WO1999029319A2 - Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds - Google Patents

Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds Download PDF

Info

Publication number
WO1999029319A2
WO1999029319A2 PCT/DK1998/000489 DK9800489W WO9929319A2 WO 1999029319 A2 WO1999029319 A2 WO 1999029319A2 DK 9800489 W DK9800489 W DK 9800489W WO 9929319 A2 WO9929319 A2 WO 9929319A2
Authority
WO
WIPO (PCT)
Prior art keywords
gaba
concentrations
chemical compound
anxiolytic
evoked
Prior art date
Application number
PCT/DK1998/000489
Other languages
French (fr)
Other versions
WO1999029319A3 (en
Inventor
Thomas Varming
Original Assignee
Neurosearch A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Neurosearch A/S filed Critical Neurosearch A/S
Priority to AU12244/99A priority Critical patent/AU1224499A/en
Publication of WO1999029319A2 publication Critical patent/WO1999029319A2/en
Publication of WO1999029319A3 publication Critical patent/WO1999029319A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70571Assays involving receptors, cell surface antigens or cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor

Definitions

  • This invention relates to a particular group of chemical compounds for use as non-sedative anxiolytic drugs, and a method for the identification of chemical compounds with anxiolytic potential.
  • GABA A receptors are members of a ligand-gated ion channel family, and are the most abundant inhibitory receptors in the mammalian brain.
  • GABA A receptor complex comprises a chloride ion channel that controls the chloride flux across the neuronal membrane, along with multiple recognition sites for small modulatory molecules such as benzodiazepines, barbiturates, picrotoxin, and certain steroids.
  • GABA interacts with its receptor, the ion channel is opened, the chloride flux is enhanced, and the cell becomes less responsive to excitatory stimuli.
  • This GABA induced ion current can be regulated by various agents, including agents that interact with the benzodiazepine receptor or recognition site.
  • benzodiazepine receptor agonists Agents that bind or interact with the modulatory sites on the GABA A receptor complex, e.g. the benzodiazepine receptor, and have a positive modulatory effect on the action of GABA, are called benzodiazepine receptor agonists or partial agonists.
  • Agonists generally produce muscle relaxant, hypnotic, sedative, anxiolytic, and/or anticonvulsant effects.
  • Benzodiazepine receptor ligands with negative modulatory effect on the action of GABA are termed inverse agonists, while benzodiazepine receptor ligands with no intrinsic activity are termed antagonists.
  • the present invention resides in the surprising discovery that a particular chemical compound, 5-acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime, in addition to being a potent benzodiazepine receptor agonist, is also characterised by a remarkable capability to effectively potentiate responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
  • the present invention provides chemical compounds for use as non- sedative anxiolytic drugs, and pharmaceutical compositions comprising these anxiolytic compounds.
  • the present invention provides methods for the screening of chemical compounds for anxiolytic potential. More specifically it is an object of the present invention to provide a convenient tool for the identification of benzodiazepine receptor agonists or partial agonists, capable of exerting a positive modulation of the GABAA receptor complex, which benzodiazepine receptor agonists or partial agonists are devoid the sedative effects often encountered with conventional benzodiazepine receptor agonists.
  • the invention relates to the use of a chemical compound for the manufacture of a pharmaceutical composition for the treatment of anxiolytic disorders, which chemical compound potentiate responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
  • the invention provides a method for the screening of chemical compounds for their potential as anxiolytic drugs, which method comprises the steps of determining, in the presence of different concentrations of GABA, the electrophysiological response evoked by GABA on a GABAA receptor; repeating the first step in the presence of a chemical compound to be screened; and selecting the chemical compounds that selectively potentiate responses evoked by GABA at lower concentrations.
  • the invention provides chemical compounds capable of potentiating responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA, and the use of these compounds as non-sedative anxiolytic drugs.
  • pharmaceutical compositions comprising the chemical compound of the invention, and methods of treating anxiolytic disorders, by which methods a mammal, including a human, is treated with a chemical compound of the invention.
  • the present invention resides in the surprising discovery that a particular chemical compound, 5-acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime, Compound 6 obtained as described in Ex. 1 (hereinafter referred to as Compound 6), in addition to being a potent benzodiazepine receptor agonist, is also characterised by a remarkable capability to potentiate the effect of GABA at low concentrations, while inhibiting the effect of GABA at higher concentrations. I.e. the effect of Compound 6 on a GABA evoked response was dependent on the GABA concentration used.
  • Compound 6 like the classical benzidoazepines, possesses a potentiating efficacy at lower concentrations of GABA, while this compound, in contrast to the classical benzidoazepines, more quickly looses its potentiating efficacy at slightly higher concentrations of GABA, and ultimately, at high GABA concentrations, becomes an inhibitor of the GABA response.
  • the ability of Compound 6 to act as an efficient potentiator at relative low stimulus intensity while loosing its efficacy at higher stimulus intensity is a very attractive property, and in this way Compound 6 is considered a representative for an entire group of chemical compounds that is particularly useful as non-sedative anxiolytic drugs.
  • the group of chemical compounds contemplated according to this invention distinguish from the classical anxiolytic compounds and thus represents a novel and distinct group of anxiolytic compounds.
  • the chemical compounds of the invention is characterised by being a chemical compound that, like the classical benzidoazepines, possesses a potentiating efficacy at lower concentrations of GABA, while, in contrast to the classical benzidoazepines, loosing its potentiating efficacy at higher concentrations of GABA, and ultimately, at very high GABA concentrations, becomes an inhibitor of the GABA response.
  • the chemical compounds of the invention may be characterised by being any chemical compound, apart from Compound 6, which is capable of potentiating responses evoked by concentrations of GABA of below about 5 ⁇ M, preferably below about 3 ⁇ M, while inhibiting responses evoked by concentrations of GABA of above about 3 ⁇ M, preferably above about 5 ⁇ M.
  • the electrophysiological response evoked by GABA on a GABA A receptor is determined at different concentrations of GABA and in the presence and absence of the compound to be tested.
  • the response evoked by GABA alone is compared to that recorded in the presence of the compound to be tested.
  • compounds capable of increasing the response evoked by GABA at lower concentrations of GABA are selected.
  • the method shall be carried out at concentrations of GABA in the range of from about 0.001 ⁇ M to about 10,000 ⁇ M, preferably in the range of from about 0.01 ⁇ M to about 1000 ⁇ M, more preferred in the range of from about 0.1 ⁇ M to about 500 ⁇ M, most preferred of from about 0.3 ⁇ M to about 300 ⁇ M.
  • chemical compounds that selectively potentiate responses evoked by GABA at concentrations lower than 10 ⁇ M, preferably lower than 5 ⁇ M, most preferred lower than 3 ⁇ M, are selected.
  • benzodiazepine receptor agonists or partial agonists are agents having a positive modulatory effect on the response on GABA A receptors evoked by the action of GABA.
  • a positive modulatory response evoked on GABA A receptors by the action of GABA may be measured using known electrophysiological methods, in particular the well known patch clamp technology.
  • the response evoked on GABA A receptors by the action of GABA may be measured using whole-cell patch clamp electrophysiological methods.
  • the whole-cell patch clamp method may be applied to any cell holding a GABAA receptor.
  • the cell applied in this method is a CHO cell, a HEK cell, or a HeLa cell.
  • GABAA Receptors are structurally constituted macromolecular heteropentameric assemblies, containing a combinations of , ⁇ , and ⁇ / ⁇ protein subunits.
  • Several subunits of such GABA A receptors ( ⁇ - 6 , ⁇ - 3 , ⁇ and ⁇ . 3 ) have been characterised using techniques of modern molecular biology.
  • the largest population (representing almost half of all GABA A receptors in the brain) is that containing the ⁇ 1 subunit in combination with the ⁇ 2 and ⁇ 2 subunits.
  • a receptor comprising an 1 , an ⁇ 2, an ⁇ 3, an o5, or an ⁇ 6 subunit; a ⁇ 1 , a ⁇ 2, or a ⁇ 3 subunit; and a ⁇ 2S or a ⁇ 2L subunit, or any combination hereof, is employed.
  • ⁇ GABAA receptor subunits, or ⁇ GABA A receptor subunits may be employed.
  • the use of co-expression of the GABA A receptor subunits ⁇ 1 , ⁇ 2 and ⁇ 2 is found to be very useful.
  • the ⁇ 1 ⁇ 2 ⁇ 2L; the ⁇ 1 ⁇ 3 ⁇ 2L; the ⁇ 2 ⁇ 2 ⁇ 2L; the cc2 ⁇ 3 ⁇ 2L; the ⁇ 3 ⁇ 2 ⁇ 2L; the ⁇ 3 ⁇ 3 ⁇ 2L; the ⁇ 5 ⁇ 2 ⁇ 2L; the oc5 ⁇ 3 ⁇ 2L; the ⁇ 6 ⁇ 2 ⁇ 2L; and the ⁇ 6 ⁇ 3 ⁇ 2L GABA A receptors are found to be useful.
  • the whole-cell patch clamp method is applied to a CHO cell expressing the oc1 ⁇ 2 ⁇ 2 GABAA receptor.
  • the invention relates to the use of the chemical compound of the invention for the manufacture of a pharmaceutical composition for the treatment of anxiolytic disorders.
  • anxiety disorders include generalised anxiety disorders, panic attacks, phobic disorders, obsessive-compulsive disorders, post-traumatic disorders, and mixed anxiety and depressive disorders.
  • the invention provides novel pharmaceutical compositions comprising a therapeutically effective amount of the chemical compound of the invention.
  • a chemical compound of the invention for use in therapy may be administered in the form of the raw chemical compound, it is preferred to introduce the active ingredient, optionally in the form of a physiologically acceptable salt, in a pharmaceutical composition together with one or more adjuvants, excipients, carriers, buffers, diluents, and/or other customary pharmaceutical auxiliaries.
  • the invention provides pharmaceutical compositions comprising the chemical compound of the invention, or a pharmaceutically acceptable salt or derivative thereof, together with one or more pharmaceutically acceptable carriers therefor, and, optionally, other therapeutic and/or prophylactic ingredients, know and used in the art.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not harmful to the recipient thereof.
  • the pharmaceutical composition of the invention may be administered by any convenient route which suite the desired therapy. Preferred routes of administration include oral administration, in particular in tablet, in capsule, in drage, in powder, or in liquid form, and parenteral administration, in particular cutaneous, subcutaneous, intramuscular, or intravenous injection.
  • the pharmaceutical composition may be prepared by the skilled person using standard and conventional techniques appropriate to the desired formulation.
  • the actual dosage depend on the nature and severity of the disease being treated, and is within the discretion of the physician, and may be varied by titration of the dosage to the particular circumstances of this invention to produce the desired therapeutic effect.
  • the pharmaceutical composition of the invention contain, as the active compound, of from about 1 to about 1000 mg of the chemical compound of the invention per individual dose.
  • Fig. 1 shows responses to 0.3, 3, 30 and 300 ⁇ M GABA, recorded from a CHO cell expressing GABA A 1 ⁇ 2 ⁇ 2 receptors;
  • Fig. 2 shows the full dose-response curve for GABA, recorded from a CHO cell expressing GABA A ⁇ 1 ⁇ 2 ⁇ 2 receptors (the EC 5 o-value was calculated to 18.8 ⁇ M and the Hill coefficient to 1.74);
  • Fig. 3 shows responses to 6 ⁇ M GABA, 1 ⁇ M GABA, and 10 ⁇ M GABA, recorded in the absence and the presence of 10 ⁇ M of a test compound (Compound 6 of Ex. 1), recorded from a CHO cell expressing GABAA ⁇ 1 ⁇ 2 ⁇ 2 receptors;
  • Fig. 4 shows the effect of 10 ⁇ M of a test compound (Compound 6 of Ex. 1 ) as a function of the GABA concentration, recorded from a CHO cell expressing GABA A ⁇ 1 ⁇ 2 ⁇ 2 receptors
  • Fig. 5 shows the effect of 1 ⁇ M of a test compound (Compound 6 of Ex. 1 ) as a function of the GABA concentration, and compared to 1 ⁇ M of a classical benzodiazepine agonist (DiazepamTM), recorded from a CHO cell expressing GABA A ⁇ 1 ⁇ 2 ⁇ 2 receptors
  • Fig. 6 shows the simultaneous effect of 10 ⁇ M of a test compound (Compound 6 of Ex.
  • DiazepamTM a classical benzodiazepine agonist
  • 3-(3-Pyridyl)aniline (Compound 2): A mixture of diethyl 3-pyridylborane (16.3 g, 0.11 mol), 3-bromoaniline (12.2 ml, 0.11 mol), potassium carbonate (45.8 g, 0.33 mol) and tetrakis(triphenylphosphine)palladium(0) (80 mg) in a mixture of water (40 ml) and dimethoxyethane (80 ml) is heated to 80°C under a stream of nitrogen over night. After cooling the mixture is diluted with water and ethyl acetate and filtered through a fluted filter paper. The layers are separated.
  • hGABA A subunits The cloning of hGABA A subunits was carried out as follows. Human brain hippocampus poly A + mRNA (Clontech) was reverse transcribed with oligo dT primer and MMLV reverse transcriptase (Pharmacia). 100 ng first strand cDNA was subjected to PCR using Expand HF polymerase (Boehringer Mannheim) and specific primer sets spanning the full length cDNA sequence. The primer sets were:
  • hGABA A ⁇ (s) 5OCTTAGCTGCTCCAGCCCGCGATGAGGA3'; (as) 5TTCCCAGTGCAGAGGAACTGAACAACAGAATG3';
  • hGABA A ⁇ 2 (s) 5 ⁇ AAACTAAAGGGATGTGGAGAGTGCGGAAAAG3'; (as) 5TCCAGTGGGAGGCCATGTTTTAGTTCACATAA3';
  • hGABA A ⁇ 2 (s) 5OTGCAACCAAGAGGCAAGAGGCGAGAG3'; and (as) 5'CCATATCAGTAAAACCCATACCTCCTCACAGG3'.
  • PCR conditions were 1) 94 °C 60 seconds; 2) 15 times (94 °C for 60 seconds, 55°C for 60 seconds, 72°C for 120 seconds); 3) 20 times (94 °C for 60 seconds, 55°C for 60 seconds, 72°C for 180 seconds); 4) 72°C for 10 minutes.
  • the amplified products were polished with pfu polymerase (Stratagene), purified on Quiaquick column (Qiagen) and subcloned into pCRScript (Stratagene). Maxiprep DNA (Qiagen) was prepared from positive clones and two to three individual clones of each subunits was sequenced bidirectionally on an ABI 377 sequencer using standard chain termination dyes (Perkin Elmer).
  • Subcloninq The ⁇ subunit was subcloned into pUbi2z (a custom designed mammalian expression vector using a human ubiquitin promoter to drive the inserted cDNA and the cDNA for enhanced green fluorescence protein (EGFP) fused to the selection marker conferring Zeocin resistance), the ⁇ 2 -subunit was subcloned into pNS1 n (a custom designed mammalian expression vector using a CMV promoter to drive the inserted cDNA and kanamycin/neomycin selection marker), and the ⁇ 2s - subunit was subcloned into pZeoSV (Invitrogen).
  • pUbi2z a custom designed mammalian expression vector using a human ubiquitin promoter to drive the inserted cDNA and the cDNA for enhanced green fluorescence protein (EGFP) fused to the selection marker conferring Zeocin resistance
  • EGFP enhanced green fluorescence protein
  • a single plasmid containing expression cassettes for both the ⁇ 2 subunit and the ⁇ 2s -subunit was constructed as follows.
  • the entire expression cassette for the ⁇ 2s -subunit in pZeoSV (SV40 promoter, cDNA, SV40-polyA) was excised as a BamHI fragment and subcloned into the Bglll site at position 1 in pNS1 n/hGABAA ⁇ 2-
  • a clones in which the driving promoters SV40 and CMV were pointing in opposite directions was selected.
  • Transfection For transient transfection experiments 1 ⁇ g of pUbi2z/ ⁇ was mixed with 1 ⁇ g of pNS1 n/ ⁇ 2 ⁇ 2S and transfected into Chinese hamster ovary cells (CHO, ATCC) using lipofectamine (Gibco) according to manufactures instructions. 24 hours after transfection, cells were plated into 35 mm Petri dishes containing 3.5 mm glass coverslips. Cells were kept at 37°C, in 5% C0 2 /95% air and used for electrophysiological studies 2-50 hours after plating.
  • Electrophysioloqy All experiments were performed in voltage clamp using conventional whole cell patch clamp methods. The following salt solutions were used: Extracellular solution (mM): NaCI (140), KCI (4), CaCI 2 (2), MgS0 4 (4),
  • the amplifier used was an EPC-9 (HEKA-electronics, Lambrect, Germany) run by a Macintosh 7600/120 computer via an ITC-16 interface. Experimental conditions were set with the Pulse- software accompanying the amplifier. Data were low pass filtered and sampled directly to hard-disk at a rate of 3 times the cut-off frequency.
  • Pipettes and electrodes Pipettes were pulled from borosilicate glass (Modulohm, Copenhagen, Denmark) using a horizontal electrode puller (Zeitz- Instrumente, Augsburg, Germany). The pipette resistance was 1.6 - 2.6 mW in the salt solutions used in these experiments.
  • the pipette electrode was a chloridised silver wire, and the reference was a silverchloride pellet electrode (In Vivo Metric, Healdsburg, USA) fixed to the experimental chamber. The electrodes were zeroed with the open pipette in the bath just prior to sealing.
  • the cells were held at a holding potential of -60 mV and at the start of each experiment the current was continuously measured for 20 sec to ensure a stable baseline.
  • GABA-containing solutions were delivered to the chamber through a custom- made gravity-driven flowpipe, the tip of which was placed approximately 50 ⁇ m from the cell.
  • Application was triggered by compressing the tubing connected to the flow pipe with a valve controlled by the Pulse-software.
  • GABA was applied for 4 sec. every 30 sec. The sampling interval during application was 1 msec.
  • the extracellular saline as well as the agonist containing solution were switched to solutions containing the compound (i.e. Compound 6 or Diazepam) to be tested. The compound was present until responses of a stable amplitude were achieved.
  • Fig. 3 the cell was voltage clamped at -60 mV and exposed to 4-sec pulses of 6 ⁇ M GABA every 20 sec. After assessment of the effect of Compound 6 (indicated by the horizontal bar, top panel), the GABA concentration was changed to 1 ⁇ M, and the effect of Compound 6 was evaluated (middle panel). Finally, the effect of Compound 6 on responses evoked by 10 ⁇ M GABA was evaluated (bottom) (please note the different scaling).
  • Fig. 5 the cells were voltage clamped at -60 mV and exposed to 4-sec pulses of GABA at 0.3-100 ⁇ M.
  • Compound 6 (1 ⁇ M, closed symbols) or Diazepam (1 ⁇ M, open symbols) was included in the bath-Ringer and in the GABA solutions.
  • Fig. 6 the cells were voltage clamped at -60 mV and exposed to 4-sec pulses of 0.3 ⁇ M GABA every 20 sec. After two control responses of the same size, Compound 6 (10 ⁇ M) and Diazepam (3 ⁇ M) were included in the solutions. After one minute (3 responses under the hatched bar), Diazepam was omitted from the solutions. After one minute (3 responses under the open bar only) solutions were changed back to control conditions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention relates to a particular group of chemical compounds for use as non-sedative anxiolytic drugs, and a method for the identification of chemical compounds with anxiolytic potential.

Description

CHEMICAL COMPOUNDS FOR USE AS ANXIOLYTIC AGENTS AND A METHOD FOR THE IDENTIFICATION OF ANXIOLYTIC COMPOUNDS
TECHNICAL FIELD
This invention relates to a particular group of chemical compounds for use as non-sedative anxiolytic drugs, and a method for the identification of chemical compounds with anxiolytic potential.
BACKGROUND ART
Receptors for the major inhibitory neurotransmitter, γ-aminobutyric acid (GABA), are divided into two main classes, GABAA receptors and GABAB receptors. GABAA receptors are members of a ligand-gated ion channel family, and are the most abundant inhibitory receptors in the mammalian brain. Each GABAA receptor complex comprises a chloride ion channel that controls the chloride flux across the neuronal membrane, along with multiple recognition sites for small modulatory molecules such as benzodiazepines, barbiturates, picrotoxin, and certain steroids. When GABA interacts with its receptor, the ion channel is opened, the chloride flux is enhanced, and the cell becomes less responsive to excitatory stimuli. This GABA induced ion current can be regulated by various agents, including agents that interact with the benzodiazepine receptor or recognition site.
Agents that bind or interact with the modulatory sites on the GABAA receptor complex, e.g. the benzodiazepine receptor, and have a positive modulatory effect on the action of GABA, are called benzodiazepine receptor agonists or partial agonists. Agonists generally produce muscle relaxant, hypnotic, sedative, anxiolytic, and/or anticonvulsant effects.
Benzodiazepine receptor ligands with negative modulatory effect on the action of GABA are termed inverse agonists, while benzodiazepine receptor ligands with no intrinsic activity are termed antagonists.
The present invention resides in the surprising discovery that a particular chemical compound, 5-acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime, in addition to being a potent benzodiazepine receptor agonist, is also characterised by a remarkable capability to effectively potentiate responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
SUMMARY OF THE INVENTION
The present invention provides chemical compounds for use as non- sedative anxiolytic drugs, and pharmaceutical compositions comprising these anxiolytic compounds.
Moreover, the present invention provides methods for the screening of chemical compounds for anxiolytic potential. More specifically it is an object of the present invention to provide a convenient tool for the identification of benzodiazepine receptor agonists or partial agonists, capable of exerting a positive modulation of the GABAA receptor complex, which benzodiazepine receptor agonists or partial agonists are devoid the sedative effects often encountered with conventional benzodiazepine receptor agonists.
Accordingly, in its first aspect, the invention relates to the use of a chemical compound for the manufacture of a pharmaceutical composition for the treatment of anxiolytic disorders, which chemical compound potentiate responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
In another aspect the invention provides a method for the screening of chemical compounds for their potential as anxiolytic drugs, which method comprises the steps of determining, in the presence of different concentrations of GABA, the electrophysiological response evoked by GABA on a GABAA receptor; repeating the first step in the presence of a chemical compound to be screened; and selecting the chemical compounds that selectively potentiate responses evoked by GABA at lower concentrations. In a further aspect, the invention provides chemical compounds capable of potentiating responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA, and the use of these compounds as non-sedative anxiolytic drugs. Finally the invention provides pharmaceutical compositions comprising the chemical compound of the invention, and methods of treating anxiolytic disorders, by which methods a mammal, including a human, is treated with a chemical compound of the invention.
DETAILED DISCLOSURE OF THE INVENTION
The Chemical Compounds
The present invention resides in the surprising discovery that a particular chemical compound, 5-acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime, Compound 6 obtained as described in Ex. 1 (hereinafter referred to as Compound 6), in addition to being a potent benzodiazepine receptor agonist, is also characterised by a remarkable capability to potentiate the effect of GABA at low concentrations, while inhibiting the effect of GABA at higher concentrations. I.e. the effect of Compound 6 on a GABA evoked response was dependent on the GABA concentration used.
This effect is considered unexpected because Compound 6, while being a benzodiazepine receptor agonists, in itself has a positive modulatory effect on the action of GABA, and because of this positive modulatory effect would be expected to show an effect that is additive to the effect of GABA or even synergistic, no matter at what GABA concentration.
In particular it was noted that at low GABA concentrations (of from about 0.3 to about 1 μM), Compound 6 show a 4-5 fold potentiation of the GABA response. This potentiation is higher than for the classic benzodiazepines. However, at higher stimulus intensity (at GABA concentrations of more than about 3 μM), Compound 6 show little or no potentiation of GABA responses, and at very high GABA concentrations (of more than about 10 μM), Compound 6 actually blocked GABA responses.
In other words, Compound 6, like the classical benzidoazepines, possesses a potentiating efficacy at lower concentrations of GABA, while this compound, in contrast to the classical benzidoazepines, more quickly looses its potentiating efficacy at slightly higher concentrations of GABA, and ultimately, at high GABA concentrations, becomes an inhibitor of the GABA response. The ability of Compound 6 to act as an efficient potentiator at relative low stimulus intensity while loosing its efficacy at higher stimulus intensity is a very attractive property, and in this way Compound 6 is considered a representative for an entire group of chemical compounds that is particularly useful as non-sedative anxiolytic drugs.
The group of chemical compounds contemplated according to this invention distinguish from the classical anxiolytic compounds and thus represents a novel and distinct group of anxiolytic compounds.
The chemical compounds of the invention is characterised by being a chemical compound that, like the classical benzidoazepines, possesses a potentiating efficacy at lower concentrations of GABA, while, in contrast to the classical benzidoazepines, loosing its potentiating efficacy at higher concentrations of GABA, and ultimately, at very high GABA concentrations, becomes an inhibitor of the GABA response. In a preferred embodiment, the chemical compounds of the invention may be characterised by being any chemical compound, apart from Compound 6, which is capable of potentiating responses evoked by concentrations of GABA of below about 5 μM, preferably below about 3 μM, while inhibiting responses evoked by concentrations of GABA of above about 3 μM, preferably above about 5 μM.
A Method of Screening
Based on this finding, we now provide an efficient tool for use in the screening of new chemical compounds for their potential for use as anxiolytic drugs, in particular for use as non-sedative anxiolytic drugs. According to the method of the invention the electrophysiological response evoked by GABA on a GABAA receptor is determined at different concentrations of GABA and in the presence and absence of the compound to be tested.
After the electrophysiological response has been recorded, the response evoked by GABA alone is compared to that recorded in the presence of the compound to be tested. According to the present invention, compounds capable of increasing the response evoked by GABA at lower concentrations of GABA are selected.
It is at present contemplated that the method shall be carried out at concentrations of GABA in the range of from about 0.001 μM to about 10,000 μM, preferably in the range of from about 0.01 μM to about 1000 μM, more preferred in the range of from about 0.1 μM to about 500 μM, most preferred of from about 0.3 μM to about 300 μM.
Also it is contemplated that chemical compounds that selectively potentiate responses evoked by GABA at concentrations lower than 10 μM, preferably lower than 5 μM, most preferred lower than 3 μM, are selected.
Positive Modulatory Effect
As noted above, benzodiazepine receptor agonists or partial agonists are agents having a positive modulatory effect on the response on GABAA receptors evoked by the action of GABA. A positive modulatory response evoked on GABAA receptors by the action of GABA may be measured using known electrophysiological methods, in particular the well known patch clamp technology.
In a preferred embodiment the response evoked on GABAA receptors by the action of GABA may be measured using whole-cell patch clamp electrophysiological methods.
Cells
The whole-cell patch clamp method may be applied to any cell holding a GABAA receptor.
In a preferred embodiment, the cell applied in this method is a CHO cell, a HEK cell, or a HeLa cell.
GABAA Receptors The GABAA receptors are structurally constituted macromolecular heteropentameric assemblies, containing a combinations of , β, and γ/δ protein subunits. Several subunits of such GABAA receptors (αι-6, βι-3, δ and γι.3) have been characterised using techniques of modern molecular biology. By far the largest population (representing almost half of all GABAA receptors in the brain) is that containing the α1 subunit in combination with the β2 and γ2 subunits.
In a preferred embodiment, a receptor comprising an 1 , an α2, an α3, an o5, or an α6 subunit; a β1 , a β2, or a β3 subunit; and a γ2S or a γ2L subunit, or any combination hereof, is employed. Also αβδ GABAA receptor subunits, or αβε GABAA receptor subunits may be employed.
According to the present invention the use of co-expression of the GABAA receptor subunits α1 , β2 and γ2 is found to be very useful. Also the α1 β2γ2L; the α1 β3γ2L; the α2β2γ2L; the cc2β3γ2L; the α3β2γ2L; the α3β3γ2L; the α5β2γ2L; the oc5β3γ2L; the α6β2γ2L; and the α6β3γ2L GABAA receptors are found to be useful.
In a preferred embodiment, the whole-cell patch clamp method is applied to a CHO cell expressing the oc1 β2γ2 GABAA receptor.
Anxiolytic Drugs
In another aspect the invention relates to the use of the chemical compound of the invention for the manufacture of a pharmaceutical composition for the treatment of anxiolytic disorders.
As defined herein, anxiety disorders include generalised anxiety disorders, panic attacks, phobic disorders, obsessive-compulsive disorders, post-traumatic disorders, and mixed anxiety and depressive disorders.
Pharmaceutical Compositions
In another aspect the invention provides novel pharmaceutical compositions comprising a therapeutically effective amount of the chemical compound of the invention.
While a chemical compound of the invention for use in therapy may be administered in the form of the raw chemical compound, it is preferred to introduce the active ingredient, optionally in the form of a physiologically acceptable salt, in a pharmaceutical composition together with one or more adjuvants, excipients, carriers, buffers, diluents, and/or other customary pharmaceutical auxiliaries.
In a preferred embodiment, the invention provides pharmaceutical compositions comprising the chemical compound of the invention, or a pharmaceutically acceptable salt or derivative thereof, together with one or more pharmaceutically acceptable carriers therefor, and, optionally, other therapeutic and/or prophylactic ingredients, know and used in the art. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not harmful to the recipient thereof. The pharmaceutical composition of the invention may be administered by any convenient route which suite the desired therapy. Preferred routes of administration include oral administration, in particular in tablet, in capsule, in drage, in powder, or in liquid form, and parenteral administration, in particular cutaneous, subcutaneous, intramuscular, or intravenous injection. The pharmaceutical composition may be prepared by the skilled person using standard and conventional techniques appropriate to the desired formulation.
The actual dosage depend on the nature and severity of the disease being treated, and is within the discretion of the physician, and may be varied by titration of the dosage to the particular circumstances of this invention to produce the desired therapeutic effect. However, it is at present contemplated that the pharmaceutical composition of the invention contain, as the active compound, of from about 1 to about 1000 mg of the chemical compound of the invention per individual dose.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention is further illustrated by reference to the accompanying drawing, in which:
Fig. 1 shows responses to 0.3, 3, 30 and 300 μM GABA, recorded from a CHO cell expressing GABAA 1 β2γ2 receptors;
Fig. 2 shows the full dose-response curve for GABA, recorded from a CHO cell expressing GABAA α1 β2γ2 receptors (the EC5o-value was calculated to 18.8 μM and the Hill coefficient to 1.74);
Fig. 3 shows responses to 6 μM GABA, 1 μM GABA, and 10 μM GABA, recorded in the absence and the presence of 10 μM of a test compound (Compound 6 of Ex. 1), recorded from a CHO cell expressing GABAA α1 β2γ2 receptors;
Fig. 4 shows the effect of 10 μM of a test compound (Compound 6 of Ex. 1 ) as a function of the GABA concentration, recorded from a CHO cell expressing GABAA α1 β2γ2 receptors; Fig. 5 shows the effect of 1 μM of a test compound (Compound 6 of Ex. 1 ) as a function of the GABA concentration, and compared to 1 μM of a classical benzodiazepine agonist (Diazepam™), recorded from a CHO cell expressing GABAA α1 β2γ2 receptors; and Fig. 6 shows the simultaneous effect of 10 μM of a test compound (Compound 6 of Ex. 1 ) and 3 μM of a classical benzodiazepine agonist (Diazepam™), compared the effect of 10 μM of the test compound alone, recorded from a CHO cell expressing GABAA α1 β2γ2 receptors.
EXAMPLES
The invention is further illustrated with reference to the following examples which are not intended to be in any way limiting to the scope of the invention as claimed.
Example 1
Preparatory Example
5-Acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime (Compound 6)
4-Fluoro-3-nitroacetophenone (Compound 1): Concentrated sulphuric acid (200 ml) is cooled to 5°C. 4-Fluoroacetophenone (20 ml, 0.16 mol) is added keeping the temperature below 10°C. The mixture is cooled to 0-5°C and potassium nitrate (25 g, 0.25 mol) is added portionwise over 2 hours keeping the temperature within this range. Following the addition the mixture is stirred in the cold for additionally 2 hours. The mixture is poured on ice (600 g) and the crude product is filtered off. Column- chromatographic purification on silica gel using a mixture of ethyl acetate and petroleum ether (1 :9) as the eluent affords pure Compound 1 (18.2 g, 60%). Mp 48- 49°C.
3-(3-Pyridyl)aniline (Compound 2): A mixture of diethyl 3-pyridylborane (16.3 g, 0.11 mol), 3-bromoaniline (12.2 ml, 0.11 mol), potassium carbonate (45.8 g, 0.33 mol) and tetrakis(triphenylphosphine)palladium(0) (80 mg) in a mixture of water (40 ml) and dimethoxyethane (80 ml) is heated to 80°C under a stream of nitrogen over night. After cooling the mixture is diluted with water and ethyl acetate and filtered through a fluted filter paper. The layers are separated. The aqueous layer is extracted once with ethyl acetate. The combined organic phases are dried over sodium sulphate and concentrated under reduced pressure. The residue is dissolved in ethanol. Water is added and the mixture is evaporated to dryness. This residue crystallises upon trituration with ice-cold water. The crystals are collected, dried and washed with petroleum ether to afford pure Compound 2 (16.3g, 87%). Mp 75-76°C. 4-Acetyl-2-nitro-N-(3-(3-pyridyl)phenyl)aniline (Compound 3): A mixture of Compound 1 (5 g, 27.3 mmol) and Compound 2 (4.62 g, 27.2 mmol) in dry N-methyl- 2-pyrrolidone (10 ml) is stirred at 40-50°C over night. The resulting solid reaction mixture is suspended in ice-water and rendered alkaline by addition of aqueous sodium carbonate (1 M). The product is filtered off, washed with water and dried to yield 7.68 g (Compound 3) (85%). Mp 112-113°C.
5-Acetyl-2-(3-(3φyridyl)phenylamino)aniline (Compound 4): Compound 3 (2 g, 6 mmol) is suspended in a mixture of ethanol (50 ml) and dichloromethane (10 ml) and is hydrogenated at ambient pressure using palladium (5% on activated carbon) as the catalyst. Filtration of the resulting solution through celite followed by evaporation of solvent leaves an oil. Trituration with a mixture of diethyl ether and petroleum ether (1 :1) affords 1.46 g pure Compound 4 (80%). Mp 175-176°C.
5-Acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole (Compound 5): A solution of Compound 4 (7 g, 23.1 mmol) in formic acid (20 ml) is stirred overnight at ambient temperature. The mixture is poured into water (750 ml) and rendered basic with concentrated aqueous ammonia. The precipitate is filtered off, washed with water and dried. The crude product is dissolved in refluxing ethanol. Water is added until onset of precipitation. The mixture is allowed to cool. The product is filtered off and dried to yield 4.3 (60%). Mp. 200 - 202°C. 5-Acetyl-1-(3-(3-pyridyl)phenyl)benzimidazole O-ethyl oxime (Compound 6):
Compound 5 (5 g, 15.97 mmol) is suspended in absolute ethanol (50 ml) and heated to 70°C. O-Ethyl hydroxylamine hydrochloride (2.4 g, 24.61 mmol) is added and the mixture is heated to reflux for 1.5 hours. After cooling the solvent is removed by evaporation, and the residue is stirred with aqueous sodium hydroxide (50 ml, 1 M). The crude product is filtered off. Column-chromatographic workup on silica gel using a mixture of ethyl acetate and ethanol (9:1 ) as the eluent affords pure Compound 6. Yield: 3.7g (65%). Mp 105-106°C.
Example 2 Preparation of CHO Cells expressing GABAA Receptors
Cloning: The cloning of hGABAA subunits was carried out as follows. Human brain hippocampus poly A+ mRNA (Clontech) was reverse transcribed with oligo dT primer and MMLV reverse transcriptase (Pharmacia). 100 ng first strand cDNA was subjected to PCR using Expand HF polymerase (Boehringer Mannheim) and specific primer sets spanning the full length cDNA sequence. The primer sets were:
1. hGABAAαι : (s) 5OCTTAGCTGCTCCAGCCCGCGATGAGGA3'; (as) 5TTCCCAGTGCAGAGGAACTGAACAACAGAATG3';
2. hGABAAβ2: (s) 5ΑAAACTAAAGGGATGTGGAGAGTGCGGAAAAG3'; (as) 5TCCAGTGGGAGGCCATGTTTTAGTTCACATAA3';
3. hGABAAγ2: (s) 5OTGCAACCAAGAGGCAAGAGGCGAGAG3'; and (as) 5'CCATATCAGTAAAACCCATACCTCCTCACAGG3'. PCR conditions were 1) 94 °C 60 seconds; 2) 15 times (94 °C for 60 seconds, 55°C for 60 seconds, 72°C for 120 seconds); 3) 20 times (94 °C for 60 seconds, 55°C for 60 seconds, 72°C for 180 seconds); 4) 72°C for 10 minutes. The amplified products were polished with pfu polymerase (Stratagene), purified on Quiaquick column (Qiagen) and subcloned into pCRScript (Stratagene). Maxiprep DNA (Qiagen) was prepared from positive clones and two to three individual clones of each subunits was sequenced bidirectionally on an ABI 377 sequencer using standard chain termination dyes (Perkin Elmer).
Subcloninq: The α subunit was subcloned into pUbi2z (a custom designed mammalian expression vector using a human ubiquitin promoter to drive the inserted cDNA and the cDNA for enhanced green fluorescence protein (EGFP) fused to the selection marker conferring Zeocin resistance), the β2-subunit was subcloned into pNS1 n (a custom designed mammalian expression vector using a CMV promoter to drive the inserted cDNA and kanamycin/neomycin selection marker), and the γ2s- subunit was subcloned into pZeoSV (Invitrogen). A single plasmid containing expression cassettes for both the β2 subunit and the γ2s-subunit was constructed as follows. The entire expression cassette for the γ2s-subunit in pZeoSV (SV40 promoter, cDNA, SV40-polyA) was excised as a BamHI fragment and subcloned into the Bglll site at position 1 in pNS1 n/hGABAAβ2- A clones in which the driving promoters SV40 and CMV were pointing in opposite directions was selected.
Transfection: For transient transfection experiments 1 μg of pUbi2z/αι was mixed with 1 μg of pNS1 n/β2γ2S and transfected into Chinese hamster ovary cells (CHO, ATCC) using lipofectamine (Gibco) according to manufactures instructions. 24 hours after transfection, cells were plated into 35 mm Petri dishes containing 3.5 mm glass coverslips. Cells were kept at 37°C, in 5% C02/95% air and used for electrophysiological studies 2-50 hours after plating.
Example 3
Patch Clamp Experiments
Electrophysioloqy: All experiments were performed in voltage clamp using conventional whole cell patch clamp methods. The following salt solutions were used: Extracellular solution (mM): NaCI (140), KCI (4), CaCI2 (2), MgS04 (4),
HEPES (10, pH =7.4);
Intracellular solution (mM): CsCI (120), CsF (20), MgS04 (4), EGTA (10), ATP (4), HEPES (10, pH = 7.2).
Electronics, programs and data acquisition: The amplifier used was an EPC-9 (HEKA-electronics, Lambrect, Germany) run by a Macintosh 7600/120 computer via an ITC-16 interface. Experimental conditions were set with the Pulse- software accompanying the amplifier. Data were low pass filtered and sampled directly to hard-disk at a rate of 3 times the cut-off frequency.
Pipettes and electrodes: Pipettes were pulled from borosilicate glass (Modulohm, Copenhagen, Denmark) using a horizontal electrode puller (Zeitz- Instrumente, Augsburg, Germany). The pipette resistance was 1.6 - 2.6 mW in the salt solutions used in these experiments. The pipette electrode was a chloridised silver wire, and the reference was a silverchloride pellet electrode (In Vivo Metric, Healdsburg, USA) fixed to the experimental chamber. The electrodes were zeroed with the open pipette in the bath just prior to sealing.
Experimental procedure: The patch clamp experiments were performed at room temperature (20 - 22 °C).
Coverslips were transferred to a 15 ml experimental chamber mounted on the stage of an inverted microscope (IMT-2, Olympus) supplied with Nomarski optics and a mercury lamp (Olympus). Cells were continuously superfused with extracellular saline at a rate of 2,5 ml/min. Cells chosen for experiments were spherical and emitted bright green fluorescence when exposed to UV-light from the mercury lamp. After giga-seal formation (1 -5 GW, success-rate = 90 %) the whole cell configuration was attained by suction.
The cells were held at a holding potential of -60 mV and at the start of each experiment the current was continuously measured for 20 sec to ensure a stable baseline. GABA-containing solutions were delivered to the chamber through a custom- made gravity-driven flowpipe, the tip of which was placed approximately 50 μm from the cell. Application was triggered by compressing the tubing connected to the flow pipe with a valve controlled by the Pulse-software. In general, GABA was applied for 4 sec. every 30 sec. The sampling interval during application was 1 msec. Having obtained responses of a stable amplitude, the extracellular saline as well as the agonist containing solution were switched to solutions containing the compound (i.e. Compound 6 or Diazepam) to be tested. The compound was present until responses of a stable amplitude were achieved.
Currents were measured at the peak of the responses. Effect of compounds was calculated as the peak current at compound equilibrium divided by the current evoked by the pulse immediately before the compound was included. The results of these experiments are presented in Figs. 1-6. In Fig. 1 the cell was voltage clamped at -60 mV and exposed to GABA at varying concentrations ejected from the flowpipe. The time between pulses was 30 sec.
In Fig. 2 the magnitude of the peak currents were expressed as fractions of the peak current obtained with a maximal concentration of GABA. Symbols represent mean ±SEM, n=3-10. The curve is a fit to the Michaelis-Menten equation and the parameters EC50 and n were calculated from the equation. In Fig. 3 the cell was voltage clamped at -60 mV and exposed to 4-sec pulses of 6 μM GABA every 20 sec. After assessment of the effect of Compound 6 (indicated by the horizontal bar, top panel), the GABA concentration was changed to 1 μM, and the effect of Compound 6 was evaluated (middle panel). Finally, the effect of Compound 6 on responses evoked by 10 μM GABA was evaluated (bottom) (please note the different scaling).
In Fig. 4 cells were voltage clamped at -60 mV and exposed to 4-sec pulses of GABA at 0.3-100 μM. After obtaining responses of a stable amplitude, Compound 6 (10 μM) was included in the bath-Ringer and in the GABA solutions. When responses again were stable, solutions were switched back to control. Results are expressed as % of control, referring to the response obtained just before Compound 6 was included (100 % = no effect on peak amplitude). Data points are mean ±SEM (n=3-10).
In Fig. 5 the cells were voltage clamped at -60 mV and exposed to 4-sec pulses of GABA at 0.3-100 μM. After obtaining responses of a stable amplitude, Compound 6 (1 μM, closed symbols) or Diazepam (1 μM, open symbols) was included in the bath-Ringer and in the GABA solutions. When responses again were stable, solutions were switched back to control. Results are expressed as % of control, referring to the response obtained just before the drug in question was included (100 % = no effect on peak amplitude). Data points are mean ±SEM (n=3-5).
In Fig. 6 the cells were voltage clamped at -60 mV and exposed to 4-sec pulses of 0.3 μM GABA every 20 sec. After two control responses of the same size, Compound 6 (10 μM) and Diazepam (3 μM) were included in the solutions. After one minute (3 responses under the hatched bar), Diazepam was omitted from the solutions. After one minute (3 responses under the open bar only) solutions were changed back to control conditions.

Claims

1. Use of a chemical compound for the manufacture of a pharmaceutical composition for the treatment of anxiolytic disorders, which chemical compound potentiate responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
2. The use according to claim 1 , in which the chemical compound potentiate responses evoked by concentrations of GABA of below 3 ╬╝M, while inhibiting responses evoked by concentrations of GABA of more above 3 ╬╝M.
3. The use according to claim 1 , in which the chemical compound potentiate responses evoked by concentration of GABA in the range of from about 0.001 to about 10 ╬╝M, more preferred in the range of from about 0.1 to about 5 ╬╝M, most preferred in the range of from about 0.3 to about 3 ╬╝M.
4. The use according to claim 1 , in which the chemical compound inhibit responses evoked by concentrations of GABA of more than about 50 ╬╝M, more preferred of more than about 10 ╬╝M, yet more preferred of more than about 5 ╬╝M, most preferred of more than about 3 ╬╝M.
5. A method for the screening of chemical compounds for their potential as anxiolytic drugs, which method comprises the steps of
(i) determining, in the presence of different concentrations of GABA, the electrophysiological response evoked by GABA on a GABAA receptor;
(ii) repeating step (i) in the presence of a chemical compound to be screened; and (iii) selecting the chemical compounds that selectively potentiate responses evoked by GABA at lower concentrations.
6. The method according to claim 5, wherein, in steps (i) and (ii), the concentration of GABA range of from about 0.001 ╬╝M to about 10,000 ╬╝M, preferably of from about 0.01 ╬╝M to about 1000 ╬╝M, more preferred of from about 0.1 ╬╝M to about 500 ╬╝M, most preferred of from about 0.3 ╬╝M to about 300 ╬╝M.
7. The method according to claim 5, wherein in step (iii) chemical compounds that 5 selectively potentiate responses evoked by GABA at concentrations lower than 5 ╬╝M, preferably lower than 3 ╬╝M, are selected.
8. The method according to any of claims 5-7, wherein the determination of the electrophysiological response in step (i) and (ii) is performed by voltage clamp
10 using whole cell patch clamp methods.
9. The method according to claim 8, wherein the GABAA receptor is located on a CHO cell, a HEK cell, or a HeLa cell.
15 10. The method according to claim 9, wherein the cell is expressing the ╬▒1 ╬▓2╬│2; the ╬▒1 ╬▓2╬│2L; the ╬▒1 ╬▓3╬│2L; the ╬▒2╬▓2╬│2L; the oc2╬▓3╬│2L; the oc3╬▓2╬│2L; the ╬▒3╬▓3╬│2L; the ╬▒5╬▓2╬│2L; the ╬▒5╬▓3╬│2L; the ╬▒6╬▓2╬│2L; or the ╬▒6╬▓3╬│2L GABAA receptor.
11. The method according to claim 9, wherein cell is a CHO cell expressing the 20 1 ╬▓2╬│2 GABAA receptor.
12. A chemical compound having anxiolytic activity identified by the method according to any of claims 5-11.
25 13. A chemical compound capable of potentiating responses evoked by lower concentrations of GABA, while inhibiting responses evoked by higher concentrations of GABA.
14. The chemical compound of claim 13, which is capable of potentiating responses 30 evoked by concentrations of GABA of below 3 ╬╝M, while inhibiting responses evoked by concentrations of GABA of more above 3 ╬╝M.
15. The chemical compound of either of claims 13-14 for use as a non-sedative anxiolytic drug.
16. A pharmaceutical composition comprising the chemical compound of any of claims 12-15.
17. A method of treating an anxiolytic disorder, by which method a mammal, including a human, is treated with a chemical compound according to any of claims 12-15.
PCT/DK1998/000489 1997-12-09 1998-11-13 Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds WO1999029319A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU12244/99A AU1224499A (en) 1997-12-09 1998-11-13 Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DK1433/97 1997-12-09
DK143397 1997-12-09

Publications (2)

Publication Number Publication Date
WO1999029319A2 true WO1999029319A2 (en) 1999-06-17
WO1999029319A3 WO1999029319A3 (en) 1999-07-22

Family

ID=8104787

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK1998/000489 WO1999029319A2 (en) 1997-12-09 1998-11-13 Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds

Country Status (2)

Country Link
AU (1) AU1224499A (en)
WO (1) WO1999029319A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002000623A2 (en) * 2000-06-26 2002-01-03 Neurogen Corporation Aryl fused substituted 4-oxy-pyridines
WO2002012442A2 (en) * 2000-08-07 2002-02-14 Neurogen Corporation Heterocyclic compounds as ligands of the gabaa receptor
WO2002098907A3 (en) * 2001-06-06 2003-04-24 Neurosearch As Cation conducting gabaa receptors and their use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5130430A (en) * 1990-10-31 1992-07-14 Neurogen Corporation 2-substituted imidazoquinoxaline diones, a new class of gaba brain receptor ligands
WO1996033191A1 (en) * 1995-04-21 1996-10-24 Neurosearch A/S Benzimidazole compounds and their use as modulators of the gabaa receptor complex
WO1998017651A1 (en) * 1996-10-21 1998-04-30 Neurosearch A/S 1-phenyl-benzimidazole compounds and their use as baga-a receptor modulators
WO1998034923A1 (en) * 1997-02-07 1998-08-13 Merck Sharp & Dohme Limited Phenylbenzimidazole derivatives as ligands for gaba receptors
EP0889044A1 (en) * 1997-06-30 1999-01-07 Ortho-McNeil Pharmaceutical, Inc. 5-(heteroaryl)alkyl)-3-oxo-pyrido(1,2-a)benzimidazole-4-carboxamide (PBI) derivatives useful in treating central nervous system disorders

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5130430A (en) * 1990-10-31 1992-07-14 Neurogen Corporation 2-substituted imidazoquinoxaline diones, a new class of gaba brain receptor ligands
WO1996033191A1 (en) * 1995-04-21 1996-10-24 Neurosearch A/S Benzimidazole compounds and their use as modulators of the gabaa receptor complex
WO1998017651A1 (en) * 1996-10-21 1998-04-30 Neurosearch A/S 1-phenyl-benzimidazole compounds and their use as baga-a receptor modulators
WO1998034923A1 (en) * 1997-02-07 1998-08-13 Merck Sharp & Dohme Limited Phenylbenzimidazole derivatives as ligands for gaba receptors
EP0889044A1 (en) * 1997-06-30 1999-01-07 Ortho-McNeil Pharmaceutical, Inc. 5-(heteroaryl)alkyl)-3-oxo-pyrido(1,2-a)benzimidazole-4-carboxamide (PBI) derivatives useful in treating central nervous system disorders

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MARYANOFF B E ET AL: "POTENTIAL ANIOLYTIC AGENTS. 2. IMPROVEMENT OF ORAL EFFICACY FOR THEPYRIDO 1,2-ABENZIMIDAZOLE (PBI) CLASS OF GABA-A RECEPTOR MODULATORS" BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 6, no. 3, 1996, pages 333-338, XP002054104 *
MARYANOFF B E ET AL: "POTENTIAL ANXIOLYTIC AGENTS. PYRIDOL 1,2-ABENZIMIDAZOLES: A NEW STRUCTURAL CLASS OF LIGANDS FOR THE BENZODIAZEPINE BINDING SITE ON GABA-A RECEPTORS" JOURNAL OF MEDICINAL CHEMISTRY, vol. 38, 1995, pages 16-20, XP002053732 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002000623A2 (en) * 2000-06-26 2002-01-03 Neurogen Corporation Aryl fused substituted 4-oxy-pyridines
WO2002000623A3 (en) * 2000-06-26 2002-07-18 Neurogen Corp Aryl fused substituted 4-oxy-pyridines
US6828329B2 (en) 2000-06-26 2004-12-07 Neurogen Corporation Aryl fused substituted 4-oxy-pyridines
WO2002012442A2 (en) * 2000-08-07 2002-02-14 Neurogen Corporation Heterocyclic compounds as ligands of the gabaa receptor
WO2002012442A3 (en) * 2000-08-07 2003-05-01 Neurogen Corp Heterocyclic compounds as ligands of the gabaa receptor
US6653471B2 (en) 2000-08-07 2003-11-25 Neurogen Corporation Heterocyclic compounds as ligands of the GABAA receptor
US6949562B2 (en) 2000-08-07 2005-09-27 Neurogen Corporation Heterocyclic compounds as ligands of the GABAA receptor
WO2002098907A3 (en) * 2001-06-06 2003-04-24 Neurosearch As Cation conducting gabaa receptors and their use
US7282344B2 (en) 2001-06-06 2007-10-16 Neurosearch A/S Cation conducting GABAA receptors and their use

Also Published As

Publication number Publication date
AU1224499A (en) 1999-06-28
WO1999029319A3 (en) 1999-07-22

Similar Documents

Publication Publication Date Title
Pearson et al. Characterization of Ca2+ channel currents in cultured rat cerebellar granule neurones.
EP3074377B1 (en) Substituted benzamides and methods of use thereof
Yamakage et al. Calcium channels—basic aspects of their structure, function and gene encoding; anesthetic action on the channels—a review
Ludwig et al. Functional expression of a rat homologue of the voltage gated either á go‐go potassium channel reveals differences in selectivity and activation kinetics between the Drosophila channel and its mammalian counterpart.
Britton et al. Corticotropin releasing factor and amphetamine exaggerate partial agonist properties of benzodiazepine antagonist Ro 15-1788 in the conflict test
CN106008504A (en) Pyrrolopyrazine-spirocyclic piperidine amides as modulators of ion channels
CN103443105A (en) Chroman-spirocyclic piperidine amides as modulators of ion channels
EP2841435B1 (en) Pyrrolopyridinone derivatives as ttx-s blockers
EP0603312A1 (en) Novel gaba a? receptor with steroid binding sites
KR101449348B1 (en) Piperidinyl pyrimidine amides as kv7 potassium channel openers
KR19990008068A (en) Mixture of 5-HT Absorption Inhibitor and Selective 5-HT1A Antagonist
JP2001521505A (en) Pharmacological substances
JP2004513125A (en) N-type calcium channel antagonists for the treatment of pain
US5856124A (en) DNA encoding high-affinity melatonin receptors
Borsini Balance between cortical 5-HT1A and 5-HT2 receptor function: hypothesis for a faster antidepressant action
WO1999029319A2 (en) Chemical compounds for use as anxiolytic agents and a method for the identification of anxiolytic compounds
Okada et al. Effects of non-toxic and toxic concentrations of phenytoin on monoamines levels in rat brain
JP2010517926A (en) 4-Phenyl-3- (2-propylsulfonylamino) tetrahydrofuran derivatives useful for the treatment of schizophrenia that enhance glutamate receptors
DE69920681T2 (en) HUMAN K + ION CHANNEL AND THERAPEUTIC USES THEREOF
US5137910A (en) Compositions containing indole-2-carboxylate compounds for treatment of CNS disorders
Lacey et al. Actions of phencyclidine on rat locus coeruleus neurones in vitro
Busch et al. Blockade of human IsK channels expressed in Xenopus oocytes by the novel class III antiarrhythmic NE-10064
DE60132571T2 (en) DNA coding for the human VANILLOID RECEPTOR VR3
US5229413A (en) Compositions containing indole-2-carboxylate compounds for treatment of CNS disorders
TW201105324A (en) 2-(1,2-benzisoxazol-3-yl)benzylamine derivatives

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase in:

Ref country code: KR

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: CA