Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Sci Rep ; 11(1): 7700, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833333

ABSTRACT

GABAA-α5 subunit-containing receptors have been shown to play a key modulatory role in cognition and represent a promising drug target for cognitive dysfunction, as well as other disorders. Here we report on the preclinical and early clinical profile of a novel GABAA-α5 selective negative allosteric modulator (NAM), basmisanil, which progressed into Phase II trials for intellectual disability in Down syndrome and cognitive impairment associated with schizophrenia. Preclinical pharmacology studies showed that basmisanil is the most selective GABAA-α5 receptor NAM described so far. Basmisanil bound to recombinant human GABAA-α5 receptors with 5 nM affinity and more than 90-fold selectivity versus α1, α2, and α3 subunit-containing receptors. Moreover, basmisanil inhibited GABA-induced currents at GABAA-α5 yet had little or no effect at the other receptor subtypes. An in vivo occupancy study in rats showed dose-dependent target engagement and was utilized to establish the plasma exposure to receptor occupancy relationship. At estimated receptor occupancies between 30 and 65% basmisanil attenuated diazepam-induced spatial learning impairment in rats (Morris water maze), improved executive function in non-human primates (object retrieval), without showing anxiogenic or proconvulsant effects in rats. During the Phase I open-label studies, basmisanil showed good safety and tolerability in healthy volunteers at maximum GABAA-α5 receptor occupancy as confirmed by PET analysis with the tracer [11C]-Ro 15-4513. An exploratory EEG study provided evidence for functional activity of basmisanil in human brain. Therefore, these preclinical and early clinical studies show that basmisanil has an ideal profile to investigate potential clinical benefits of GABAA-α5 receptor negative modulation.


Subject(s)
GABA-A Receptor Agonists/pharmacology , Receptors, GABA-A/drug effects , Allosteric Regulation , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Female , HEK293 Cells , Healthy Volunteers , Humans , Learning/drug effects , Macaca fascicularis , Positron-Emission Tomography , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis
2.
Neuropsychopharmacology ; 41(5): 1319-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26354045

ABSTRACT

A growing body of evidence indicates that neuronal oscillations in the gamma frequency range (30-80 Hz) are disturbed in schizophrenic patients during cognitive processes and may represent an endophenotype of the disease. N-methyl-D-aspartate (NMDA) receptor antagonists have been used experimentally to induce schizophrenia-like symptoms including cognitive deficits in animals and humans. Here we characterized neuronal oscillations and event-related potentials (ERPs) in Cynomolgus macaques fully trained to perform a continuous performance test (CPT) in the presence and absence of the NMDA antagonist phencyclidine (PCP). Macaques (n=8) were trained to touch 'target' stimuli and ignore 'distractor' stimuli presented randomly on a touchscreen. Subsequently, all subjects were implanted with epidural EEG electrodes over frontal (FC) and parietal cortices (PC) and later tested under vehicle (saline, i.m.) or acute PCP (0.1-0.3 mg/kg, i.m.) conditions. Compared with vehicle treatment, PCP produced a significant dose-dependent decrease in CPT performance accuracy and increased reaction times. Furthermore, PCP elevated the amplitudes of 'low' (30-50 Hz) and 'high' (51-80 Hz) gamma oscillations in FC and PC around target presentations for all correct responses. The CPT accuracy was inversely correlated with the gamma band amplitude in the presence of PCP. Additionally, PCP delayed the N100 peak latency in FC, and prolonged and suppressed the cognitively relevant P300 component of mean ERPs in FC and PC, respectively. The NMDA receptor antagonist-induced alteration in neuronal oscillations and ERPs may contribute to the observed cognitive deficits in macaques, and enhance our understanding of EEG recordings as a translatable biomarker.


Subject(s)
Attention/physiology , Frontal Lobe/physiology , Gamma Rhythm , Parietal Lobe/physiology , Phencyclidine/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/physiopathology , Animals , Attention/drug effects , Dose-Response Relationship, Drug , Electroencephalography , Excitatory Amino Acid Antagonists/administration & dosage , Frontal Lobe/drug effects , Gamma Rhythm/drug effects , Macaca fascicularis , Male , Parietal Lobe/drug effects
3.
Proc Natl Acad Sci U S A ; 108(20): 8485-90, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21525407

ABSTRACT

The trace amine-associated receptor 1 (TAAR1), activated by endogenous metabolites of amino acids like the trace amines p-tyramine and ß-phenylethylamine, has proven to be an important modulator of the dopaminergic system and is considered a promising target for the treatment of neuropsychiatric disorders. To decipher the brain functions of TAAR1, a selective TAAR1 agonist, RO5166017, was engineered. RO5166017 showed high affinity and potent functional activity at mouse, rat, cynomolgus monkey, and human TAAR1 stably expressed in HEK293 cells as well as high selectivity vs. other targets. In mouse brain slices, RO5166017 inhibited the firing frequency of dopaminergic and serotonergic neurons in regions where Taar1 is expressed (i.e., the ventral tegmental area and dorsal raphe nucleus, respectively). In contrast, RO5166017 did not change the firing frequency of noradrenergic neurons in the locus coeruleus, an area devoid of Taar1 expression. Furthermore, modulation of TAAR1 activity altered the desensitization rate and agonist potency at 5-HT(1A) receptors in the dorsal raphe, suggesting that TAAR1 modulates not only dopaminergic but also serotonergic neurotransmission. In WT but not Taar1(-/-) mice, RO5166017 prevented stress-induced hyperthermia and blocked dopamine-dependent hyperlocomotion in cocaine-treated and dopamine transporter knockout mice as well as hyperactivity induced by an NMDA antagonist. These results tie TAAR1 to the control of monoamine-driven behaviors and suggest anxiolytic- and antipsychotic-like properties for agonists such as RO5166017, opening treatment opportunities for psychiatric disorders.


Subject(s)
Biogenic Monoamines/metabolism , Receptors, G-Protein-Coupled/metabolism , Synaptic Transmission/physiology , Animals , Benzodioxoles/pharmacology , Dopamine/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Mental Disorders , Mice , Phenylpropionates/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency
4.
Proc Natl Acad Sci U S A ; 106(47): 20081-6, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19892733

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) that is nonselectively activated by endogenous metabolites of amino acids. TAAR1 is considered a promising drug target for the treatment of psychiatric and neurodegenerative disorders. However, no selective ligand to identify TAAR1-specific signaling mechanisms is available yet. Here we report a selective TAAR1 antagonist, EPPTB, and characterize its physiological effects at dopamine (DA) neurons of the ventral tegmental area (VTA). We show that EPPTB prevents the reduction of the firing frequency of DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 agonist. When applied alone, EPPTB increases the firing frequency of DA neurons, suggesting that TAAR1 either exhibits constitutive activity or is tonically activated by ambient levels of endogenous agonist(s). We further show that EPPTB blocks the TAAR1-mediated activation of an inwardly rectifying K(+) current. When applied alone, EPPTB induces an apparent inward current, suggesting the closure of tonically activated K(+) channels. Importantly, these EPPTB effects were absent in Taar1 knockout mice, ruling out off-target effects. We additionally found that both the acute application of EPPTB and the constitutive genetic lack of TAAR1 increase the potency of DA at D2 receptors in DA neurons. In summary, our data support that TAAR1 tonically activates inwardly rectifying K(+) channels, which reduces the basal firing frequency of DA neurons in the VTA. We hypothesize that the EPPTB-induced increase in the potency of DA at D2 receptors is part of a homeostatic feedback mechanism compensating for the lack of inhibitory TAAR1 tone.


Subject(s)
Benzamides/chemistry , Benzamides/metabolism , Dopamine/metabolism , Limbic System , Neurons/metabolism , Pyrrolidines/chemistry , Pyrrolidines/metabolism , Receptors, G-Protein-Coupled , Ventral Tegmental Area , Action Potentials/physiology , Animals , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Limbic System/cytology , Limbic System/metabolism , Mice , Mice, Knockout , Molecular Structure , Neurons/cytology , Oocytes/cytology , Oocytes/physiology , Patch-Clamp Techniques , Rats , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism , Xenopus laevis
5.
Bioorg Med Chem Lett ; 19(19): 5746-52, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19726184

ABSTRACT

Through iterative design cycles we have discovered a number of novel new classes where the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine was deemed the most promising GABA(A) alpha5 inverse agonist class with potential for cognitive enhancement. This class combines a modest subtype binding selectivity with inverse agonism and has the most favourable molecular properties for further lead optimisation towards a central nervous system (CNS) acting medicine.


Subject(s)
Benzodiazepines/chemistry , Nootropic Agents/chemistry , Receptors, GABA-A/metabolism , Triazoles/chemistry , Animals , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Drug Discovery , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Nootropic Agents/chemical synthesis , Nootropic Agents/pharmacology , Oocytes/drug effects , Triazoles/chemical synthesis , Triazoles/pharmacology , Xenopus laevis
6.
Bioorg Med Chem Lett ; 19(20): 5940-4, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19762240

ABSTRACT

Lead optimisation of the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine class led to the identification of two clinical leads [RO4882224 (11) and RO4938581 (44)] functioning as novel potent and selective GABAA alpha5 inverse agonists. The unique pharmacological profiles and optimal pharmacokinetic profiles resulted in in vivo activity in selected cognition models.


Subject(s)
Anticonvulsants/pharmacokinetics , Benzodiazepines/pharmacokinetics , Cognition Disorders/drug therapy , Imidazoles/pharmacokinetics , Receptors, GABA-A/metabolism , Triazoles/pharmacokinetics , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Benzodiazepines/chemical synthesis , Benzodiazepines/chemistry , Cell Line , Disease Models, Animal , Drug Discovery , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice , Protein Binding , Rats , Seizures/chemically induced , Seizures/drug therapy , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
7.
Bioorg Med Chem Lett ; 19(20): 5958-61, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19740657

ABSTRACT

In a search for GABAA alpha5 ligands that combine high subtype binding selectivity with a marked inverse agonism imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepines were identified as a promising class. A short tandem reaction allowed rapid access to this chemical series, thereby facilitating rapid SAR generation which guided the optimization process. Two compounds (10e and 11f) were found to be active in an in vivo paradigm for cognitive improvement.


Subject(s)
Anticonvulsants/chemistry , Benzodiazepines/chemistry , Cognition Disorders/drug therapy , Receptors, GABA-A/metabolism , Triazoles/chemistry , Adjuvants, Anesthesia/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacokinetics , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacokinetics , Benzodiazepines/pharmacology , Cell Line , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Memory, Short-Term/drug effects , Microsomes, Liver/metabolism , Rats , Scopolamine/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology
8.
Psychopharmacology (Berl) ; 202(1-3): 207-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18936916

ABSTRACT

RATIONALE: GABAA alpha5 subunit-containing receptors are primarily expressed in the hippocampus and their role in learning and memory has been demonstrated recently by both genetic and pharmacological approaches. OBJECTIVES: The objective of the study is to evaluate the cognitive effects of a novel GABAA alpha5 receptor inverse agonist, RO4938581 in rats and monkeys. MATERIALS AND METHODS: The in vitro profile was determined using radioligand binding and electrophysiological assays for the GABAA alpha1, alpha2, alpha3, and alpha5 receptors. Long-term potentiation (LTP) was performed in mouse hippocampal slices. Cognitive effects were assessed in rats in the delayed match to position (DMTP) task and the Morris water maze. In monkeys, the object retrieval task was used. Pro-convulsant and anxiogenic potentials were evaluated in mice and rats. In vivo receptor occupancy was determined using [3H]-RO0154513. RESULTS: RO4938581 is a potent inverse agonist at the GABAA alpha5 receptor, with both binding and functional selectivity, enhancing hippocampal LTP. RO4938581 reversed scopolamine-induced working memory impairment in the DMTP task (0.3-1 mg/kg p.o.) and diazepam-induced spatial learning impairment (1-10 mg/kg p.o.). RO4938581 improved executive function in monkeys (3-10 mg/kg p.o.). Importantly, RO4938581 showed no anxiogenic and pro-convulsive potential. RO4938581 dose-dependently bound to GABAA alpha5 receptors and approximately 30% receptor occupancy was sufficient to produce enhanced cognition in the rat. CONCLUSIONS: The data further support the potential of GABAA alpha5 receptors as a target for cognition-enhancing drugs. The dual binding and functional selectivity offers an ideal profile for cognition-enhancing effects without the unwanted side effects associated with activity at other GABAA receptor subtypes.


Subject(s)
Benzodiazepines/pharmacology , Cognition/drug effects , Imidazoles/pharmacology , Nootropic Agents/pharmacology , Receptors, GABA-A/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Haplorhini , Hippocampus/drug effects , In Vitro Techniques , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Membranes/drug effects , Memory/drug effects , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Patch-Clamp Techniques , Plasmids , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Recombinant Proteins , Seizures/chemically induced
9.
J Biomol Screen ; 10(4): 339-47, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15964935

ABSTRACT

The number of projects in drug development that fail in late phases because of cardiac side effects such as QT prolongation can impede drug discovery and development of projects. The molecular target responsible for QT prolongation by a wide range of pharmaceutical agents is the myocardial hERG potassium channel. It is therefore desirable to screen for compound interactions with the hERG channel at an early stage of drug development. Here, the authors report a cell-based fluorescence assay using membrane potential-sensitive fluorescent dyes and stably transfected hERG channels from CHO cells. The assay allows semiautomated screening of compounds for hERG activity on 384-well plates and is sufficiently rapid for testing a large number of compounds. The assay is robust as indicated by a Z' factor larger than 0.6. The throughput is in the range of 10,000 data points per day, which is significantly higher than any other method presently available for hERG. The data obtained with the fluorescence assay were in qualitative agreement with those from patch-clamp electrophysiological analysis. There were no false-positive hits, and the rate of false-negative compounds is currently 12% but might be further reduced by testing compounds at higher concentration. Quantitative differences between fluorescence and electrophysiological methods may be due to the use- or voltage-dependent activity of the antagonists.


Subject(s)
Potassium Channels, Voltage-Gated/antagonists & inhibitors , Spectrometry, Fluorescence/methods , Animals , CHO Cells , Cricetinae , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Fluorescent Dyes/chemistry , Patch-Clamp Techniques , Reproducibility of Results
10.
Bioorg Med Chem Lett ; 13(19): 3155-9, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-12951084

ABSTRACT

Screening of the Roche compound depository led to the identification of (1-benzyloxy-4,5-dihydro-1H-imidazol-2-yl)-butyl amine 4, a structurally novel NR1/2B subtype selective NMDA receptor antagonist. The structure-activity relationships developed in this series resulted in the discovery of a novel class of potent and selective NMDA receptor blockers displaying activity in vivo.


Subject(s)
Excitatory Amino Acid Antagonists/chemistry , Imidazoles/chemistry , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cells, Cultured , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Imidazoles/administration & dosage , Imidazoles/pharmacology , Injections, Intraperitoneal , Mice , Mice, Inbred DBA , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Seizures/prevention & control
11.
Bioorg Med Chem Lett ; 13(10): 1759-62, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12729659

ABSTRACT

A series of 4-(3,4-dihydro-1H-isoquinolin-2yl)-pyridines and analogous quinolines was prepared and evaluated as NR1/2B subtype selective NMDA receptor antagonists. 2-Hydroxyalkylamino substitution combines high affinity with selectivity (vs alpha1 and M1 receptors) and activity in vivo.


Subject(s)
Pyridines/chemical synthesis , Quinolines/chemical synthesis , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Mice , Pyridines/pharmacology , Quinolines/pharmacology , Seizures/drug therapy , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 13(5): 829-32, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12617901

ABSTRACT

Recently, we disclosed 4-aminoquinolines as structurally novel NR1/2B subtype selective NMDA receptor antagonists. We would now like to report our findings on structurally related pyridine analogues. The SAR developed in this series resulted in the discovery of high affinity antagonists which are selective (vs alpha1 and M1 receptors) and active in vivo.


Subject(s)
Isoquinolines/chemistry , Isoquinolines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Acoustic Stimulation , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , Hydrogen-Ion Concentration , Mice , Mice, Inbred DBA , Seizures/etiology , Seizures/prevention & control , Structure-Activity Relationship
13.
Neurobiol Dis ; 11(2): 298-307, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12505422

ABSTRACT

Guanidinoacetate methyltransferase (GAMT) deficiency is an autosomal recessively inherited disorder of creatine biosynthesis. The disease occurs in early life with developmental delay or arrest and several neurological symptoms, e.g., seizures and dyskinesia. Both the deficiency of high-energy phosphates in neurons and the neurotoxic action of the accumulated metabolite guanidinoacetate (GAA) are candidate mechanisms for the pathophysiology of this disease. To examine a potential role of GAA accumulation, we analyzed the electrophysiological responses of neurons induced by GAA application in primary culture and acute murine brain slices. GAA evoked picrotoxin- and bicuculline-sensitive GABA(A) receptor-mediated chloride currents with an EC(50) of 167 microM in cortical neurons. Pathophysiologically relevant GAA concentrations hyperpolarized globus pallidus neurons and reduced their spontaneous spike frequency with an EC(50) of 15.1 microM. Furthermore, GAA acted as a partial agonist at heterologously expressed GABA(A) but not GABA(B) receptors. The interaction of GAA with neuronal GABA(A) receptors represents a candidate mechanism explaining neurological dysfunction in GAMT deficiency.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Brain/metabolism , Creatine/deficiency , Glycine/analogs & derivatives , Glycine/metabolism , Methyltransferases/deficiency , Neurons/metabolism , Receptors, GABA-A/metabolism , Animals , Animals, Newborn , Binding Sites/drug effects , Binding Sites/physiology , Brain/physiopathology , Brain Diseases, Metabolic, Inborn/physiopathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CHO Cells , Chloride Channels/drug effects , Chloride Channels/metabolism , Creatine/biosynthesis , Cricetinae , Female , GABA Antagonists/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , GABA-B Receptor Agonists , GABA-B Receptor Antagonists , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Glycine/pharmacology , Guanidinoacetate N-Methyltransferase , Membrane Potentials/drug effects , Membrane Potentials/physiology , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , Neurons/drug effects , Oocytes , Receptors, GABA-B/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Sulfur Radioisotopes , Xenopus
14.
Bioorg Med Chem Lett ; 12(18): 2615-9, 2002 Sep 16.
Article in English | MEDLINE | ID: mdl-12182873

ABSTRACT

Screening of the Roche compound library led to the identification of 4-aminoquinoline 4 as structurally novel NR1/2B subtype selective NMDA receptor antagonist. The SAR which was developed in this series resulted in the discovery of highly potent and in vivo active blockers.


Subject(s)
Aminoquinolines/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Aminoquinolines/chemistry , Excitatory Amino Acid Antagonists/chemistry , Structure-Activity Relationship
15.
Chembiochem ; 3(5): 455-9, 2002 May 03.
Article in English | MEDLINE | ID: mdl-12007180

ABSTRACT

A computer-based method has been developed for prediction of the hERG (human ether-à-go-go related gene) K(+)-channel affinity of low molecular weight compounds. hERG channel blockage is a major concern in drug design, as such blocking agents can cause sudden cardiac death. Various techniques were applied to finding appropriate molecular descriptors for modeling structure-activity relationships: substructure analysis, self-organizing maps (SOM), principal component analysis (PCA), partial least squares fitting (PLS), and supervised neural networks. The most accurate prediction system was based on an artificial neural network. In a validation study, 93 % of the nonblocking agents and 71 % of the hERG channel blockers were correctly classified. This virtual screening method can be used for general compound-library shaping and combinatorial library design.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/chemistry , Trans-Activators , Combinatorial Chemistry Techniques , Databases, Factual , Drug Design , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Linear Models , Molecular Structure , Neural Networks, Computer , Nonlinear Dynamics , Structure-Activity Relationship , Transcriptional Regulator ERG
SELECTION OF CITATIONS
SEARCH DETAIL