Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 28(11): 103758, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37660984

ABSTRACT

The suitability of small molecules as oral drugs is often assessed by simple physicochemical rules, the application of ligand efficiency scores or by composite scores based on physicochemical compound properties. These rules and scores are empirical and typically lack mechanistic background, such as information on pharmacokinetics (PK). We introduce new types of Compound Quality Scores (CQS, specifically called dose scores and cmax scores), which explicitly include predicted or, when available, experimental PK parameters and combine these with on-target potency. These CQS scores are surrogates for an estimated dose and corresponding cmax and allow prioritizing of compounds within test cascades as well as before synthesis. We demonstrate the complementarity and, in most cases, superior performance relative to existing efficiency metrics by project examples.


Subject(s)
Benchmarking , Ligands
2.
J Pharm Sci ; 112(8): 2285-2291, 2023 08.
Article in English | MEDLINE | ID: mdl-37062414

ABSTRACT

Many biotherapeutics such as monoclonal antibodies (mAbs) consist of various glycoforms, which can have different PK properties upon administration to animals and human. As a result, it is necessary to monitor the abundance of glycoforms and limit lot-to-lot variability during the manufacturing process. However, limited information is known about the clearance of mAb glycoforms from ocular space upon intravitreal injection. We present here an assessment of glycoform clearance of a biotherapeutic mAb (IgG1) from rabbit vitreous humor, aqueous humor and retina tissue using LC/MS. The results show that G0, G0F and G1F have similar T1/2, while mannose-5 has a longer T1/2 and is cleared slower in rabbit ocular space, which contradicted with what has been reported in the literature in which Mann5 was cleared faster systematically.


Subject(s)
Antibodies, Monoclonal , Retina , Animals , Rabbits , Humans , Chromatography, Liquid , Kinetics , Mass Spectrometry
3.
Elife ; 112022 05 26.
Article in English | MEDLINE | ID: mdl-35617485

ABSTRACT

Chronic liver injury causes fibrosis, characterized by the formation of scar tissue resulting from excessive accumulation of extracellular matrix (ECM) proteins. Hepatic stellate cell (HSC) myofibroblasts are the primary cell type responsible for liver fibrosis, yet there are currently no therapies directed at inhibiting the activity of HSC myofibroblasts. To search for potential anti-fibrotic compounds, we performed a high-throughput compound screen in primary human HSC myofibroblasts and identified 19 small molecules that induce HSC inactivation, including the polyether ionophore nanchangmycin (NCMC). NCMC induces lipid re-accumulation while reducing collagen expression, deposition of collagen in the extracellular matrix, cell proliferation, and migration. We find that NCMC increases cytosolic Ca2+ and reduces the phosphorylated protein levels of FYN, PTK2 (FAK), MAPK1/3 (ERK2/1), HSPB1 (HSP27), and STAT5B. Further, depletion of each of these kinases suppress COL1A1 expression. These studies reveal a signaling network triggered by NCMC to inactivate HSC myofibroblasts and reduce expression of proteins that compose the fibrotic scar. Identification of the antifibrotic effects of NCMC and the elucidation of pathways by which NCMC inhibits fibrosis provide new tools and therapeutic targets that could potentially be utilized to combat the development and progression of liver fibrosis.


Subject(s)
Cicatrix , Hepatic Stellate Cells , Cicatrix/pathology , Collagen/metabolism , Ethers , Extracellular Matrix Proteins/metabolism , Fibrosis , Focal Adhesion Kinase 1/metabolism , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Spiro Compounds
4.
Int J Pharm ; 613: 121361, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34896561

ABSTRACT

Quantitation of ocular drug metabolism is important, but only sparse data is currently available. Herein, the pharmacokinetics of four drugs, substrates of metabolizing enzymes, was investigated in albino rabbit eyes after intracameral and intravitreal administrations. Acetaminophen, brimonidine, cefuroxime axetil, and sunitinib and their corresponding metabolites were quantitated in the cornea, iris-ciliary body, aqueous humor, lens, vitreous humor, and neural retina with LC-MS/MS analytics. Non-compartmental analysis was employed to estimate the pharmacokinetic parameters of the parent drugs and metabolites. The area under the curve (AUC) values of metabolites were 12-70 times lower than the AUC values of the parent drugs in the tissues with the highest enzymatic activity. The ester prodrug cefuroxime axetil was an exception because it was efficiently and quantitatively converted to cefuroxime in the ocular tissues. In contrast to the liver, sulfotransferases, aldehyde oxidase, and cytochrome P450 3A activities were low in the eye and they had negligible impact on ocular drug clearance. With the exception of esterase substrates, metabolism seems to be a minor player in ocular pharmacokinetics. However, metabolites might contribute to ocular toxicity, and drug metabolism in various eye tissues should be investigated and understood thoroughly.


Subject(s)
Pharmaceutical Preparations , Animals , Chromatography, Liquid , Rabbits , Retina , Tandem Mass Spectrometry , Vitreous Body
5.
Pharmaceutics ; 13(8)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34452112

ABSTRACT

Bidirectional permeability measurement with cellular models grown on Transwell inserts is widely used in pharmaceutical research since it not only provides information about the passive permeability of a drug, but also about transport proteins involved in the active transport of drug substances across physiological barriers. With the increasing number of investigative drugs coming from chemical space beyond Lipinski's Rule of 5, it becomes more and more challenging to provide meaningful data with the standard permeability assay. This is exemplified here by the difficulties we encountered with the cyclic depsipeptides emodepside and its close analogs with molecular weight beyond 1000 daltons and cLogP beyond 5. The aim of this study is to identify potential reasons for these challenges and modify the permeability assays accordingly. With the modified assay, intrinsic permeability and in vitro efflux of depsipeptides could be measured reliably. The improved correlation to in vivo bioavailability and tissue distribution data indicated the usefulness of the modified permeability assay for the in vitro screening of compounds beyond the Rule of 5.

6.
Mol Pharm ; 18(7): 2703-2713, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34151575

ABSTRACT

The discovery of new small-molecule drugs for intravitreal administration would benefit from simple models to predict vitreal clearance (CL). The current models available have limitations in their applicability to small-molecule drugs and translatability to humans. We developed a mechanistic model combining the diffusion rate of the molecule in the vitreous and permeability across posterior segment tissues and applied it to 30 small molecules with observed CL available mostly from literature. We used Caco-2 permeability as a surrogate for ocular tissue permeability. The model predicted rabbit vitreal CL well, with 80% of the predictions being within a 2-fold range of the observed CL. For an accurate prediction, it was crucial to consider the anterior diffusion CL from the vitreous to the aqueous and a limiting diffusion CL for the whole eye. We observed no major differences in model accuracy when using literature permeability values from retinal pigment epithelial cell models. Importantly, by adopting the specific dimensions of the human eye, the model was able to accurately predict vitreal CL of four compounds for which human vitreal CL data are available. In summary, this mechanistic model enables a simple, accurate, and translatable estimation of small-molecule vitreal CL.


Subject(s)
Cell Membrane Permeability , Models, Biological , Pharmaceutical Preparations/metabolism , Retinal Pigment Epithelium/metabolism , Small Molecule Libraries/metabolism , Vitreous Body/metabolism , Animals , Caco-2 Cells , Diffusion , Humans , Kinetics , Rabbits
7.
Mol Pharm ; 18(3): 1305-1316, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33595329

ABSTRACT

Hydrolytic reactions constitute an important pathway of drug metabolism and a significant route of prodrug activation. Many ophthalmic drugs and prodrugs contain ester groups that greatly enhance their permeation across several hydrophobic barriers in the eye before the drugs are either metabolized or released, respectively, via hydrolysis. Thus, the development of ophthalmic drug therapy requires the thorough profiling of substrate specificities, activities, and expression levels of ocular esterases. However, such information is scant in the literature, especially for preclinical species often used in ophthalmology such as rabbits and pigs. Therefore, our aim was to generate systematic information on the activity and expression of carboxylesterases (CESs) and arylacetamide deacetylase (AADAC) in seven ocular tissue homogenates from these two species. The hydrolytic activities were measured using a generic esterase substrate (4-nitrophenyl acetate) and, in the absence of validated substrates for rabbit and pig enzymes, with selective substrates established for human CES1, CES2, and AADAC (d-luciferin methyl ester, fluorescein diacetate, procaine, and phenacetin). Kinetics and inhibition studies were conducted using these substrates and, again due to a lack of validated rabbit and pig CES inhibitors, with known inhibitors for the human enzymes. Protein expression levels were measured using quantitative targeted proteomics. Rabbit ocular tissues showed significant variability in the expression of CES1 (higher in cornea, lower in conjunctiva) and CES2 (higher in conjunctiva, lower in cornea) and a poor correlation of CES expression with hydrolytic activities. In contrast, pig tissues appear to express only CES1, and CES3 and AADAC seem to be either low or absent, respectively, in both species. The current study revealed remarkable species and tissue differences in ocular hydrolytic enzymes that can be taken into account in the design of esterase-dependent prodrugs and drug conjugates, the evaluation of ocular effects of systemic drugs, and in translational and toxicity studies.


Subject(s)
Carboxylesterase/metabolism , Eye/metabolism , Animals , Female , Humans , Hydrolysis/drug effects , Male , Nitrophenols/metabolism , Prodrugs/metabolism , Proteomics/methods , Rabbits , Substrate Specificity/physiology , Swine
8.
Pharmaceutics ; 12(12)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276439

ABSTRACT

The rat is a commonly used species in ocular drug research. Detailed methods of separating rat ocular tissues have not been described in literature. To understand the intraocular drug distribution, we developed a robust method for the separation of individual anterior and posterior substructures of pigmented Brown Norway (BN) and albino Wistar Han (WH) rat eyes, followed by quantification of drug concentration in these substructures. A short formalin incubation, which did not interfere with drug quantification, enabled the preservation of individual tissue sections while minimizing cross-tissue contamination, as demonstrated by histological analysis. Following oral administration, we applied the tissue separation method, in order to determine the ocular concentrations of dexamethasone and levofloxacin, as well as two in-house molecules BI 113823 and BI 1026706, compounds differing in their melanin binding. The inter-individual variability in tissue partitioning coefficients (Kp) was low, demonstrating the reproducibility of the separation method. Kp values of individual tissues varied up to 100-fold in WH and up to 46,000-fold in BN rats highlighting the importance of measuring concentration directly from the ocular tissue of interest. Additionally, clear differences were observed in the BN rat tissue partitioning compared to the WH rat. Overall, the developed method enables a reliable determination of small molecule drug concentrations in ocular tissues to support ocular drug research and development.

9.
Pharmaceutics ; 11(11)2019 Nov 11.
Article in English | MEDLINE | ID: mdl-31718023

ABSTRACT

Movement of xenobiotic substances across the blood-brain barrier (BBB) is tightly regulated by various transporter proteins, especially the efflux transporters P-glycoprotein (P-gp/MDR1) and breast cancer resistance protein (BCRP). Avoiding drug efflux at the BBB is a unique challenge for the development of new central nervous system (CNS) drugs. Drug efflux at the BBB is described by the partition coefficient of unbound drug between brain and plasma (Kp,uu,brain) which is typically obtained from in vivo and often additionally in vitro measurements. Here, we describe a new method for the rapid estimation of the in vivo drug efflux at the BBB of rats: the measurement of the partition coefficient of a drug between brain and skeletal muscle (Kp,brain/muscle). Assuming a closely similar distribution of drugs into the brain and muscle and that the efflux transporters are only expressed in the brain, Kp,brain/muscle, similar to Kp,uu,brain, reflects the efflux at the BBB. The new method requires a single in vivo experiment. For 64 compounds from different research programs, we show the comparability to other approaches used to obtain Kp,uu,brain. P-gp- and BCRP-overexpressing cell systems are valuable in vitro tools for prescreening. Drug efflux at the BBB can be most accurately predicted based on a simple algorithm incorporating data from both in vitro assays. In conclusion, the combined use of our new in vivo method and the in vitro tools allows an efficient screening method in drug discovery with respect to efflux at the BBB.

10.
Proc Natl Acad Sci U S A ; 116(20): 10156-10161, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028142

ABSTRACT

Transient receptor potential canonical type 6 (TRPC6) is a nonselective receptor-operated cation channel that regulates reactive fibrosis and growth signaling. Increased TRPC6 activity from enhanced gene expression or gain-of-function mutations contribute to cardiac and/or renal disease. Despite evidence supporting a pathophysiological role, no orally bioavailable selective TRPC6 inhibitor has yet been developed and tested in vivo in disease models. Here, we report an orally bioavailable TRPC6 antagonist (BI 749327; IC50 13 nM against mouse TRPC6, t1/2 8.5-13.5 hours) with 85- and 42-fold selectivity over the most closely related channels, TRPC3 and TRPC7. TRPC6 calcium conductance results in the stimulation of nuclear factor of activated T cells (NFAT) that triggers pathological cardiac and renal fibrosis and disease. BI 749327 suppresses NFAT activation in HEK293T cells expressing wild-type or gain-of-function TRPC6 mutants (P112Q, M132T, R175Q, R895C, and R895L) and blocks associated signaling and expression of prohypertrophic genes in isolated myocytes. In vivo, BI 749327 (30 mg/kg/day, yielding unbound trough plasma concentration ∼180 nM) improves left heart function, reduces volume/mass ratio, and blunts expression of profibrotic genes and interstitial fibrosis in mice subjected to sustained pressure overload. Additionally, BI 749327 dose dependently reduces renal fibrosis and associated gene expression in mice with unilateral ureteral obstruction. These results provide in vivo evidence of therapeutic efficacy for a selective pharmacological TRPC6 inhibitor with oral bioavailability and suitable pharmacokinetics to ameliorate cardiac and renal stress-induced disease with fibrosis.


Subject(s)
Cardiomegaly/drug therapy , Nephrosclerosis/drug therapy , TRPC6 Cation Channel/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical , Fibrosis , HEK293 Cells , Heart/drug effects , Humans , Kidney/drug effects , Mice
11.
Adipocyte ; 7(4): 277-284, 2018.
Article in English | MEDLINE | ID: mdl-30161013

ABSTRACT

Despite increased knowledge of nutrient intake regulation and energy homeostasis, treatment options for obesity remain limited. Food reward consists of two branches: gustatory and post-ingestive nutritive information. Drosophila lacking dSLC5A11 (sodium/glucose cotransporter 6-SGLT6) prefer L-glucose over D-glucose independently of their state of satiety. Human SGLT6 is an active transporter of myo-inositol and D-glucose. We investigated expression of SGLT6 in human tissue and found a significant expression in the small intestine and brain. The preference between a metabolizable and a non-metabolizable sugar was tested in 3 mouse models with a selective and potent SGLT6 inhibitor. No influence on sugar preference was seen with SGLT6 inhibition. These studies suggest that SGLT6 does not play a significant role in nutrient sensing in mammals.


Subject(s)
Anti-Obesity Agents/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Obesity/drug therapy , Obesity/metabolism , Symporters/antagonists & inhibitors , Symporters/metabolism , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/therapeutic use , Caco-2 Cells , Food Preferences/drug effects , Glucose/metabolism , HEK293 Cells , Humans , Inositol/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy
12.
PLoS One ; 13(1): e0191225, 2018.
Article in English | MEDLINE | ID: mdl-29385160

ABSTRACT

BACKGROUND: Forty million adults in the US suffer from anxiety disorders, making these the most common forms of mental illness. Transient receptor potential channel canonical subfamily (TRPC) members 4 and 5 are non-selective cation channels highly expressed in regions of the cortex and amygdala, areas thought to be important in regulating anxiety. Previous work with null mice suggests that inhibition of TRPC4 and TRPC5 may have anxiolytic effects. HC-070 IN VITRO: To assess the potential of TRPC4/5 inhibitors as an avenue for treatment, we invented a highly potent, small molecule antagonist of TRPC4 and TRPC5 which we call HC-070. HC-070 inhibits recombinant TRPC4 and TRPC5 homomultimers in heterologous expression systems with nanomolar potency. It also inhibits TRPC1/5 and TRPC1/4 heteromultimers with similar potency and reduces responses evoked by cholecystokinin tetrapeptide (CCK-4) in the amygdala. The compound is >400-fold selective over a wide range of molecular targets including ion channels, receptors, and kinases. HC-070 IN VIVO: Upon oral dosing in mice, HC-070 achieves exposure levels in the brain and plasma deemed sufficient to test behavioral activity. Treatment with HC-070 attenuates the anxiogenic effect of CCK-4 in the elevated plus maze (EPM). The compound recapitulates the phenotype observed in both null TRPC4 and TRPC5 mice in a standard EPM. Anxiolytic and anti-depressant effects of HC-070 are also observed in pharmacological in vivo tests including marble burying, tail suspension and forced swim. Furthermore, HC-070 ameliorates the increased fear memory induced by chronic social stress. A careful evaluation of the pharmacokinetic-pharmacodynamic relationship reveals that substantial efficacy is observed at unbound brain levels similar to, or even lower than, the 50% inhibitory concentration (IC50) recorded in vitro, increasing confidence that the observed effects are indeed mediated by TRPC4 and/or TRPC5 inhibition. Together, this experimental data set introduces a novel, high quality, small molecule antagonist of TRPC4 and TRPC5 containing channels and supports the targeting of TRPC4 and TRPC5 channels as a new mechanism of action for the treatment of psychiatric symptoms.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , TRPC Cation Channels/antagonists & inhibitors , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacokinetics , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacokinetics , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/psychology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Behavior, Animal/drug effects , Depression/drug therapy , Depression/metabolism , Depression/psychology , Disease Models, Animal , Fear/drug effects , Fear/physiology , Fear/psychology , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , High-Throughput Screening Assays , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL
13.
Mol Pharm ; 14(10): 3436-3447, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28880093

ABSTRACT

Transporters at the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) play a pivotal role as gatekeepers for efflux or uptake of endogenous and exogenous molecules. The protein expression of a number of them has already been determined in the brains of rodents, nonhuman primates, and humans using quantitative targeted absolute proteomics (QTAP). The dog is an important animal model for drug discovery and development, especially for safety evaluations. The purpose of the present study was to clarify the relevance of the transporter protein expression for drug distribution in the dog brain and CSF. We used QTAP to examine the protein expression of 17 selected transporters and receptors at the dog BBB and BCSFB. For the first time, we directly linked the expression of two efflux transporters, P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), to regional brain and CSF distribution using specific substrates. Two cocktails, each containing one P-gp substrate (quinidine or apafant) and one BCRP substrate (dantrolene or daidzein) were infused intravenously prior to collection of the brain. Transporter expression varied only slightly between the capillaries of different brain regions and did not result in region-specific distribution of the investigated substrates. There were, however, distinct differences between brain capillaries and choroid plexus. Largest differences were observed for BCRP and P-gp: both were highly expressed in brain capillaries, but no BCRP and only low amounts of P-gp were detected in the choroid plexus. Kp,uu,brain and Kp,uu,CSF of both P-gp substrates were indicative of drug efflux. Also, Kp,uu,brain for the BCRP substrates was low. In contrast, Kp,uu,CSF for both BCRP substrates was close to unity, resulting in Kp,uu,CSF/Kp,uu,brain ratios of 7 and 8, respectively. We conclude that the drug transporter expression profiles differ between the BBB and BCSFB in dogs, that there are species differences in the expression profiles, and that CSF is not a suitable surrogate for unbound brain concentrations of BCRP substrates in dogs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Brain/blood supply , Capillaries/metabolism , Choroid Plexus/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/blood , ATP Binding Cassette Transporter, Subfamily B, Member 1/cerebrospinal fluid , ATP Binding Cassette Transporter, Subfamily G, Member 2/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2/cerebrospinal fluid , Animals , Azepines/pharmacokinetics , Biological Transport , Blood-Brain Barrier , Brain/metabolism , Dantrolene/pharmacokinetics , Dogs , Female , Gene Expression Profiling , Isoflavones/pharmacokinetics , Male , Proteomics/methods , Quinidine/pharmacokinetics , Tissue Distribution , Triazoles/pharmacokinetics
14.
Bioorg Med Chem Lett ; 25(3): 581-6, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25556092

ABSTRACT

Through a ligand-based pharmacophore model (S)-proline based compounds were identified as potent cannabinoid receptor 2 (CB2) agonists with high selectivity over the cannabinoid receptor 1 (CB1). Structure-activity relationship investigations for this compound class lead to oxo-proline compounds 21 and 22 which combine an impressive CB1 selectivity profile with good pharmacokinetic properties. In a streptozotocin induced diabetic neuropathy model, 22 demonstrated a dose-dependent reversal of mechanical hyperalgesia.


Subject(s)
Isoxazoles/chemistry , Proline/chemistry , Pyrrolidonecarboxylic Acid/analogs & derivatives , Receptor, Cannabinoid, CB2/agonists , Animals , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/drug therapy , Half-Life , Humans , Isoxazoles/pharmacokinetics , Isoxazoles/therapeutic use , Ligands , Male , Microsomes, Liver/metabolism , Proline/pharmacokinetics , Proline/therapeutic use , Protein Binding , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/pharmacokinetics , Pyrrolidonecarboxylic Acid/therapeutic use , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Solubility , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 25(3): 587-92, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25575658

ABSTRACT

A novel class of potent cannabinoid receptor 2 (CB2) agonists based on a (S)-piperidine scaffold was identified using ligand-based pharmacophore models. Optimization of solubility and metabolic stability led to the identification of several potent CB2 agonists (e.g., 30) that displayed selectivity over cannabinoid receptor 1 (CB1) and acceptable drug like properties. In rats, compound 30 demonstrated a favorable pharmacokinetic profile and efficacy in a Streptozotocin-induced diabetic neuropathy model, with full reversal of mechanical hyperalgesia.


Subject(s)
Pipecolic Acids/chemistry , Piperidines/chemistry , Receptor, Cannabinoid, CB2/agonists , Thiazines/chemistry , Animals , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/drug therapy , Half-Life , Humans , Ligands , Male , Microsomes, Liver/metabolism , Pain/drug therapy , Pipecolic Acids/pharmacokinetics , Pipecolic Acids/therapeutic use , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Binding , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Solubility , Structure-Activity Relationship , Thiazines/pharmacokinetics , Thiazines/therapeutic use
16.
Bioorg Med Chem Lett ; 21(7): 2011-6, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21354795

ABSTRACT

A high-throughput screening campaign has identified 1,4-diazepane compounds which are potent Cannabinoid receptor 2 agonists with excellent selectivity against the Cannabinoid receptor 1. This class of compounds suffered from low metabolic stability. Following various strategies, compounds with a good stability in liver microsomes and rat PK profile have been identified.


Subject(s)
Azepines/pharmacology , Receptor, Cannabinoid, CB2/agonists , Animals , Azepines/chemistry , Microsomes, Liver/metabolism , Rats , Rats, Wistar
17.
Eur J Pharm Sci ; 16(3): 119-28, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12128165

ABSTRACT

The aim of the present study was to determine a potential impact of P-glycoprotein (P-gp) on the tissue distribution and disposition of apafant (WEB 2086, CAS 105219-56-5), a selective platelet-activating factor antagonist, and on digoxin in mdr1a(-/-) and wildtype mice. Transport experiments in Caco-2 monolayers at low concentrations (<10 microM) showed that the secretory flux of [(14)C]apafant and [(3)H]digoxin exceeded the absorptive flux nine times. This efflux was concentration dependent and subject to inhibition by the P-gp substrates verapamil and cyclosporin A. This indicates that active drug transporter P-gp was involved in apafant and digoxin absorption. Mdr1a(-/-) mice showed a more than 70-fold higher concentration of digoxin-related radioactivity (P<0.001) in the brain than wildtype mice after intravenous doses of 0.05 mg/kg [(3)H]digoxin. Differences were less pronounced in other tissues. Both liquid scintillation counting and whole body autoradiography yielded comparable results and they also matched recently published data. Apafant-related radioactivity was about ten-fold higher in the brain of mdr1a(-/-) mice compared to wildtype mice following intravenous doses of 2 mg/kg radiolabelled apafant. Only slight or negligible differences were observed in other tissues. In wildtype mice, intestinal excretion of [(14)C]apafant (54.9%) exceeded biliary excretion (26.5%). However, in mdr1a(-/-) mice biliary excretion (50.7%) exceeded intestinal excretion (6.8%). These differences were mirrored in the urinary and faecal excretion. Pharmacokinetic parameters of apafant and radioactivity did not differ between wildtype and mdr1a(-/-) mice. The conclusions were: (1) apafant and digoxin are P-gp substrates, and (2) absence of mdr1a encoded P-gp significantly alters tissue distribution (especially in brain) and excretion routes (biliary and intestinal) of apafant.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Azepines/pharmacokinetics , Platelet Activating Factor/antagonists & inhibitors , Triazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Animals , Area Under Curve , Azepines/blood , Azepines/chemistry , Bile Ducts/metabolism , Caco-2 Cells/metabolism , Carbon Radioisotopes/pharmacokinetics , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Knockout , Platelet Activating Factor/metabolism , Tissue Distribution/physiology , Triazoles/blood , Triazoles/chemistry
18.
ALTEX ; 13(1): 17-23, 1996.
Article in English | MEDLINE | ID: mdl-11178441

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) represents a central distal mediator of inflammation. It is a protein released upon infections, traumatic, lesions or autoimmune disorders from immunocompetent cells and thus maintains or enhances the inflammatory reaction. The determination of such mediators in experimentally challenged animals is a mean for testing the efficacy of putative drugs. Alternative attempts for the assessment of mediator release from cell cultures are limited by profound differences in the time course of mediator release in vitro. We checked the hypothesis that these kinetic differences are due to the lack of elimination of mediators formed and released in vitro. We used the release of TNF-alpha from liver macrophage cultures stimulated with the bacterial cell wall component endotoxin as a model. The discrepancy between the in vivo release of the cytokines during endotoxic shock in the rat and the in vitro release from Kupffer cells was confirmed. By using a continuous open perfusion system instead of a static culture, the simulation of an elimination resulted in a mediator release that closely resembled the kinetics seen in vivo. Perfusion cultures appear to be suitable for relevant in vitro screening models in drug development and testing.

SELECTION OF CITATIONS
SEARCH DETAIL
...