in vitro drug-target binding kinetics in drug discovery requires insight into the relation between in vivo drug effect and in vitro measured drug-target binding kinetics. AREAS COVERED: In this review, the authors discuss both the relation between in vitro and in vivo measured binding kinetics and the relation between in vivo binding kinetics, target occupancy and effect profiles. EXPERT OPINION: More scientific evidence is required for the rational selection and development of drug-candidates on the basis of in vitro estimates of drug-target binding kinetics. To elucidate the value of in vitro binding kinetics measurements, it is necessary to obtain information on system-specific properties which influence the kinetics of target occupancy and drug effect. Mathematical integration of this information enables the identification of drug-specific properties which lead to optimal target occupancy and drug effect in patients.


Subject(s)
Drug Delivery Systems , Drug Discovery/methods , Models, Biological , Animals , High-Throughput Screening Assays/methods , Humans , Pharmaceutical Preparations/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL