Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Toxicol Methods ; 127: 107504, 2024.
Article in English | MEDLINE | ID: mdl-38678804

ABSTRACT

We developed a novel, stress-free blood sampling method for minipigs, allowing continuous cortisol monitoring over 24 h. Baseline cortisol levels exhibited both ultradian and diurnal rhythms. During nighttime, smaller ultradian rhythms overlaid a lower baseline cortisol, which increased in sleeping pigs before lights were turned on. Additionally, we developed an analytical tool based on the R package "pracma" to quantify ultradian peak and circadian components of the cortisol profiles. To validate our model, we investigated the effects of Verucerfont, a CRH receptor antagonist, and Venlafaxine, a serotonin-norepinephrine reuptake inhibitor. Verucerfont reduced cortisol levels during the first 9 h without affecting diurnal rhythm. Cortisol peak parameters decreased, with a 31% reduction in overall area under the curve (AUC) and a 38% reduction in ultradian average AUC. Ultradian peaks decreased from 7 to 4.5, with 34% lower amplitude. Venlafaxine maintained plasma concentrations within the targeted human effective range. This method enables us to enhance our understanding of cortisol regulation and provide valuable insights for the impact of investigation drugs on the diurnal and ultradian rhythms of cortisol.


Subject(s)
Circadian Rhythm , Hydrocortisone , Swine, Miniature , Venlafaxine Hydrochloride , Animals , Swine , Hydrocortisone/blood , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Venlafaxine Hydrochloride/pharmacology , Ultradian Rhythm/drug effects , Ultradian Rhythm/physiology , Blood Specimen Collection/methods , Area Under Curve , Male , Female
2.
Xenobiotica ; 52(6): 591-607, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36000364

ABSTRACT

The use of the Göttingen minipig as an animal model for drug safety testing and prediction of human pharmacokinetics (PK) continues to gain momentum in pharmaceutical research and development. The aim of this study was to evaluate in vitro to in vivo extrapolation (IVIVE) methodologies for prediction of hepatic, metabolic clearance (CLhep,met) in Göttingen minipig, using a comprehensive set of compounds.In vivo clearance was determined in Göttingen minipig by intravenous cassette dosing and hepatocyte intrinsic clearance, plasma protein binding and non-specific incubation binding were determined in vitro. Prediction of CLhep,met was performed by IVIVE using conventional and adapted formats of the well-stirred liver model.The best prediction of in vivo CLhep,met from scaled in vitro kinetic data was achieved using an empirical correction factor based on a 'regression offset' of the IVIV relationship.In summary, these results expand the in vitro and in vivo PK knowledge in Göttingen minipig. We show regression corrected IVIVE provides superior prediction of in vivo CLhep,met in minipig offering a practical, unified scaling approach to address systematic under-predictions. Finally, we propose a reference set for researchers to establish their own 'lab-specific' regression correction for IVIVE in minipig.


Subject(s)
Hepatocytes , Models, Biological , Animals , Hepatocytes/metabolism , Humans , Kinetics , Liver/metabolism , Metabolic Clearance Rate , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Swine , Swine, Miniature
3.
Drug Metab Bioanal Lett ; 15(1): 12-21, 2022.
Article in English | MEDLINE | ID: mdl-35196975

ABSTRACT

BACKGROUND AND OBJECTIVE: Compound X is a drug candidate for the treatment of neurodegenerative diseases. Its brain distribution was evaluated as part of the lead identification and optimization activities undertaken in early drug discovery. METHODS: The brain distribution of compound X was studied in genetic transporter knockout rodent models, in vivo models with a chemical inhibitor, and in vitro transporter cell systems. RESULTS: Compound X was found to be a substrate for human Breast Cancer-Resistance Protein (BCRP) in vitro (efflux ratio 8.1) and rodent Bcrp in vivo (Kp, uuKO/Kp, uuWT = 0.15/0.057 = 2.7, p< 0.05) but not a substrate for human P-glycoprotein (P-gp) in vitro (efflux ratio 1.0) nor rodent P-gp in vivo (Kp, uuKO/Kp, uuWT = 0.056/ 0.051 = 1.1, p> 0.05). When both transporters were knocked out in vivo, Kp, uu increased to 0.51±0.02. A similar pattern observed across compounds with related chemistry corroborating the structure-activity relationship. CONCLUSION: While in vitro assays showed compound X to be a substrate for human BCRP and not P-gp, in vivo studies indicated a synergistic effect between rodent efflux transporters. However, this only accounted for ~50% of restricted BBB-transport, suggesting involvement of other efflux transporters. Considering Kp, uu as a key criterion for assessing the technical quality of CNS candidates before progression into clinical development, it is important to identify relevant screening assays for a better understanding of low Kp, uu and brain distribution in pre-clinical models for translation to humans.


Subject(s)
Blood-Brain Barrier , Rodentia , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Discovery , Humans , Membrane Transport Proteins/metabolism , Neoplasm Proteins , Rodentia/metabolism
4.
Eur J Pharm Biopharm ; 169: 211-219, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34756975

ABSTRACT

Drug efflux by P-glycoprotein (P-gp, ABCB1) is considered as a major obstacle for brain drug delivery for small molecules. P-gp-expressing cell monolayers are used for screening of new drug candidates during early states of drug development. It is, however, uncertain how well the in vitro studies can predict the in vivo P-gp mediated efflux at the blood-brain barrier (BBB). We previously developed a novel cell line of porcine origin, the iP-gp cell line, with high transepithelial resistance and functional expression of human P-gp. The aim of the present study was to evaluate the applicability of the cell line for screening of P-gp interactions of novel drug candidates. For this purpose, bidirectional fluxes of 14 drug candidates were measured in iP-gp cells and in MDCK-MDR1 cells, and compared with pharmacokinetic data obtained in male C57BL/6 mice. The iP-gp cells formed extremely tight monolayers (>15 000 Ω∙cm2) as compared to the MDCK- MDR1 cells (>250 Ω∙cm2) and displayed lower Papp,a-b values. The efflux ratios obtained with iP-gp and MDCK-MDR1 monolayers correlated with Kp,uu,brain values from the in vivo studies, where compounds with the lowest Kp,uu,brain generally displayed the highest efflux ratios. 12 of the tested compounds displayed a poor BBB penetration in mice as judged by Kp,uu less than 1. Of these compounds, nine compounds were categorized as P-gp substrates in the iP-gp screening, whereas analysis of data estimated in MDCK-MDR1 cells indicated four compounds as potential substrates. The results suggest that the iP-gp cell model may be a sensitive and useful screening tool for drug screening purposes to identify possible substrates of human P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Availability , Blood-Brain Barrier , Central Nervous System Agents/pharmacokinetics , Drug Evaluation, Preclinical/methods , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Central Nervous System Agents/classification , Drug Development/methods , Humans , Membrane Transport Proteins/metabolism , Mice , Swine , Technology, Pharmaceutical/methods , Tissue Distribution
5.
Neuropharmacology ; 186: 108454, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33444639

ABSTRACT

Positive modulation of cAMP signalling by phosphodiesterase (PDE) inhibitors has recently been explored as a potential target for the reversal of cognitive and behavioural deficits implicating the corticoaccumbal circuit. Previous studies show that PDE type 1 isoform B (PDE1B) inhibition may improve memory function in rodent models; however, the contribution of PDE1B inhibition to impulsivity, attentional and motivational functions as well as its neurophysiological effects have not been investigated. To address this, we recorded single unit activity in medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) in Lister Hooded rats treated with the PDE1B inhibitor Lu AF64386 and tested in the 5-choice serial reaction time task (5-CSRTT). We also asked whether PDE1B inhibition modulates neurophysiological deficits produced by subchronic phencyclidine (PCP) treatment, a rat pharmacological model of schizophrenia. Lu AF64386 significantly affected behavioural parameters consistent with a reduction in goal-directed behaviour, however without affecting accuracy. Additionally, it reduced mPFC neuronal activity. Pre-treatment with PCP did not affect behavioural parameters, however it significantly disrupted overall neuronal firing while increasing phasic responses to reward-predicting cues and disrupting mPFC-NAc cross-talk. The latter two effects were reversed by Lu AF64386. These findings suggest PDE1B inhibition may be beneficial in disorders implicating a dysfunction of the mPFC-NAc network.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Goals , Phencyclidine/toxicity , Phosphodiesterase Inhibitors/therapeutic use , Prefrontal Cortex/enzymology , Schizophrenia/enzymology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Disease Models, Animal , Female , Hallucinogens/toxicity , Phosphodiesterase Inhibitors/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Rats , Schizophrenia/chemically induced , Schizophrenia/drug therapy
6.
Neuropharmacology ; 103: 104-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26700248

ABSTRACT

P-glycoprotein (P-gp)-mediated brain efflux of xenobiotics is a well-known process, which may result in suboptimal target engagement and consequently reduced efficacy of drugs exerting their therapeutic effects in the central nervous system. In the present study the role of P-gp in transport across the blood-brain barrier (BBB) was investigated with a series of newer antidepressants (levomilnacipran, vilazodone and vortioxetine) and a control substrate (escitalopram) using P-gp knock-out (KO) and P-gp competent wild-type (WT) mice. Brain and plasma exposure time-courses were measured after an acute subcutaneous dose and at steady-state obtained after subcutaneous drug infusion by osmotic minipumps. Following acute dosing, the brain-to-plasma KO/WT exposure enhancement ratios ((AUCbrain ko/AUCplasma ko)/(AUCbrain WT/AUCplasma WT)) were 5.8 (levomilnacipran), 5.4 (vilazodone), 3.1 (escitalopram) and 0.9 (vortioxetine), respectively. At steady-state, assessment of Kp,uu (unbound brain concentrations/unbound plasma concentrations) revealed a restriction in the brain distribution in WT mice for all compounds except vortioxetine. Levomilnacipran exhibited the most pronounced efflux with a Kp,uu-value of 0.038 in WT mice which was increased to 0.37 in KO mice. Based on both the acute and steady-state distribution data, the results suggest that levomilnacipran, vilazodone and escitalopram are susceptible to P-gp mediated efflux at the BBB in vivo in mice, whereas vortioxetine was practically devoid of being affected by P-gp in vivo. The functional impact of the drug transport-controlling role of P-gp at the BBB was demonstrated by in vivo cortical serotonin transporter occupancy of vilazodone, which exhibited a 20-fold higher plasma EC50 in WT mice compared to KOs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacokinetics , Antidepressive Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Animals , Antidepressive Agents/blood , Biological Transport , Brain Chemistry/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Citalopram/blood , Citalopram/pharmacokinetics , Cyclopropanes/blood , Cyclopropanes/pharmacokinetics , Male , Mice , Milnacipran , Piperazines/blood , Piperazines/pharmacokinetics , Serotonin Plasma Membrane Transport Proteins/pharmacokinetics , Sulfides/blood , Sulfides/pharmacokinetics , Vilazodone Hydrochloride/blood , Vilazodone Hydrochloride/pharmacokinetics , Vortioxetine
SELECTION OF CITATIONS
SEARCH DETAIL
...