Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 50(1): 65-75, 2022 01.
Article in English | MEDLINE | ID: mdl-34620695

ABSTRACT

Small molecules that present complex absorption, distribution, metabolism, and elimination (ADME) properties can be challenging to investigate as potential therapeutics. Acquiring data through standard methods can yield results that are insufficient to describe the in vivo situation, which can affect downstream development decisions. Implementing in vitro-in vivo-in silico strategies throughout the drug development process is effective in identifying and mitigating risks while speeding up their development. Risdiplam (Evrysdi)-an orally bioavailable, small molecule approved by the US Food and Drug Administration and more recently by the European Medicines Agency for the treatment of patients ≥2 months of age with spinal muscular atrophy-is presented here as a case study. Risdiplam is a low-turnover compound whose metabolism is mediated through a non-cytochrome P450 enzymatic pathway. Four main challenges of risdiplam are discussed: predicting in vivo hepatic clearance, determining in vitro metabolites with regard to metabolites in safety testing guidelines, elucidating enzymes responsible for clearance, and estimating potential drug-drug interactions. A combination of in vitro and in vivo results was successfully extrapolated and used to develop a robust physiologically based pharmacokinetic model of risdiplam. These results were verified through early clinical studies, further strengthening the understanding of the ADME properties of risdiplam in humans. These approaches can be applied to other compounds with similar ADME profiles, which may be difficult to investigate using standard methods. SIGNIFICANCE STATEMENT: Risdiplam is the first approved, small-molecule, survival of motor neuron 2 mRNA splicing modifier for the treatment of spinal muscular atrophy. The approach taken to characterize the absorption, distribution, metabolism, and excretion (ADME) properties of risdiplam during clinical development incorporated in vitro-in vivo-in silico techniques, which may be applicable to other small molecules with challenging ADME. These strategies may be useful in improving the speed at which future drug molecules can be developed.


Subject(s)
Azo Compounds/metabolism , Azo Compounds/pharmacokinetics , Pharmaceutical Preparations/metabolism , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , RNA Splicing/drug effects , RNA, Messenger/metabolism , Tissue Distribution , Animals , Humans , In Vitro Techniques , Survival of Motor Neuron 2 Protein/metabolism
2.
Br J Clin Pharmacol ; 87(9): 3550-3560, 2021 09.
Article in English | MEDLINE | ID: mdl-33576513

ABSTRACT

AIMS: RO5459072, a cathepsin-S inhibitor, Biopharmaceutics Classification System class 2 and P-glycoprotein substrate, exhibited complex, nonlinear pharmacokinetics (PK) while fasted that seemed to impact both the absorption and the disposition phases. When given with food, all nonlinearities disappeared. Physiologically based PK (PBPK) modelling attributed those nonlinearities to dose-dependent solubilisation and colonic absorption. The objective of this population PK analysis was to complement the PBPK analysis. METHODS: PK profiles in 39 healthy volunteers after first oral dosing (1-600 mg) while fasted or fed in single and multiple ascending dose studies were analysed using population compartmental modelling. RESULTS: The PK of RO5459072 while fed was characterized by a 1-compartmental PK model with linear absorption and elimination. The nonlinearities while fasted were captured using dose dependent bioavailability and 2 sequential first-order absorption phases: one following drug administration and one occurring 11 hours later and only for doses >10 mg. The bioavailability in the first absorption phase increased between 1 and 10 mg and then decreased with dose, in agreement with in vitro dissolution and solubility studies. The remaining fraction of doses to be absorbed by the second absorption phase was found to have a bioavailability similar to that in the first absorption phase. CONCLUSION: The population PK model supported that dissolution- and solubility-limited absorption from the proximal and distal intestine alone explains the nonlinear PK of RO5459072 in fasted state and the linear PK in fed state. This work, together with the PBPK analysis, raised our confidence in the understanding of this complex PK.


Subject(s)
Food-Drug Interactions , Pharmaceutical Preparations , Administration, Oral , Humans , Intestinal Absorption , Models, Biological , Pyrazoles , Pyrrolidines , Solubility , Water
3.
Sci Rep ; 9(1): 4074, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858459

ABSTRACT

As more and more protein biotherapeutics enter the drug discovery pipelines, there is an increasing interest in tools for mechanistic drug metabolism investigations of biologics in order to identify and prioritize the most promising candidates. Understanding or even predicting the in vivo clearance of biologics and to support translational pharmacokinetic modeling activities is essential, however there is a lack of effective and validated in vitro cellular tools. Although different mechanisms have to be adressed in the context of biologics disposition, the scope is not comparable to the nowadays widely established tools for early characterization of small molecule disposition. Here, we describe a biotransformation study of the fusion protein tetranectin apolipoprotein A1 by cellular systems. The in vivo biotransformation of tetranectin apolipoprotein A1 has been described previously, and the same major biotransformation product could also be detected in vitro, by a targeted and highly sensitive detection method based on chymotrypsin digest. In addition, the protease responsible for the formation of this biotransformation product could be elucidated to be DPP4. To our knowledge, this is one of the first reports of an in vitro biotransformation study by cells of a therapeutic protein.


Subject(s)
Apolipoprotein A-I/genetics , Biotransformation/genetics , Dipeptidyl Peptidase 4/chemistry , Lectins, C-Type/genetics , Recombinant Fusion Proteins/genetics , Apolipoprotein A-I/chemistry , Chymotrypsin/pharmacology , Dipeptidyl Peptidase 4/pharmacology , Drug Discovery , Humans , Lectins, C-Type/chemistry , Protein Processing, Post-Translational , Proteomics/methods , Recombinant Fusion Proteins/chemistry
4.
PLoS One ; 13(10): e0205435, 2018.
Article in English | MEDLINE | ID: mdl-30332475

ABSTRACT

New therapeutic biological entities such as bispecific antibodies targeting tissue or specific cell populations form an increasingly important part of the drug development portfolio. However, these biopharmaceutical agents bear the risk of extensive target-mediated drug disposition or atypical pharmacokinetic properties as compared to canonical antibodies. Pharmacokinetics and bio-distribution studies become therefore more and more important during lead optimization. Biologics present, however, greater analytical challenges than small molecule drugs due to the mass and selectivity limitation of mass spectrometry and ligand-binding assay, respectively. Radiocarbon (14C) and its detection methods, such as the emerging 14C cavity ring down spectroscopy (CRDS), thus can play an important role in the large molecule quantitation where a 14C-tag is covalently bound through a stable linker. CRDS has the advantage of a simplified sample preparation and introduction system as compared to accelerator mass spectrometry (AMS) and can be accommodated within an ordinary research laboratory. In this study, we report on the labeling of an anti-IL17 IgG1 model antibody with 14C propionate tag and its detection by CRDS using it as nanotracer (2.1 nCi or 77.7 Bq blended with the therapeutic dose) in a pharmacokinetics study in a preclinical species. We compare these data to data generated by AMS in parallel processed samples. The derived concentration time profiles for anti-IL17 by CRDS were in concordance with the ones derived by AMS and γ-counting of an 125I-labeled anti-IL17 radiotracer and were well described by a 2-compartment population pharmacokinetic model. In addition, antibody tissue distribution coefficients for anti-IL17 were determined by CRDS, which proved to be a direct and sensitive measurement of the extravascular tissue concentration of the antibody when tissue perfusion was applied. Thus, this proof-of-concept study demonstrates that trace 14C-radiolabels and CRDS are an ultrasensitive approach in (pre)clinical pharmacokinetics and bio-distribution studies of new therapeutic entities.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Interleukin-17/antagonists & inhibitors , Carbon Radioisotopes , Humans , Iodine Radioisotopes , Mass Spectrometry , Spectrum Analysis , Tissue Distribution
5.
J Pharmacol Exp Ther ; 365(2): 237-248, 2018 05.
Article in English | MEDLINE | ID: mdl-29453199

ABSTRACT

Long-term in vitro liver models are now widely explored for human hepatic metabolic clearance prediction, enzyme phenotyping, cross-species metabolism, comparison of low clearance drugs, and induction studies. Here, we present studies using a long-term liver model, which show how metabolism and active transport, drug-drug interactions, and enzyme induction in healthy and diseased states, such as hepatitis B virus (HBV) infection, may be assessed in a single test system to enable effective data integration for physiologically based pharmacokinetic (PBPK) modeling. The approach is exemplified in the case of (3S)-4-[[(4R)-4-(2-Chloro-4-fluorophenyl)-5-methoxycarbonyl-2-thiazol-2-yl-1,4-dihydropyrimidin-6-yl]methyl]morpholine-3-carboxylic acid RO6889678, a novel inhibitor of HBV with a complex absorption, distribution, metabolism, and excretion (ADME) profile. RO6889678 showed an intracellular enrichment of 78-fold in hepatocytes, with an apparent intrinsic clearance of 5.2 µl/min per mg protein and uptake and biliary clearances of 2.6 and 1.6 µl/min per mg protein, respectively. When apparent intrinsic clearance was incorporated into a PBPK model, the simulated oral human profiles were in good agreement with observed data at low doses but were underestimated at high doses due to unexpected overproportional increases in exposure with dose. In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. Furthermore, we report on the first evaluation of in vitro pharmacokinetics studies using HBV-infected HepatoPac cocultures. Thus, long-term liver models have great potential as translational research tools exploring pharmacokinetics of novel drugs in vitro in health and disease.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Liver/metabolism , Antiviral Agents/pharmacokinetics , Biological Transport , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Hepatocytes/metabolism , Humans , Kinetics , Liver/drug effects , Time Factors , Tissue Distribution
6.
AAPS J ; 19(2): 534-550, 2017 03.
Article in English | MEDLINE | ID: mdl-28050713

ABSTRACT

Early prediction of human clearance is often challenging, in particular for the growing number of low-clearance compounds. Long-term in vitro models have been developed which enable sophisticated hepatic drug disposition studies and improved clearance predictions. Here, the cell line HepG2, iPSC-derived hepatocytes (iCell®), the hepatic stem cell line HepaRG™, and human hepatocyte co-cultures (HµREL™ and HepatoPac®) were compared to primary hepatocyte suspension cultures with respect to their key metabolic activities. Similar metabolic activities were found for the long-term models HepaRG™, HµREL™, and HepatoPac® and the short-term suspension cultures when averaged across all 11 enzyme markers, although differences were seen in the activities of CYP2D6 and non-CYP enzymes. For iCell® and HepG2, the metabolic activity was more than tenfold lower. The micropatterned HepatoPac® model was further evaluated with respect to clearance prediction. To assess the in vitro parameters, pharmacokinetic modeling was applied. The determination of intrinsic clearance by nonlinear mixed-effects modeling in a long-term model significantly increased the confidence in the parameter estimation and extended the sensitive range towards 3% of liver blood flow, i.e., >10-fold lower as compared to suspension cultures. For in vitro to in vivo extrapolation, the well-stirred model was used. The micropatterned model gave rise to clearance prediction in man within a twofold error for the majority of low-clearance compounds. Further research is needed to understand whether transporter activity and drug metabolism by non-CYP enzymes, such as UGTs, SULTs, AO, and FMO, is comparable to the in vivo situation in these long-term culture models.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , Models, Biological , Pharmacokinetics , Coculture Techniques , Cytochrome P-450 CYP2D6/metabolism , Enzymes/metabolism , Hep G2 Cells , Hepatocytes/enzymology , Humans , Liver/enzymology , Nonlinear Dynamics , Pharmaceutical Preparations/metabolism , Time Factors
7.
ChemMedChem ; 11(19): 2216-2239, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27629993

ABSTRACT

The modulation of pharmacologically relevant properties of N-alkyl-piperidine-2-carboxamides was studied by selective introduction of 1-3 fluorine atoms into the n-propyl and n-butyl side chains of the local anesthetics ropivacaine and levobupivacaine. The basicity modulation by nearby fluorine substituents is essentially additive and exhibits an exponential attenuation as a function of topological distance between fluorine and the basic center. The intrinsic lipophilicity of the neutral piperidine derivatives displays the characteristic response noted for partially fluorinated alkyl groups attached to neutral heteroaryl systems. However, basicity decrease by nearby fluorine substituents affects lipophilicities at neutral pH, so that all partially fluorinated derivatives are of similar or higher lipophilicity than their non-fluorinated parents. Aqueous solubilities were found to correlate inversely with lipophilicity with a significant contribution from crystal packing energies, as indicated by variations in melting point temperatures. All fluorinated derivatives were found to be somewhat more readily oxidized in human liver microsomes, the rates of degradation correlating with increasing lipophilicity. Because the piperidine-2-carboxamide core is chiral, pairs with enantiomeric N-alkyl groups are diastereomeric. While little response to such stereoisomerism was observed for basicity or lipophilicity, more pronounced variations were observed for melting point temperatures and oxidative degradation.


Subject(s)
Piperidines/chemistry , Piperidines/pharmacology , Dose-Response Relationship, Drug , Halogenation , Humans , Hydrophobic and Hydrophilic Interactions , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Piperidines/chemical synthesis , Piperidines/metabolism , Structure-Activity Relationship , Temperature
9.
Pharm Res ; 33(10): 2565-79, 2016 10.
Article in English | MEDLINE | ID: mdl-27469324

ABSTRACT

PURPOSE: This study aims to expand our understanding of the mechanisms of drug absorption, distribution, metabolism and excretion in the Göttingen minipig to aid a knowledge-driven selection of the optimal species for preclinical pharmaceutical research. METHODS: The pharmacokinetics of seven reference compounds (antipyrine, atenolol, cimetidine, diazepam, hydrochlorothiazide, midazolam and theophylline) was investigated after intravenous and oral dosing in minipigs. Supportive in vitro data were generated on hepatocellularity, metabolic clearance in hepatocytes, blood cell and plasma protein binding and metabolism routes. RESULTS: Systemic plasma clearance for the seven drugs ranged from low (1.1 ml/min/kg, theophylline) to close to liver blood flow (37.4 ml/min/kg, cimetidine). Volume of distribution in minipigs ranged from 0.7 L/kg for antipyrine to 3.2 L/kg for hydrochlorothiazide. A gender-related difference of in vivo metabolic clearance was observed for antipyrine. The hepatocellularity for minipig was determined as 124 Mcells/g liver, similar to the values reported for human. Based on these data a preliminary in vitro to in vivo correlation (IVIVC) for metabolic clearance measured in hepatocytes was investigated. Metabolite profiles of diazepam and midazolam compared well between minipig and human. CONCLUSIONS: The results of the present study support the use of in vitro metabolism data for the evaluation of minipig in preclinical research and safety testing.


Subject(s)
Hepatocytes/metabolism , Models, Animal , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Animals , Female , Hepatocytes/drug effects , Humans , Male , Protein Binding/physiology , Species Specificity , Swine , Swine, Miniature
10.
BMC Genomics ; 16: 932, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26573612

ABSTRACT

BACKGROUND: In the past decade the Göttingen minipig has gained increasing recognition as animal model in pharmaceutical and safety research because it recapitulates many aspects of human physiology and metabolism. Genome-based comparison of drug targets together with quantitative tissue expression analysis allows rational prediction of pharmacology and cross-reactivity of human drugs in animal models thereby improving drug attrition which is an important challenge in the process of drug development. RESULTS: Here we present a new chromosome level based version of the Göttingen minipig genome together with a comparative transcriptional analysis of tissues with pharmaceutical relevance as basis for translational research. We relied on mapping and assembly of WGS (whole-genome-shotgun sequencing) derived reads to the reference genome of the Duroc pig and predict 19,228 human orthologous protein-coding genes. Genome-based prediction of the sequence of human drug targets enables the prediction of drug cross-reactivity based on conservation of binding sites. We further support the finding that the genome of Sus scrofa contains about ten-times less pseudogenized genes compared to other vertebrates. Among the functional human orthologs of these minipig pseudogenes we found HEPN1, a putative tumor suppressor gene. The genomes of Sus scrofa, the Tibetan boar, the African Bushpig, and the Warthog show sequence conservation of all inactivating HEPN1 mutations suggesting disruption before the evolutionary split of these pig species. We identify 133 Sus scrofa specific, conserved long non-coding RNAs (lncRNAs) in the minipig genome and show that these transcripts are highly conserved in the African pigs and the Tibetan boar suggesting functional significance. Using a new minipig specific microarray we show high conservation of gene expression signatures in 13 tissues with biomedical relevance between humans and adult minipigs. We underline this relationship for minipig and human liver where we could demonstrate similar expression levels for most phase I drug-metabolizing enzymes. Higher expression levels and metabolic activities were found for FMO1, AKR/CRs and for phase II drug metabolizing enzymes in minipig as compared to human. The variability of gene expression in equivalent human and minipig tissues is considerably higher in minipig organs, which is important for study design in case a human target belongs to this variable category in the minipig. The first analysis of gene expression in multiple tissues during development from young to adult shows that the majority of transcriptional programs are concluded four weeks after birth. This finding is in line with the advanced state of human postnatal organ development at comparative age categories and further supports the minipig as model for pediatric drug safety studies. CONCLUSIONS: Genome based assessment of sequence conservation combined with gene expression data in several tissues improves the translational value of the minipig for human drug development. The genome and gene expression data presented here are important resources for researchers using the minipig as model for biomedical research or commercial breeding. Potential impact of our data for comparative genomics, translational research, and experimental medicine are discussed.


Subject(s)
Genome , Swine, Miniature/genetics , Aging/genetics , Animals , Chromosomes , Gene Expression , Gene Expression Profiling , Humans , Liver/metabolism , Pharmaceutical Preparations/metabolism , Pseudogenes , Species Specificity , Swine , Transcription, Genetic
11.
J Med Chem ; 58(22): 9041-60, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26523333

ABSTRACT

The synthesis of a collection of 3-substituted indole derivatives incorporating partially fluorinated n-propyl and n-butyl groups is described along with an in-depth study of the effects of various fluorination patterns on their properties, such as lipophilicity, aqueous solubility, and metabolic stability. The experimental observations confirm predictions of a marked lipophilicity decrease imparted by a vic-difluoro unit when compared to the gem-difluoro counterparts. The data involving the comparison of the two substitution patterns is expected to benefit molecular design in medicinal chemistry and, more broadly, in life as well as materials sciences.


Subject(s)
Drug Discovery/methods , Fluorine Compounds/chemical synthesis , Fluorine Compounds/pharmacology , Animals , Biotransformation , Chemistry, Physical , Drug Design , Fluorine Compounds/pharmacokinetics , Halogenation , Humans , In Vitro Techniques , Lipids/chemistry , Microsomes, Liver/metabolism , Protein Structure, Tertiary , Rats , Solubility , Structure-Activity Relationship , Thermodynamics
12.
J Pharmacokinet Pharmacodyn ; 42(3): 275-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25822652

ABSTRACT

Real time cell analysis (RTCA) is an impedance-based technology which tracks various living cell characteristics over time, such as their number, morphology or adhesion to the extra cellular matrix. However, there is no consensus about how RTCA data should be used to quantitatively evaluate pharmacodynamic parameters which describe drug efficacy or toxicity. The purpose of this work was to determine how RTCA data can be analyzed with mathematical modeling to explore and quantify drug effect in vitro. The pharmacokinetic-pharmacodynamic erlotinib concentration profile predicted by the model and its effect on the human epidermoïd carcinoma cell line A431 in vitro was measured through RTCA output, designated as cell index. A population approach was used to estimate model parameter values, considering a plate well as the statistical unit. The model related the cell index to the number of cells by means of a proportionality factor. Cell growth was described by an exponential model. A delay between erlotinib pharmacokinetics and cell killing was described by a transit compartment model, and the effect potency, by an E max function of erlotinib concentration. The modeling analysis performed on RTCA data distinguished drug effects in vitro on cell number from other effects likely to modify the relationship between cell index and cell number. It also revealed a time-dependent decrease of erlotinib concentration over time, described by a mono-exponential pharmacokinetic model with nonspecific binding.


Subject(s)
Computer Systems , Erlotinib Hydrochloride/pharmacokinetics , Models, Biological , Protein Kinase Inhibitors/pharmacokinetics , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Humans
13.
Curr Top Med Chem ; 11(15): 1902-24, 2011.
Article in English | MEDLINE | ID: mdl-21470172

ABSTRACT

G protein-coupled receptors (GPCRs) share a common architecture consisting of seven transmembrane (TM) domains. Various lines of evidence suggest that this fold provides a generic binding pocket within the TM region for hosting agonists, antagonists, and allosteric modulators. Hence, an automated method was developed that allows a fast analysis and comparison of these generic ligand binding pockets across the entire GPCR family by providing the relevant information for all GPCRs in the same format. This methodology compiles amino acids lining the TM binding pocket including parts of the ECL2 loop in a so-called 1D ligand binding pocket vector and translates these 1D vectors in a second step into 3D receptor pharmacophore models. It aims to support various aspects of GPCR drug discovery in the pharmaceutical industry. Applications of pharmacophore similarity analysis of these 1D LPVs include definition of receptor subfamilies, prediction of species differences within subfamilies in regard to in vitro pharmacology and identification of nearest neighbors for GPCRs of interest to generate starting points for GPCR lead identification programs. These aspects of GPCR research are exemplified in the field of melanopsins, trace amine-associated receptors and somatostatin receptor subtype 5. In addition, it is demonstrated how 3D pharmacophore models of the LPVs can support the prediction of amino acids involved in ligand recognition, the understanding of mutational data in a 3D context and the elucidation of binding modes for GPCR ligands and their evaluation. Furthermore, guidance through 3D receptor pharmacophore modeling for the synthesis of subtype-specific GPCR ligands will be reported. Illustrative examples are taken from the GPCR family class C, metabotropic glutamate receptors 1 and 5 and sweet taste receptors, and from the GPCR class A, e.g. nicotinic acid and 5-hydroxytryptamine 5A receptor.


Subject(s)
Drug Discovery/methods , Receptors, G-Protein-Coupled/chemistry , Amino Acid Sequence , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
14.
Bioorg Med Chem Lett ; 20(18): 5426-30, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20724150

ABSTRACT

Pyrido pyrimidinones are selective agonists of the human high affinity niacin receptor GPR109A (HM74A). They show no activity on the highly homologous low affinity receptor GPR109B (HM74). Starting from a high throughput screening hit the in vitro activity of the pyrido pyrimidinones was significantly improved providing lead compounds suitable for further optimization.


Subject(s)
Niacin/metabolism , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Animals , Microsomes, Liver/metabolism , Pyrimidinones/administration & dosage , Pyrimidinones/metabolism , Rats , Rats, Wistar , Structure-Activity Relationship
15.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(23): 2101-5, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20627230

ABSTRACT

This manuscript describes the determination of the nociceptin/orphanin FQ peptide (NOP) receptor agonist RO0646198 in rat and Cynomolgus monkey plasma using liquid chromatography coupled to tandem mass spectrometry. The structural analogue RO0658791 served as internal standard. After protein precipitation with ethanol, clean up and enrichment was carried out by on-line solid-phase extraction on a 10mm C18 trapping column. Gradient separation was performed on a 50 mm C18 analytical column, followed by electrospray ionization and detection in positive ion selected reaction monitoring (SRM) mode. The total run time was 2.5 min. The lower limit of quantification (LLOQ) was 10 pg/mL. Precisions were below 19% and accuracies were between 86% and 118%. The recoveries were above 90%, and no significant matrix effect was observed. The method was successfully applied to low dose pharmacokinetic studies. The bioavailability after oral dosing was below 5% for rats and below 1% in Cynomolgus monkeys, limiting the application of RO0646198 in the clinic to parenteral routes.


Subject(s)
Imidazoles/pharmacology , Imidazoles/pharmacokinetics , Macaca fascicularis/blood , Receptors, Opioid/agonists , Spectrometry, Mass, Electrospray Ionization/methods , Spiro Compounds/pharmacology , Spiro Compounds/pharmacokinetics , Animals , Drug Administration Routes , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Rats , Rats, Wistar , Reference Standards , Spiro Compounds/administration & dosage , Spiro Compounds/blood , Time Factors , Nociceptin Receptor
16.
J Med Chem ; 50(25): 6291-4, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18020390

ABSTRACT

We disclose the first selective, nonpeptidic, small-molecule somatostatin receptor subtype 5 (SST5R) antagonists that were identified by a chemogenomics approach based on the analysis of the homology of amino acids defining the putative consensus drug binding site of SST5R. With this strategy, opioid, histamine, dopamine, and serotonine receptors were identified as the closest neighbors of SST5R. The H1 antagonist astemizole was chosen as a seed structure and subsequently transformed into a SST5 receptor antagonist with nanomolar binding affinity devoid of the original target activity.


Subject(s)
Benzoxazoles/chemical synthesis , Piperidines/chemical synthesis , Receptors, Somatostatin/antagonists & inhibitors , Amino Acid Sequence , Astemizole/chemistry , Astemizole/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Binding Sites , Histamine H1 Antagonists/chemistry , Histamine H1 Antagonists/pharmacology , Humans , Molecular Sequence Data , Piperidines/chemistry , Piperidines/pharmacology , Radioligand Assay , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Sequence Homology, Amino Acid , Structure-Activity Relationship
17.
J Med Chem ; 50(25): 6295-8, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18020391

ABSTRACT

The H1R antagonist astemizole was identified as a somatostatin 5 (SST5) receptor antagonist by a comparative sequence analysis of the consensus drug binding pocket of GPCRs. Subsequently, a similarity analysis of GPCR affinity profiles of astemizole versus a set of in-house GPCR-biased combinatorial libraries revealed new chemical entry points that led to a second lead series with nanomolar binding affinity.


Subject(s)
Astemizole/chemistry , Histamine H1 Antagonists/chemistry , Piperidines/chemical synthesis , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Somatostatin/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Astemizole/pharmacology , Binding Sites , Combinatorial Chemistry Techniques , Databases, Factual , Histamine H1 Antagonists/pharmacology , Humans , Piperidines/chemistry , Piperidines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Structure-Activity Relationship
18.
BMC Struct Biol ; 7: 66, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17935609

ABSTRACT

BACKGROUND: Differences in sweet taste perception among species depend on structural variations of the sweet taste receptor. The commercially used isovanillyl sweetener neohesperidin dihydrochalcone activates the human but not the rat sweet receptor TAS1R2+TAS1R3. Analysis of interspecies combinations and chimeras of rat and human TAS1R2+TAS1R3 suggested that the heptahelical domain of human TAS1R3 is crucial for the activation of the sweet receptor by neohesperidin dihydrochalcone. RESULTS: By mutational analysis combined with functional studies and molecular modeling we identified a set of different amino acid residues within the heptahelical domain of human TAS1R3 that forms the neohesperidin dihydrochalcone binding pocket. Sixteen amino acid residues in the transmembrane domains 2 to 7 and one in the extracellular loop 2 of hTAS1R3 influenced the receptor's response to neohesperidin dihydrochalcone. Some of these seventeen residues are also part of the binding sites for the sweetener cyclamate or the sweet taste inhibitor lactisole. In line with this observation, lactisole inhibited activation of the sweet receptor by neohesperidin dihydrochalcone and cyclamate competitively, whereas receptor activation by aspartame, a sweetener known to bind to the N-terminal domain of TAS1R2, was allosterically inhibited. Seven of the amino acid positions crucial for activation of hTAS1R2+hTAS1R3 by neohesperidin dihydrochalcone are thought to play a role in the binding of allosteric modulators of other class C GPCRs, further supporting our model of the neohesperidin dihydrochalcone pharmacophore. CONCLUSION: From our data we conclude that we identified the neohesperidin dihydrochalcone binding site at the human sweet taste receptor, which overlaps with those for the sweetener cyclamate and the sweet taste inhibitor lactisole. This readily delivers a molecular explanation of our finding that lactisole is a competitive inhibitor of the receptor activation by neohesperidin dihydrochalcone and cyclamate. Some of the amino acid positions crucial for activation of hTAS1R2+hTAS1R3 by neohesperidin dihydrochalcone are involved in the binding of allosteric modulators in other class C GPCRs, suggesting a general role of these amino acid positions in allosterism and pointing to a common architecture of the heptahelical domains of class C GPCRs.


Subject(s)
Chalcones/chemistry , Chalcones/metabolism , Hesperidin/analogs & derivatives , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Benzene Derivatives , Binding Sites , Hesperidin/chemistry , Hesperidin/metabolism , Humans , Models, Theoretical , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Sequence Alignment
19.
J Neurochem ; 98(2): 601-15, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805850

ABSTRACT

Fenobam [N-(3-chlorophenyl)-N'-(4,5-dihydro-1-methyl-4-oxo-1H-imidazole-2-yl)urea], a clinically validated non-benzodiazepine anxiolytic, has been shown to be a potent and non-competitive metabotropic glutamate (mGlu)-5 receptor antagonist. In the present study, we have used the site-directed mutagenesis coupled with three-dimensional receptor-based pharmacophore modelling to elucidate the interacting mode of fenobam within the seven-transmembrane domain (7TMD) of mGlu5 receptor and its comparison with that of 2-methyl-6-(phenylethynyl)pyridine (MPEP), the prototype antagonist. The common residues involved in the recognition of MPEP and fenobam include Pro654(3.36), Tyr658(3.40), Thr780(6.44), Trp784(6.48), Phe787(6.51), Tyr791(6.55) and Ala809(7.47). The differentiating residues between both modulators' interacting modes are Arg647(3.29), Ser657(3.39) and Leu743(5.47). Our data suggest that these chemically unrelated mGlu5 antagonists act similarly, probing a functionally unique region of the 7TMD. Using [3H]inositol phosphates accumulation assay, we have also identified the critical residues involved in the inverse agonist effect of MPEP. The mutation W784(6.48)A completely blocked the inverse agonist activity of MPEP; two mutations F787(6.51)A and Y791(6.55)A, caused a drastic decrease in the MPEP inverse agonism. Furthermore, these three mutations led to an increased efficacy of quisqualate without having any effect on its potency. The fact that the residues Trp784(6.48) and Phe787(6.51) are essential equally in antagonism and inverse agonism effects emphasizes again the key role of these residues and the involvement of a common transmembrane network in receptor inactivation by MPEP.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Binding Sites , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Fluorometry , Humans , Imidazoles/metabolism , Inositol Phosphates/metabolism , Models, Molecular , Mutation/physiology , Plasmids , Quisqualic Acid/antagonists & inhibitors , Quisqualic Acid/pharmacology , Receptor, Metabotropic Glutamate 5 , Thiazoles/pharmacology
20.
Eur J Pharmacol ; 529(1-3): 95-104, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16352303

ABSTRACT

Several mutations in the seven-transmembrane region of rat metabotropic glutamate 5 (rmGlu5) receptors were produced by site-directed mutagenesis and expressed in CHO cells. Using functional intracellular calcium ([Ca(2+)](i)) mobilisation, we identified amino acids implicated in the positive allosteric modulation of quisqualate-induced response by 3,3'-difluorobenzaldazine (DFB). Human and rat mGlu5 receptors displayed a higher potency and a higher efficacy in the presence of DFB. Mutant receptors S657(3.39)C, T780(6.44)A and M801(7.39)T disrupted the DFB-mediated increase in functional response. DFB-induced increase in potency was abolished in mutant receptors N733(45.51)A, Y791(6.55)A, A809(7.47)V, P654(3.36)S/S657(3.39)C and P654(3.36)S/S657(3.39)C/L743(5.47)V without affecting the enhancement of efficacy observed in wild type receptors. Mutations at positions Leu-743(5.47) and Trp-784(6.48) resulted in significantly larger DFB-induced potentiation of EC(50) and E(max) values than in wild type receptors. DFB-mediated increase of efficacy was abolished and EC(50) values were right-shifted in mutant receptor F787A, resulting in DFB acting as a weak partial antagonist at this mutant receptor. Based on these findings, we constructed a homology model concluding that six key residues in transmembranes 3, 5, 6 and 7 are necessary for the allosteric modulation of rmGlu5a receptor by DFB. The model confirms an overlapping but distinct binding site to 2-methyl-6-(phenylethynyl)-pyridine (MPEP), and in particular emphasises the key role of W784 in transmembrane (TM) 6 for controlling the receptor's activation state.


Subject(s)
Hydrazines/pharmacology , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Rats , Receptor, Metabotropic Glutamate 5 , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...