WO1996029316A1 - NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS - Google Patents

NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS Download PDF

Info

Publication number
WO1996029316A1
WO1996029316A1 PCT/SE1996/000344 SE9600344W WO9629316A1 WO 1996029316 A1 WO1996029316 A1 WO 1996029316A1 SE 9600344 W SE9600344 W SE 9600344W WO 9629316 A1 WO9629316 A1 WO 9629316A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
dibenzo
methyl
alkyl
Prior art date
Application number
PCT/SE1996/000344
Other languages
French (fr)
Inventor
Håkan WIKSTRÖM
Peter De Boer
Yi Liao
Original Assignee
Wikstroem Haakan
Peter De Boer
Yi Liao
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 Wikstroem Haakan, Peter De Boer, Yi Liao filed Critical Wikstroem Haakan
Priority to AU51305/96A priority Critical patent/AU5130596A/en
Publication of WO1996029316A1 publication Critical patent/WO1996029316A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D281/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D281/02Seven-membered rings
    • C07D281/04Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D281/08Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D281/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • C07D281/14[b, e]-condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/38[b, e]- or [b, f]-condensed with six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D267/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D267/02Seven-membered rings
    • C07D267/08Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D267/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D267/16Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with two six-membered rings
    • C07D267/18[b, e]-condensed

Definitions

  • the present invention is directed toward new sulfone ester analogues of iso-Clozapine [RN 1977-08-81 and their pharmaceutically acceptable salts, to processes for preparing such compounds, pharmaceutical preparations of such compounds and the use of such compounds in manufacture of a pharmaceutical preparation.
  • Pharmaceutical preparations of these compounds are useful for central nervous system disorders, such as for a therapeutic effect on one or several of the following receptor systems: dopamine (DA; D1 - D4), 5-HT2A, 5-HT2C, ⁇ 1 and muscarinic (M1 -M4) in mammals.
  • Clozapine [RN 54241-01 -9] is an efficient anti-psychotic agent 5 and binds with high affinity to DA D1 and D4 receptors and with moderate affinity to DA D2 and D3 receptors. 6 In addition, Clozapine binds with high affinity to ⁇ 1 receptors, muscarinic 1-M4 receptors and 5-HT2A and 5-HT2C receptors. 6 Clozapine has a low propensity to induce extrapyramidal side-effects in humans and catalepsy in rats. In addition, Clozapine does not increase the prolactine levels, something which the classical neuroleptics (e. g. haloperidol and chlorpromazine) potently do.
  • the classical neuroleptics e. g. haloperidol and chlorpromazine
  • Clozapine is hampered by rare incidences of a serious side-effect (blood toxicity, aranulocytosis) .
  • B Expensive, continuous blood monitoring makes the use of Clozapine tedious and expensive and the use of Clozapine is therefore limited to psychotic patients not responding to the first and second choice of drug therapy. 7
  • Clozapine analogues have been evaluated but, to date, no marketed compound with a Clozapine-like structure and Clozapine-like pharmacological and clinical profile, i. e. an analogue that is also free from side-effects, has been found. 7
  • Loxapine references 1 2, 13, 14, 15, 16, 17, 18, 19, 20, 21
  • This invention is related to novel iso-Clozapine, Loxapine and Clothiapine analogues of Formula 1 :
  • R* is H, (C*-C 8 ) alkyl or haloalkyl (e. g. CF 3 (triflates) and CF 2 CF 3 ) or hydroxyalkyl, alkenyl, alkynyl. cyclopropylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyi;
  • R 2 is H, (C* -C 8 ) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (C r C 8 ) haloalkyl (e. g. CH 2 CF 3 CH 2 CH 2 CF 3 , CH 2 CH 2 F,
  • alkyl-2-imidazolidinone (e. g. 1 -(CH 2 CH 2 -2-imidazolidinone) and 1 -(CH 2 CH 2 CH 2 -2- imidazolidinonel);
  • X is NH, NR 0, S, SO, S0 2 .
  • the compounds of this invention possess affinity to one or several receptor systems, e. g. DA (D1 -D4), ol , muscarinic (M1 -M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7).
  • the central nervous system disorders to be treated with the compounds of the present invention include psychoses/schizophrenia, autism, Tourette's syndrome, restless legs, Huntington's chorea, motion sickness, nausea, vomiting and severe anxiety.
  • the invention is directed to compounds of Formula 1 wherein R. is haloalkyl R 2 is H or CH 3 and X is NH, 0 or S.
  • R. is haloalkyl
  • R 2 is H or CH 3 and X is NH, 0 or S.
  • R - is CF 3
  • R 2 is H or CH 3 and X is NH, 0 or S.
  • R * is CF 3
  • R 2 is CH 3 and X is NH, O or S.
  • the most preferred compound is the compound of Formula I wherein R * is CF 3 , R 2 is CH 3 and X is NH.
  • An object of the invention is to provide compounds for therapeutic use, especially compounds having a therapeutic activity in the central nervous system. Another object is to provide compounds having an effect on receptors (DA (D1 -D4), ⁇ 1 , muscarinic (M1 - M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7)) in mammals including man.
  • DA D1 -D4
  • M1 - M4 muscarinic
  • 5-HT 5-HT2A, 5-HT2C and 5-HT7
  • alkyl refers to an aliphatic hydrocarbon radical and - 4 - includes branched or unbranched forms such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neo-pentyl, n-hexyl, isohex ⁇ l, n-heptyl, isoheptyl, and n-octyl.
  • Alkenyl refers to a radical of an aliphatic unsaturated hydrocarbon having a double bond and includes both branched and unbranched forms such as ethenyl, 1 -methyl- 1-ethenyl, 1- propenyl, 2-propenyl, 1 -butenyl, 2-butenyl, 3-butenyl, 2-methyl-1-butenyl, l-pentenyl, 2- pentenyl, 3-pentenyl, 4-pentenyl, 1 -methyl-4-pentenyl, 3-methyl-1-pentenyl, 3-methyl-2- pentenyl, 1 -hexenyl, 2-hexenyl, 3-hexenyl , 4-hexenyl, 1 -methyl-4-hexenyl, 3-methyl-1 - hexenyl, 3-methyl-2-hexenyl, 1 -heptenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 1 -methyl-4
  • C ⁇ cloalkyl refers to a radical of a saturated cyclic hydrocarbon such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohex ⁇ l, cycloheptvl, or cyclooctyl.
  • Hydrogen means fluorine, chlorine, bromine or iodine, preferably fluorine.
  • compounds of this invention may contain chiral centers (e. g. R* and/or R 2 ).
  • the scope of this invention includes all enantiomeric or diastereomeric forms of Formula 1 compounds, either in pure form or as mixtures of enantiomers or diastereomers.
  • the therapeutic properties of the compounds may to a greater or lesser degree depend on the stereochemistry of a particular compound. Pure enantiomers, as well as enantiomeric or diastereomeric mixtures, are within the scope of the invention.
  • Both organic and inorganic acids can be employed to form non-toxic pharmaceutically acceptable acid addition salts of the compounds of this invention.
  • Illustrative acids are sulfuric, nitric, phosphoric, hydrochloric, citric, acetic, lactic, tartaric, pamoic, methanesulfonic, ethanedisulfonic, sulfamic, succinic, cyclohexylsulfamic, fumaric, maieic and benzoic acid.
  • These salts are readily prepared by methods known in the art.
  • the compounds of this invention may be obtained by one of the following methods described below, as outlined in the appropriate charts.
  • the compounds of the present invention will normally be administered orally, rectally or by injection, in the form of pharmaceutical preparations comprising the active ingredient either as a free base or as a pharmaceutically acceptable non-toxic, acid addition salt, such as the hydrochloride, lactate, acetate, methanesulfonate, sulfamate salt, in association with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable non-toxic, acid addition salt such as the hydrochloride, lactate, acetate, methanesulfonate, sulfamate salt
  • the suitable daily doses of the compounds of the invention are from about 0.1 mg to 2000 mg for oral application, preferably 0.5-500 mg, and 0.05 mg to about 100 mg for parenteral application, preferably 0.05-50 mg daily doses.
  • the daily dose will preferably be administrated in individual dosages 1 -4 times daily and the dosage amounts are based on an individual having a weight of 70 kg. w
  • R « is H, (C*-C 8 ) alkyl or haloalkyl (e. g. CF 3 (triflates) and CF 2 CF 3 ) or hydroxyalkyl, alkenyl, alkynyl, cyclopropylalkyl, aryl, ar ⁇ lalk ⁇ l, heteroaryl, heteroarylalkyl;
  • R 2 is H, (C 1 -C 8 ) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (C r C 8 ) haloalkyl (e. g. CH 2 CF 3 CH 2 CH 2 CF 3 , CH 2 CH 2 F, is CH 2 CH 2 CH 2 F), hydroxyalkyl, hydroxyalkyloxyalkyl (e. g. or 1-
  • alkyl-2-imidazolidinone (e. g. 1 -(CH 2 CH 2 -2-imidazolidinone) and 1 -(CH 2 CH 2 CH 2 -2- imidazolidinone));
  • X is NH, NR., 0, S, SO, S0 2 , display binding affinity at the receptor systems: DA (D1-D4), ⁇ 1 , muscarinic (M1 -M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7) in mammals including man.
  • DA D1-D4
  • M1 -M4 muscarinic
  • 5-HT 5-HT2A, 5-HT2C and 5-HT7
  • These compounds are particularly effective atypical anti- 0 psychotic (anti-schizophrenic) agents.
  • Other uses for these compounds include autism, Tourette's syndrome, restless legs, Huntington's chorea, motion sickness, nausea, vomiting and severe anxiety.
  • the compounds of this invention also have high oral potency and a long duration of 5 action.
  • the utility of the compounds of this invention to treat central nervous system disorders can be shown in behavioral and biochemical activity in rats.
  • biochemical effects of the compounds of this invention can be monitored with the microdial ⁇ sis technique in freely moving animals (see under Experimental Section below).
  • the propensity of the compounds of this invention to induce catalepsy was measured in rats, who were placed temporarily on a vertical grid (see under Experimental Section below).
  • Suitable starting materials either the phenols or the OMe-analooues. which can be dealkylated to the corresponding phenols with e. g. AICI 3 in ethylmercaptane at room temperature, for this invention are known:
  • 2-Trifluoromethanesulfon ⁇ loxy-5,10-dihydro-11 -oxo-dibenzo[b,f][1 ,4]-oxazepine 2- Hydroxy-5,10-dihydro-1 1 -oxo-dibenzo[b,e][1 ,4]oxazepine (1 .0 g) in 20 mL of dichloromethane and 5 mL of triethylamine was cooled to -78 °C, treated with triflic anhydride (1.54 mL, 7 mmol). The mixture was stirred for 3 h to allow the temperature back to room temperature, quenched with 3 mL of 50% methanol in water and concentrated.
  • 2-Trif luoromethanesulf onyloxy- 1 1 -(4-methhyl ⁇ iperazino)-5H-dibenzo-[b ,f ] [ 1 ,4]thiazepine 2-Trifluoromethanesulfonyloxy-5, 10-dihydro-1 1 -oxo-dibenzo-[b,e][1 ,4)thiazepine (80 mg), POCI 3 (1 mL), toluene (1 mL) and N,N-dimethyl-aniline (0.1 mL) were combined and heated to reflux for 3 h.
  • N-methyl-piperazine derivatives e. g. compounds 12, 15-20
  • an alk ⁇ lation with the appropriate alk ⁇ lating agent (e. g. 1 -(2- chloroeth ⁇ l)-2-imidazoiidinone and 1 -(3-chloroprophyl)-2-imidazolidinone, respectively), refluxing over night in methyl isobutyl ketone (MIBK) in the presence of dry, ground K 2 C0 3 (s) and a catalytic amount of Kl (s).
  • MIBK methyl isobutyl ketone
  • Microdialysis experiments Adult male Wistar rats (Harlan Centraal Proefdier Bedrijf, The Netherlands) were used. Until implantation of the dialysis probe, the rats were housed in groups of 5-10 animals in plastic cages. The rats were kept in a room maintained under constant temperature (21 °C) and humidity (40%) on a 12:12 dark:light cycle (07:00 on and 19:00 off) with food and water available ad libitum. The rats weighed between 250- 350 g at the time of the microdialysis probe implantation. Animal procedures were conducted in accordance with guidelines published in the NIH Guide for Care and Use of Laboratory Animals and all protocols were approved by an Institutional Animal Care and Use Committee.
  • microdialysis probes used in the present investigation were of a vertical, concentric design: A piece of fused silica capillary (Polymicro Technologies, Phoenix, AZ, USA) with I.D. 50 ⁇ m and O.D. 1 15 ⁇ m was threaded through a piece of PE-20 tubing (Clay Adams, Parsippan ⁇ , NJ, USA) 0.5 cm from the end. A dialysis membrane (8 mm) (Hospal, Meyzieu, France) with an I.D. of 250 ⁇ m and an O.D.
  • the animals were anesthetized with chloral hydrate (400 mg/kg) and a microdialysis probe was implanted into the CPu at the following coordinates: (flat skull) AP 1.0 mm, ML + 2.5 mm relative to bregma, and -6.0 mm below the dura and/or nucleus accumbens at the following coordinates: (flat skull) A. P. 2.5 mm, ML -1.3 relative to bregma, and -7.3 mm below the dura.
  • the probes were lowered using an electrode carrier (Model 1760, David Kopf, Tujunga, CA, USA) at a speed of approximately 400 ⁇ m/mi ⁇ and secured to the skull with fast-curing dental cement on three skull screws.
  • aCSF cerebrospinal fluid
  • NaCI 147, KCI 2.5, CaCI 2 1 .3, and MgCI 0.9, pH 7.4 was perfused through the probe with a syringe pump (Harvard Apparatus inc, South Natick, MA, USA).
  • the outlet of the probes were connected to the valve of an HPLC apparatus and samples were collected on-line and injected every 15 min from a 20 ⁇ sample-loop.
  • Dopamine, DOPAC, and 5-HIAA were quantified in the samples according to previously described methods. Before initiating any pharmacological manipulation, baseline samples were taken until there was less than 30% variation between the samples.
  • Catalepsy test Rats were placed in a plastic bowl which measured 30 cm in diameter and 20 cm in height. After 10 minutes, the rats were placed on a vertical metal grid (grids 1 cm square) which measured 15 x 40 cm (width x heigth). The time until the first paw movement and the time to get off the grid were measured in three consecutive sessions. Thereafter, the rat was injected with either saline of test compound and left in the cage for another 30 minutes before catalepsy was retested.
  • Locomotor activity Directly after the second catalepsy test (see above), the animals were transferred to a locomotion chamber (Columbus Instruments, Ohio, USA) and locomotion was measured for 10 minutes (referred to as exploratory behavior due to the new environment). Apomorphine (1 mg/kg, s.c.) was subsequently injected and locomotion was measured for 30 minutes after the injection.
  • Rats were injected with thest compounds 15 min prior to the administration of 0.5 or 2 mg/kg amphetamine in their home cages. Immediately after the administration of amphetamine, the rats were transferred to a locomotor chamber (Columbus Instruments, Ohio, USA) and locomotion was measured for 60 min.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A compound of formula (I), or pharmaceutically acceptable acid addition salts thereof, wherein R1 is H, (C1-C8) alkyl or haloalkyl or hydroxyalkyl, alkenyl, alkynyl, cyclopropylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl; R2 is H, (C1-C8) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (C1-C8) haloalkyl, hydroxyalkyl, hydroxyalkyloxyalkyl or 1-(alkyl-2-imidazolidinone); X is NH, NR1, O, S, SO, SO2. The compounds of this invention possess affinity to one or several receptor systems, e.g. DA (D1-D4), α1, muscarinic (M1-M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7). The central nervous system disorders to be treated with the compounds of the present invention include psychoses-schizophrenia, autism, Tourette's syndrome, restless legs, Huntington's chorea, motion sickness, nausea, vomiting and severe anxiety.

Description

NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS.
Field of the Invention
The present invention is directed toward new sulfone ester analogues of iso-Clozapine [RN 1977-08-81 and their pharmaceutically acceptable salts, to processes for preparing such compounds, pharmaceutical preparations of such compounds and the use of such compounds in manufacture of a pharmaceutical preparation. Pharmaceutical preparations of these compounds are useful for central nervous system disorders, such as for a therapeutic effect on one or several of the following receptor systems: dopamine (DA; D1 - D4), 5-HT2A, 5-HT2C, α1 and muscarinic (M1 -M4) in mammals.
Background of the Invention
In psychotic/schizofrenic patients evidence indicates that the neurotransmission in the central nervous system (CNS) may be disturbed. These disturbances seem to involve the neurotransmitters dopamine (DA, D1 -D4 receptors), noradrenaline |NA, α1 receptors), acetylcholine (ACh, 1-M4 receptors) and 5-hydroxytryptamine (5-HT, 5-HT2A and 5- HT2C receptors).1 The drugs most frequently used in the treatment of psychoses are considered to act by altering the neurotransmission of one or several of these receptor systems. The mechanism of action for typical/classical anti-psychotic drugs used to treat psychosis is generally believed to be through blockage of DA D2 receptors.2, 3- 4 The atypical anti-psychotic drug Clozapine [RN 54241-01 -9] is an efficient anti-psychotic agent5 and binds with high affinity to DA D1 and D4 receptors and with moderate affinity to DA D2 and D3 receptors.6 In addition, Clozapine binds with high affinity to α1 receptors, muscarinic 1-M4 receptors and 5-HT2A and 5-HT2C receptors.6 Clozapine has a low propensity to induce extrapyramidal side-effects in humans and catalepsy in rats. In addition, Clozapine does not increase the prolactine levels, something which the classical neuroleptics (e. g. haloperidol and chlorpromazine) potently do. However, the use of Clozapine is hampered by rare incidences of a serious side-effect (blood toxicity, aranulocytosis) . B Expensive, continuous blood monitoring makes the use of Clozapine tedious and expensive and the use of Clozapine is therefore limited to psychotic patients not responding to the first and second choice of drug therapy.7
Several Clozapine analogues have been evaluated but, to date, no marketed compound with a Clozapine-like structure and Clozapine-like pharmacological and clinical profile, i. e. an analogue that is also free from side-effects, has been found.7
A very interesting and, for the present invention, a highly relevant fact is that the 2-CI isomer of Clozapine (iso-Clozapine, [RN 1977-08-81) has been classified as a typical neuroleptic agent.8 We surprisingly found that the iso-Clozapine sulfone analogues of the present invention display atypical neuroleptic properties.
Information Disclosure Statement.
The following documents could be important in the examination of this application:
iso-Clozapine references: 9, 10, 1 1
Loxapine references: 1 2, 13, 14, 15, 16, 17, 18, 19, 20, 21
Clothiapine references: 15, 16, 19, 21 , 22, 23, 24, 25, 26
Miscellaneous references over sulfonates: 27, 28, 29, 30, 31
Sufonate patent application reference: 32
Summary of the Invention
This invention is related to novel iso-Clozapine, Loxapine and Clothiapine analogues of Formula 1 :
Figure imgf000004_0001
or pharmaceutically acceptable acid addition salts thereof, wherein R* is H, (C*-C8) alkyl or haloalkyl (e. g. CF3 (triflates) and CF2CF3) or hydroxyalkyl, alkenyl, alkynyl. cyclopropylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyi; R2 is H, (C* -C8) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (CrC8) haloalkyl (e. g. CH2CF3 CH2CH2CF3, CH2CH2F,
CH2CH2CH2F), hydroxyalkyl, hydroxyalkyloxyalkyl (e. g. or 1-
Figure imgf000005_0001
(alkyl-2-imidazolidinone) (e. g. 1 -(CH2CH2-2-imidazolidinone) and 1 -(CH2CH2CH2-2- imidazolidinonel); X is NH, NR 0, S, SO, S02.
The compounds of this invention possess affinity to one or several receptor systems, e. g. DA (D1 -D4), ol , muscarinic (M1 -M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7). The central nervous system disorders to be treated with the compounds of the present invention include psychoses/schizophrenia, autism, Tourette's syndrome, restless legs, Huntington's chorea, motion sickness, nausea, vomiting and severe anxiety.
In a preferred embodiment, the invention is directed to compounds of Formula 1 wherein R. is haloalkyl R2 is H or CH3 and X is NH, 0 or S. A more preferred embodiment are compounds of Formula 1 wherein R - is CF3, R2 is H or CH3 and X is NH, 0 or S. An even more preferred embodiment are compounds of Formula 1 wherein R * is CF3, R2 is CH3 and X is NH, O or S. The most preferred compound is the compound of Formula I wherein R* is CF3, R2 is CH3 and X is NH.
An object of the invention is to provide compounds for therapeutic use, especially compounds having a therapeutic activity in the central nervous system. Another object is to provide compounds having an effect on receptors (DA (D1 -D4), α1 , muscarinic (M1 - M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7)) in mammals including man.
Processes for preparation of these compounds, their pharmaceutical use and pharmaceutical preparations employing such compounds constitute further aspects of the invention.
Detailed Description of the Invention
In appropriate situations, the proper stereochemistry is represented in the structural schemes.
As used herein the term (Cn-Cm) is inclusive such that a compound of (C|-C8) would include compounds of one to 8 carbons and their isomeric forms. The various carbon moieties are defined as follows: alkyl refers to an aliphatic hydrocarbon radical and - 4 - includes branched or unbranched forms such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neo-pentyl, n-hexyl, isohexγl, n-heptyl, isoheptyl, and n-octyl.
Alkenyl refers to a radical of an aliphatic unsaturated hydrocarbon having a double bond and includes both branched and unbranched forms such as ethenyl, 1 -methyl- 1-ethenyl, 1- propenyl, 2-propenyl, 1 -butenyl, 2-butenyl, 3-butenyl, 2-methyl-1-butenyl, l-pentenyl, 2- pentenyl, 3-pentenyl, 4-pentenyl, 1 -methyl-4-pentenyl, 3-methyl-1-pentenyl, 3-methyl-2- pentenyl, 1 -hexenyl, 2-hexenyl, 3-hexenyl , 4-hexenyl, 1 -methyl-4-hexenyl, 3-methyl-1 - hexenyl, 3-methyl-2-hexenyl, 1 -heptenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 1 -methyl-4- heptenyl, 3-methyl-1 -heptenyl, 3-methyl-2-heptenyl, 1 -octenyl, 2-octenyl or 3-octenyl. Cγcloalkyl refers to a radical of a saturated cyclic hydrocarbon such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexγl, cycloheptvl, or cyclooctyl. "Halogen" means fluorine, chlorine, bromine or iodine, preferably fluorine.
It will be apparent to those skilled in the art that compounds of this invention may contain chiral centers (e. g. R* and/or R2). The scope of this invention includes all enantiomeric or diastereomeric forms of Formula 1 compounds, either in pure form or as mixtures of enantiomers or diastereomers. The compounds of Formula 1 can contain one asymmetric carbon atom in the ring system (when X = SO). The therapeutic properties of the compounds may to a greater or lesser degree depend on the stereochemistry of a particular compound. Pure enantiomers, as well as enantiomeric or diastereomeric mixtures, are within the scope of the invention.
Both organic and inorganic acids can be employed to form non-toxic pharmaceutically acceptable acid addition salts of the compounds of this invention. Illustrative acids are sulfuric, nitric, phosphoric, hydrochloric, citric, acetic, lactic, tartaric, pamoic, methanesulfonic, ethanedisulfonic, sulfamic, succinic, cyclohexylsulfamic, fumaric, maieic and benzoic acid. These salts are readily prepared by methods known in the art.
The compounds of this invention may be obtained by one of the following methods described below, as outlined in the appropriate charts.
In clinical practice the compounds of the present invention will normally be administered orally, rectally or by injection, in the form of pharmaceutical preparations comprising the active ingredient either as a free base or as a pharmaceutically acceptable non-toxic, acid addition salt, such as the hydrochloride, lactate, acetate, methanesulfonate, sulfamate salt, in association with a pharmaceutically acceptable carrier. The use and administration to a patient to be treated in the clinic would be readily apparent to a person of ordinary skill in the art.
s In therapeutical treatment the suitable daily doses of the compounds of the invention are from about 0.1 mg to 2000 mg for oral application, preferably 0.5-500 mg, and 0.05 mg to about 100 mg for parenteral application, preferably 0.05-50 mg daily doses. The daily dose will preferably be administrated in individual dosages 1 -4 times daily and the dosage amounts are based on an individual having a weight of 70 kg. w
The compounds of Formula 1 of this invention, wherein R« is H, (C*-C8) alkyl or haloalkyl (e. g. CF3 (triflates) and CF2CF3) or hydroxyalkyl, alkenyl, alkynyl, cyclopropylalkyl, aryl, arγlalkγl, heteroaryl, heteroarylalkyl; R2 is H, (C1-C8) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (CrC8) haloalkyl (e. g. CH2CF3 CH2CH2CF3, CH2CH2F, is CH2CH2CH2F), hydroxyalkyl, hydroxyalkyloxyalkyl (e. g. or 1-
Figure imgf000007_0001
(alkyl-2-imidazolidinone) (e. g. 1 -(CH2CH2-2-imidazolidinone) and 1 -(CH2CH2CH2-2- imidazolidinone)); X is NH, NR., 0, S, SO, S02, display binding affinity at the receptor systems: DA (D1-D4), α1 , muscarinic (M1 -M4) and 5-HT (5-HT2A, 5-HT2C and 5-HT7) in mammals including man. These compounds are particularly effective atypical anti- 0 psychotic (anti-schizophrenic) agents. Other uses for these compounds include autism, Tourette's syndrome, restless legs, Huntington's chorea, motion sickness, nausea, vomiting and severe anxiety.
The compounds of this invention also have high oral potency and a long duration of 5 action.
The utility of the compounds of this invention to treat central nervous system disorders can be shown in behavioral and biochemical activity in rats.
0 Effect on DA release in rats as measured bv microdialvsis in freely moving animals.
The biochemical effects of the compounds of this invention can be monitored with the microdialγsis technique in freely moving animals (see under Experimental Section below).
5 Monitoring potential catalepsy in rats,
The propensity of the compounds of this invention to induce catalepsy was measured in rats, who were placed temporarily on a vertical grid (see under Experimental Section below).
Monitoring of potential inhibition of locomotor activity in rats treated with Apomorphine or Amphetamine.
The ability of the componds of this invention to reduce the Apomorphine or Amphetamine induced increase in locomotor activity was measured in rats with motilitγ meters (see under Experimental Section below).
Experimental Section
Syntheses
Starting materials (the corresponding phenols) for the claimed target compounds may be obtained by the methods known in the art.
' H and 13C NMR spectra were recorded at 200 and 50.3 MHz, respectively, on a Varian Gemini 200 spectrometer. CDCI3 was employed as the solvent unless otherwise stated. Chemical shifts are given in units (ppm) and relative to TMS or deuterated solvent. IR spectra were obtained on a ATI-Mattson spectrometer. Elemental analyses were performed in the Micro Analytical Department of the University of Groningen or at other approved laboratories. The chemical ionization (CD mass spectra were obtained on a Finnegan 3300 system or a UNICAM Automass 150 GC/MS system, equipped with a UNICAM GC, 610 Series. Melting points were determined on a Electrothermal digital melting point apparatus and are uncorrected. Specific optical rotations were measured in methanol (c = 1.0) at 21 °C on a Perkin Elmer 241 polarimeter.
All methoxylated starting compounds were prepared according to the literature procedures.10, 18, 21 , 33, 34 Chemicals used were commercially available (Aldrich and/or Sigma) and were used without further purification.
Parts of the synthetic methods, not necessarily the preferred route, for forming the sulfone esters from the corresponding phenols are exemplified in Scheme 1 : Scheme 1
Figure imgf000009_0001
Figure imgf000009_0002
Suitable starting materials, either the phenols or the OMe-analooues. which can be dealkylated to the corresponding phenols with e. g. AICI3 in ethylmercaptane at room temperature, for this invention are known:
2-Hydroxy-1 1 -(4-methyl-1 -piperaziπyl)-5H-dibenzolb,e][1 ,4]diazepine; 2-hydroxy-8- dechloroclozapine; RN [156632-07-4]
Figure imgf000010_0001
2-Methoxy- 1 1 -(4-methγl-1 -piperazinyl)-5H-dibenzolb,e][ 1 ,4]diazepine; 2-methoxy-8- dechloroclozapine; RN [95316-97-5]
Figure imgf000010_0002
2-Methoxy-1 1 -(4-methyl-1 -piperazinyl)-dibenzo[b,f][1 ,4]oxazepine; RN [13745-79-4]
Figure imgf000010_0003
2-Methoxy-11-(4-methyl-1-piperazinyl)-dibenzo[b,f][1,4]thiazepine; RN [13745-79-4]
Figure imgf000011_0001
The following intermediates, useful for the preparation of compounds of the present invention are known:
2-Methoxγ-dibeπz[b,f][1,4]oxazepin-11(10H)-one RN [60287-33-4]35
2-Hydroxy-dibenz(b,f][1,4]oxazepin-11(10H)-one RN [60287-08-3]35
2-Methoxγ-dibenzolb,f][1,4]thiazepin-11(10H)-one RN [3158-77-8]36, 37
Figure imgf000012_0001
Figure imgf000013_0001
Scheme 4
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
Figure imgf000014_0004
Figure imgf000014_0005
Scheme 5
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
Figure imgf000015_0004
Synthesis of 2-Trifluoromethanesulfonyloxy-1 1-(4-methyl-1 - piperazinyl)-5H- dibenzo[b,e][1 ,4]diazepine (12).
2-(2-Nitrophenyl)amino-5-methoxybenzoic acid (3). A mixture of 5-methoxγ-2- aminobenzoic acid (1 , 6.68 g, 40 mmol), 2-bromo-nitrobenzene (2, 8.08 g, 40 mmol) and 10 mL of amγl alcohol was heated to 80 °C with stirring. To the homogeneous solution was immediately added copper powder (60 mg) and K2C03 (5.7 g). The reaction temperature was increased in order to distill off the water generated in the reaction and the amγl alcohol. The reaction mixture was then kept at 210 °C for 3 h. After cooling, the reaction mixture was quenched with 200 mL of water and 100 mL of 4 N aq HCI at room temperature. The precipitate was removed by filtration and the solution was extracted with ethyl acetate. The organic layers were combined and concentrated. The residue was purified by flash chromatography (Si02, ethyl acetate as eluent) to afford 6.8 g (59 %) of the title compound.
2-(2-Arninophenyl)amino-5-methoxybenzoic acid (4). Compound 3 [2-(2-nitrophenyl)amino- 5-methoxybenzoic acid] ( 1 .8 g) in THF (50 mL) was hydrogenated with a catalytic amount of palladium on carbon in a Parr-shaker apparatus under 4.5 atm H2 (g) at 60 °C for 4 h to afford 1 .4 g (88 %) of the title compound.
2-Methoxy-5,10-dihydro-1 1 -oxo-dibenzo(b,e][1 ,4]diazepine (5). Intermediate 4 [2-(2- aminophenyl)amino-5-methoxybenzoic acid] (3.0 g) in 100 mL of xylene was refluxed for 40 h. After evaporation the residue was purified by flash chromatography (Si02, hexane and ethyl acetate, 4: 1 then 1 : 1 , as eluents) to afford 2.2 g (79 %) of the title compound. MS (El) shows M + at m/e = 240.
2-Methoxy-1 1 -(4-methyl-1 -piperazinyl)-5H-dibenzo[b,el[1.4l-diazepine (7, RN [95316-97- 51). Intermediate 5 (200 mg, 0.83 mmol), phosphorus oxychloride (POCI3) (3 mL), toluene (5 mL) and N,N-dimβthylaniline (0.5 mL) were combined and heated to reflux for 3 h. The solvents of the mixture were evaporated under vacuum to afford the iminochloride intermediate 6, which was used in the next-step reaction without further purification.
Above iminochloride 6 in 3 mL of toluene and 2 mL of N-methylpiperazine were refluxed for 3 h. After evaporation the residue was taken up with chloroform, washed with 2N aq NaOH and purified by flash chromatography (Si02, ethyl acetate then THF as eluents) to afford 214 mg (80 %) of the title compound. MS (El) shows M + at m/e = 322. 2-Hydroxy-11-(4-methyl-1-piperaziπyl)-5H-dibenzo[b,e][1,4]-diazepine (8, RN [156632-07- 4]). 2-Methoxy-1 1-(4-methγl-1-piperazinyl)-5H-dibeπzo[b,el[1 ,4]diazepine (7) (200 mg, 0.62 mmol) in 3 mL of EtSH was treated with AICI3 (1 .5 g) with stirring at room temperature for 4 h. The mixture was quenched with 30 mL of ice-water. The pH value of 5 the solution was adjusted to pH 8 with 2N aq NaOH. The solution was extracted with chloroform (4 x 30 mL). The organic layers were combined, dried over MgS04 filtered, and the solvents were evaporated. The light yellow solid residue was recrystallized from ethyl acetate to afford 140 mg (73 %) of the title compound as fine crystals: mp 266 °C, with all of the expected spectra including IR, -H NMR, 1 3C NMR. MS (El) shows M at w m/e = 308.
2-Hydroxy-5,10-dihydro-1 1 -oxo-dibenzo[b,e][1 ,4]diazepine (9). 2-Methoxy-5,10-dihγdro- 1 1 -oxo-dibenzo[b,eH1 ,4]diazepine (5) (600 mg, 2.5 mmol) in 4 mL of EtSH was treated with AICI3 (1 .6 g) with stirring at room temperature for 4 h. The mixture was quenched 15 with 20 mL of ice-water and then 10 mL of 4N aq HCI. The solution was extracted with chloroform (2 x 20 mL) and ethγlacetate (2 x 20 mL). The combined organic layers were dried over MgS04, filtered, and the solvents were evaporated. The solid residue was recrystallized from ethyl acetate to afford 505 mg (88 %) of the title compound as fine crystals: mp 280 °C, with all the expected spectra including IR, *H NMR, 1 3C NMR. MS 0 (El) shows M at m/e = 226.
2-Trifluoromethanesulfonyloxy-5,10-dihydro-11-oxo-dibenzo-[b,e][1 ,4]diazepine (10). 2- Hydroxy-5, 10-dihydro-1 1-oxo-dibenzo[b,e][1 ,4]-diazepine (9) (400 mg, 1.77 mmol) in 10 mL of dioxane was treated with N-phenyl-trifluoromethanesulfonimide (800 mg) in the 5 presence of 2 mL of triethylamine with strirring at room temperature overnight. After evaporation of the solvents, the residue was purified by flash chromatography (Si02, hexane and ethyl acetate, 9:1 then 4: 1 , as eluents). Recrystallization from hexane and ethyl acetate (10: 1 ) afforded 380 mg (60 %) of the title compound as light yellow crystals. MS (El) shows M + at m/e = 358. 0
2-Trifluoromethanesulfonyloxy-11-(4-methyl-1-piperazinyl)-5H-dibenzo[b,e][1 ,4]diazepine (12). Method A. 2-Hydroxy-1 1 -(4-methyl-1 -piperazinyl)-5H-dibenzo[b,e][1 ,4]diazepine (8) (180 mg, 0.58 mmol) in 10 mL of CH2CI2 was treated with N-phenyl- trifluoromethanesulfonimide (300 mg, 0.83 mmol) in the presence of 1 mL of 5 triethylamine with strirring at room temperature overnight. After evaporation the residue was purified by flash chromatography (Si02; hexane and ethyl acetate, 1 :1 ; then ethyl acetate as eluents). Recrγstallization from hexane and ethyl acetate (15:1 ) afforded 200 mg (78 %) of the title compound as fine crystals: mp 160 °C, with the expected spectra data including IR, 1H NMR, 1 3C NMR. MS (El) shows M + at m/e = 440.
Method B. 2-Trifluoromethanesulfonyloxy-5,10-dihydro-1 1-oxo-dibenzo-[b,e][1 ,4]diazepine (10) (260 mg, 0.72 mmol), phosphorus oxychloride (P0CI3) (4 mL), toluene (10 mL) and N,N-dimethylaniline (0.5 mL) were combined and heated to reflux for 3 h. The mixture was evaporated under vacuum to afford the corresponding iminochloride intermediate 11 , which was used in the next-step reaction without further purification.
The iminochloride 11 in 10 mL of toluene was treated with 3 mL of N-methylpiperazine under refluxing for 3 h. After evaporation of the solvents, the residue was taken up in chloroform, washed with 2N aq NaOH and purified by flash chromatography and recrystallized, as described in Method A above, to afford 230 mg (73 %) of the title compound 12.
2-Methanesulf onyloxy-1 -(4-methyl-1 -piperazinyl)-5H-dibenzo-[b.e][ 1 ,4]diazepine (15).
2-Methanesulfonyl-5,10-dihydro-11-oxo-dibenzo[b,e][1 ,4]diazepine (13). 2-Hydroxy-5,10- dihydro-1 1 -oxo-dibenzo[b,e][1 ,4]diazepine (9) (50 mg, 0.22 mmol) in 6 mL of dichloromethanβ was treated with methanesulfonylchloride (34 mg, 0.30 mmol) in the presence of 0.2 mL of triethylamine at -78 °C. The mixture was stirred for 1 h, in order to allow the reaction temperature to reach room temperature, quenched with water and extracted with chloroform. Workup afforded 50 mg (76 %) of the title compound. MS (El) shows M+ at m/e = 304.
2-Methanesulfonyloxy-11-(4-methyl-1-piperazinyl)-5H-dibβnzo[b.e][1,4]diazepine (15). 2- Methanesulfonyloxy-5,10-dihydro-1 1-oxo-dibenzo(b,el[1 ,4]diazepine (13) (50 mg, 0.16 mmol), phosphorus oxychloride (POCI3) (2 mL), toluene (3 mL) and N,N-dimethylaniline (0.1 mL) were combined and heated to reflux for 3 h. The mixture was evaporated under vacuum to afford the iminochloride intermediate 14, which was used in the next reaction step without further purification.
This iminochloride 14 in 3 mL of toluene was treated with 1 mL of N-methylpiperazine under refluxing for 3 h. Similar workup as described for the preparation of 12 afforded 32 mg (50 %) of the title compound as fine crystals: mp 173-177 °C. MS (El) shows M + at m/e = 386. Preparations of 2-(4-methylphenylsulfonyloxy)-1 1-(4-methγl-1-piperazinyl|-5H- dibenzo[b,e][1 ,4]diazepine (16), 2-phenγlsulfonyloxy-1 1-<4-methyl-1 -piperazinyl)-5H- dibenzo[b,e][1 ,4]diazepine (17) and 2-(2-thienylsulfonyloxy)-1 1 -(4-methγl-1 -piperazinyl)- 5H-dibenzo[b,e][1 ,4)diazepine (18) were performed by the same procedure as for the preparation of 2-trifluoromethanesulfonyloxγ-1 1 -(4-methyl-1 -piperazinyl)-5H- dibenzo[b,e][1 ,4]diazepine (12) (Method A) from 2-hydroxy-1 1 -(4-methyl-1 -piperazinyl)- 5H-dibenzo[b,e][1 ,4]diazepine (8), however, using the sulfonation procedure with appropriate sulfonylchloride, as described for the preparation of compound 13.
Mass spectrometrγ:
Compound no M + at m/e
16 462
17 448 18 454
2-Tπfluoromethanesulf onyloxy-11 -(4-methylpiperazino)-5H-dibβnzo[b,f ][ 1 ,4)oxazepine ( 19)
(Scheme 4). 2-Methoxy-5,10-dihydro-1 1 -oxo-dibenzo[b,f][1 ,4]oxazepine (RN [60287-33- 4]) was prepared according to the literature. 35
2-Hydroxy-5,10-dihydro-1 1 -oxo-dibenzo[b,f][1,4loxazepine (RN [60287-08-3] ). 2- Methoxy-5,10-dihγdro-1 1 -oxo-dibenzo[b,e][1 ,4]oxazepine (1.2 g) in 10 mL of EtSH and 10 mL of dichloromethane was treated with AICI3 (5.0 g) with stirring at room temperature for 6 h. The solvent of the mixture was evaporated by rotavapor at reduced pressure, quenched with 20 mL of ice-water and then 20 mL of 4N aq HCI. A white precipitate was collected, washed with water and dried under vacuum. The product (1 .0 gram) showed the expected spectra including IR, 'H NMR, and MS (El, m/z 227).
2-Trifluoromethanesulfonγloxy-5,10-dihydro-11 -oxo-dibenzo[b,f][1 ,4]-oxazepine. 2- Hydroxy-5,10-dihydro-1 1 -oxo-dibenzo[b,e][1 ,4]oxazepine (1 .0 g) in 20 mL of dichloromethane and 5 mL of triethylamine was cooled to -78 °C, treated with triflic anhydride (1.54 mL, 7 mmol). The mixture was stirred for 3 h to allow the temperature back to room temperature, quenched with 3 mL of 50% methanol in water and concentrated. The residue was flashed on silical-gel column (hexane and ethyl acetate, 9:1 then 1 :1 , as eluents) to afford 1.3 g of the title compound as white crystal: MS (El) m/z 359. 2-Trifluoromethanesulfonyioxy-1 1 -(4-methylpiperazino)-5H-dibenzo-[b,f][1 ,4]oxazepinβ. 2- Trifluoromethanesulfonyl-5,10-dihydro-1 1-oxo-dibenzo-[b,e][1 ,4]oxazepine (1 .0 g), P0CI3 (5 mL), toluene (10 mL) and N,N-dimethyl-aniline (1.0 mL) were combined and heated to reflux for 3 h. The solvent of the mixture was evaporated under vacuum to afford iminochloride intermediate, which was dissolved in 10 mL of toluene and was treated with 5 mL of N-methylpiperazine under refluxing conditions for 3 h. After evaporation the residue was purified by flash chromatography (Si02, 9:1 hexane and ethyl acetate then pure ethylacetate as eluents) and recrystallization (n-hexane) to afford 1 .01 g of the title compound: mp 1 17-1 18 °C; MS(EI) m/z 441 ; 1H NMR (CDCI3) and elements (C. H. N) were analyzed.
2-Trifluoromethanesulfonyloxγ-11-(4-methylpiperazino)-5H-dibenzo-[b,f][1,4]thiazepine (20) (Scheme 5). 2-[(4-Methoxyphenyl)thio]benzoic acid (RN 19862-91 -0) was prepared according to the literature.
2-[(4-methoxyphenyl|thio]benzoyl azide. 2-[(4-methoxγphenyl)thiolbenzoic acid (14 g, 53.8 mmol) was treated with 50 mL of S0CI2 and 1 mL of DMF at refuxing for 1 h. The mixture was evaporated under vacuum and the solid residue was dissolved into dry acetone (100 mL). The resulting solution was added dropwise to a cooled 30 % aq NaN3 (60 mL) during 30 min. The suspension was stirred at 0 °C for 1 h, diluted with water
(400 mL), filtered, washed well with water and dried overnight under vacuum to afford 14 g of the title compound as a fine solid. IR and GC-MS were analyzed. Mass spectra (El) observed m/z at 257 (M + -N2, isocyanation product under GC conditions: 150-300 °C/10
°C rate program) conditions.
2-Hydroxy-dibenzo[b,f][1,4]thiazβpinβ-11(10H)-one. 2-[(4-methoxγ-phenyl)thio)benzoyl azide (1 .4 g) in 20 mL of dichlorobenzene was added to a suspension of AICI3 (2.7 g) in o-dichlorobenzene (50 mL) with stirring. The mixture was heated to reflux for 15 min, cooled to room temperature. CHCI3 (150 mL) was added, extracted with 4 N aq HCI (2 x 50 mL). The organic layer was concentrated under vacuum and the residue was purified by flash chromatography (Si02, ethyl acetate as eluent) to afford 240 mg of the title compound. Mass spectra observed m/z at 243 (M + ).
2-Triflatβ-dibenzo[b,f][1,4]thiazepine-11(10H)-one. 2-Hydroxγ-dibenzo-[b,f][1 ,4]thiazepine- 1 1 (10H)-one (100 mg) in CH2CI2 was cooled to -78 °C and treated with triflic anhydride (0.13 mL) in the presence of triethylamine (1 .0 mL). After stirring for 2 h the mixture was mixed with Si02 (3 mL), evaporated and flashed (Si02 column, hexane and ethyl acetate as eluents) to afford 130 mg of the title compound. MS (El) at m/z 375 (M + ).
2-Trif luoromethanesulf onyloxy- 1 1 -(4-methhylρiperazino)-5H-dibenzo-[b ,f ] [ 1 ,4]thiazepine . 2-Trifluoromethanesulfonyloxy-5, 10-dihydro-1 1 -oxo-dibenzo-[b,e][1 ,4)thiazepine (80 mg), POCI3 (1 mL), toluene (1 mL) and N,N-dimethyl-aniline (0.1 mL) were combined and heated to reflux for 3 h. The solvent of the mixture was evaporated under vacuum to afford iminochloride intermediate, which was dissolved in 1 mL of toluene and was treated with 1 mL of N-methylpiperazine under refluxing conditions for 3 h. After evaporation the residue was purified by flash chromatography (Si02, 9:1 hexane and ethyl acetate then pure ethγlacetate as eluents) and recrγstallization from hexane to afford 33 mg of the title compound: MS(EI) m/z 457; 1 H NMR (CDCI3) and IR were analyzed.
Derivatives with the -CH2CH2-2-imidazolidinone and -CH2CH2CH2-2-imidazolidinone substutυent R2 (see Formula I).
These derivatives are synthesized from the corresponding N-methyl-piperazine derivatives (e. g. compounds 12, 15-20) by first performing a demethγlation with 1 -chloroethyl chloroformate39 and then an alkγlation with the appropriate alkγlating agent (e. g. 1 -(2- chloroethγl)-2-imidazoiidinone and 1 -(3-chloroprophyl)-2-imidazolidinone, respectively), refluxing over night in methyl isobutyl ketone (MIBK) in the presence of dry, ground K2C03 (s) and a catalytic amount of Kl (s).1
Pharmacology
Microdialysis experiments: Adult male Wistar rats (Harlan Centraal Proefdier Bedrijf, The Netherlands) were used. Until implantation of the dialysis probe, the rats were housed in groups of 5-10 animals in plastic cages. The rats were kept in a room maintained under constant temperature (21 °C) and humidity (40%) on a 12:12 dark:light cycle (07:00 on and 19:00 off) with food and water available ad libitum. The rats weighed between 250- 350 g at the time of the microdialysis probe implantation. Animal procedures were conducted in accordance with guidelines published in the NIH Guide for Care and Use of Laboratory Animals and all protocols were approved by an Institutional Animal Care and Use Committee.
Drugs were administered s.c. in a volume of 1 mL/kg in distilled water. Sometimes a drop of acetic acid was added to improve the solubility of the test compound. The microdialysis probes used in the present investigation were of a vertical, concentric design: A piece of fused silica capillary (Polymicro Technologies, Phoenix, AZ, USA) with I.D. 50 μm and O.D. 1 15 μm was threaded through a piece of PE-20 tubing (Clay Adams, Parsippanγ, NJ, USA) 0.5 cm from the end. A dialysis membrane (8 mm) (Hospal, Meyzieu, France) with an I.D. of 250 μm and an O.D. of 300 μπrt was slipped over the fused silica, glued into the PE-20, and closed at the end with epoxy (Devcon, Danvers, MA, USA). The fused silica was then brought into close apposition to the closed end of the dialysis membrane and fixed into position by epoxy at the junction of the silica and PE- 20. The active membrane region was 2 mm in length and the region above the active area was coated with a thin layer of epoxy. After cutting the silica and PE-20 tubing 5 mm from the junction of the silica and PE-20, an inlet (a 25 G stainless steel needle that was broken at the bevel and from the plastic fixture) was glued with epoxy over the silica. The other end of the PE 20 served as the outlet.
The animals were anesthetized with chloral hydrate (400 mg/kg) and a microdialysis probe was implanted into the CPu at the following coordinates: (flat skull) AP 1.0 mm, ML + 2.5 mm relative to bregma, and -6.0 mm below the dura and/or nucleus accumbens at the following coordinates: (flat skull) A. P. 2.5 mm, ML -1.3 relative to bregma, and -7.3 mm below the dura. The probes were lowered using an electrode carrier (Model 1760, David Kopf, Tujunga, CA, USA) at a speed of approximately 400 μm/miπ and secured to the skull with fast-curing dental cement on three skull screws. At the start of the experiment, 16 hours after surgery, artificial cerebrospinal fluid (aCSF) consisting of (mmol/L): NaCI 147, KCI 2.5, CaCI2 1 .3, and MgCI 0.9, pH 7.4 was perfused through the probe with a syringe pump (Harvard Apparatus inc, South Natick, MA, USA). The outlet of the probes were connected to the valve of an HPLC apparatus and samples were collected on-line and injected every 15 min from a 20 μ sample-loop. Dopamine, DOPAC, and 5-HIAA were quantified in the samples according to previously described methods. Before initiating any pharmacological manipulation, baseline samples were taken until there was less than 30% variation between the samples.
Behavioral experiments: Adult male Wistar rats (Harlan Centraal Proefdier Bedrijf, The Netherlands) were used. The rats were housed in groups of 12 animals in plastic cages. The rats were kept in a room maintained under constant temperature (21 °C) and humidity (40%) on a 12:12 dark:light cycle (07:00 on and 19:00 off) with food and water available ad libitum. The rats weighed between 200-250 g at the start of the experiment. Animal procedures were conducted in accordance with guidelines published in the NIH Guide for Care and Use of Laboratory Animals and all protocols were approved by an Institutional Animal Care and Use Committee.
Catalepsy test: Rats were placed in a plastic bowl which measured 30 cm in diameter and 20 cm in height. After 10 minutes, the rats were placed on a vertical metal grid (grids 1 cm square) which measured 15 x 40 cm (width x heigth). The time until the first paw movement and the time to get off the grid were measured in three consecutive sessions. Thereafter, the rat was injected with either saline of test compound and left in the cage for another 30 minutes before catalepsy was retested.
Locomotor activity: Directly after the second catalepsy test (see above), the animals were transferred to a locomotion chamber (Columbus Instruments, Ohio, USA) and locomotion was measured for 10 minutes (referred to as exploratory behavior due to the new environment). Apomorphine (1 mg/kg, s.c.) was subsequently injected and locomotion was measured for 30 minutes after the injection.
Results
TABLE 1. Summary of the pharmacological characterization of compound 12, 15, 19, 20, clozapine, olanzapine, and haloperidol.
Figure imgf000024_0001
Footnotes Table 1 : Interaction with MK801 : Rats were injected with test compounds 15 min prior to the administration of 0.25 mg/kg MK801 in their home cages. Immediately after the administration of MK801 , the rats were transferred to a locomotor chamber (Columbus Instruments, Ohio, USA) and locomotion was measured for 30 min.
Interaction with AMPHETAMINE: Rats were injected with thest compounds 15 min prior to the administration of 0.5 or 2 mg/kg amphetamine in their home cages. Immediately after the administration of amphetamine, the rats were transferred to a locomotor chamber (Columbus Instruments, Ohio, USA) and locomotion was measured for 60 min.
References
1 . Liegeois, J. F. F.; Rogister, F. A.; Bruhwyler, J.; Damas, J .; Nguyen, T. P.; Inarejos, M. 0.; Chleide, E. M. G.; Mercier, M. G. A.; Delarge, J. E.
Pyridobenzoxazepine and Pyridobenzothiazepine Derivatives as Potential Central Nervous System Agents: Synthesis and Neurochemical Study. J. Med. Chem. 1994, 37, 519-25.
2. Carlsson, A.; Lindqvist, M. Effect of Chlorpromazine or Haloperidol on formation of 3-methoxytyramine and norepinephrine in the mouse brain. Acta Pharmacol.
Scand. 1963, 20, 140-144.
3. Snyder, S. H. DA hypothesis of schizophrenia. Am. J. Psychiatry 1976, 133, 197.
4. Sedvall, G. Monoamines and schizophrenia. Acta Psychiat. Scand. 1990, 82 (Sυppl. 358), 7-13.
5. Povlsen, U. J.; Noring, U.; Fog, R.; Gerlach, J. Tolerability and Therapeutic effect of Clozapine. Acta Psychiatr. Scand. 1985, 71, 176-185.
6. Phillips, S. T.; De Paulis, T.; Baron, B. M.; Siegel, B. W.; Seeman, P.; Van Tol, H. H. M.; Guan, H.-C; Smith, H. E. Binding of 5H-Dibenzo[b,e][1 ,4)diazepine and Chiral 5H-Dibenzo[a,d)cγcloheptene Analogues of Clozapine to Dopamine and
Serotonin Receptors. J. Med. Chem. 1994, 37, 2686-2696.
7. Marder, S. R.; Van Putten, T. Who Should Receive Clozapine? Arch. Gen. Psychiatry 1988, 45, 865-867.
8. Bϋrki, H. R.; Sayers, A. C; Ruch, W.; Asper, H. Effects of Clozapine and other Dibenzo-Epines on Central Dopaminergic and Cholinergic Systems. Structure- Activity Relationships. Arzneim.-Forsch. 1977, 27, 1561 -1565.
9. Howell, C. F.; Hardy, R. A. J.; Quinones, N. Q. Patent: 1 1 -Tertiary- - 24 - aminodibenz[b,f][ 1 ,4]oxazepines and thiazepines <FR 1 508536 680105 PRAI US 6601 17 (20 pp.)). Fr. Patent 1966.
10. Schmutz, J .; Hunziker, F.; Kuenzle, F. M. Patent: Substituted dibenz[b,f][1 ,4]oxazepines (CH 436297 671 1 15 (3 pp.)). Swiss Patent Coden: Swxxas 1964.
1 1 . Schmutz, J.; Hunziker, F.; Fischer, E. Patent: Diazepine and thiazepine derivatives. S. African patent application, 29 pp., PI ZA 7106668 730405, Al ZA 71-6668 71 1005 1971.
12. Krusteva, I.; Vitev, M.; Denkova, E.; Georgiev, A.; Evstatieva, A.; Zikolova, T. Synthesis of 2-chloro- 1 1 -(4-methyl- 1 -piperazinγl)dibenz[b,f ][ 1 ,4]oxazepine succinate. I. Preparation of 2-chloro-1 1 -(4-methyl-1 - piperazinyl)dibenz[b,fl[ 1 ,4]oxazepine. Farmatsiya (Sofia) 1987, 37, 1 -6.
13. Izquierdo, S. M.; Martin, E. P. U. Patent PI ES 490495 A1 810516, Al ES 80- 490495 80041 1 : Dibenzo[b,f][ 1 ,4]oxazepine and its salts. Span. Patent 1980. 14. Howell, C. F.; Hardy, R. A. J.; Quinones, N. Q. Patent PI JP 51041639 B4
761 1 1 1 Showa, Al JP 67-3069 6701 17: 1 1 -Aminodibenz[b,f][ 1 ,4]oxazepines and -thiazepines. Japan. Patent 1967.
1 5. Schneider, J. Patent PI DE 2316438 73101 1 , PRAI CH 72-4898 720404: Piperazinγl-substituted dibenzoxazepines and similar compounds. Ger. Often. Patent 1972.
16. Sheely, R. M.; Allen, G. R. J. Patent PI GB 1 164360 690917, PRAI US 661 21 6: Analgesic, antidepressant and tranquillizing 1 1 - aminodibenz[b,f][1 ,4]oxazepines and -thiazepines. Brit. Patent 1966.
1 7. Howell, C. F.; Hardy, R. A. J.; Quinones, N. Q. Patent PI US 3444169 690513, Al US 6601 17: Tranquilizing and antidepressant 1 1 - aminodibenz[b,f][1 ,4]oxazepines and thiazepines. U.S. Patent 1966.
18. Howell, C. F.; Hardy, R. A. J.; Quinones, N. Q. Patent PI FR 1508536 680105, PRAI US 6601 17: 1 1 -Tertiary-aminodibeπz(b,f][ 1 ,4]oxazepines and thiazepines. Fr. Patent 1966, 1 9. Coppola, J. A. Patent PI US 3412193 681 1 19, Al US 651213: 1 1 -(4-Methyl-
1 -piperazinyl)dibeπz[b,f][1 ,4]oxazepines or -thiazepines. U.S. Patent 1965.
20. Schmutz, J.; Hunziker, F.; Kuenzle, F. M. Patent PI CH 436297 671 1 15, Al CH 640527: Substituted dibenz[b,f][1 ,4]oxazepines. Swiss Patent 1964.
21 . Schmutz, J.; Hunziker, F.; Stillβ, G.; Lauener, H. (Clothiapine-OMe ref) Chemical structure and pharmacological activity of a new group of tricyclic neuroleptics. XI.
Heterocycles with seven atoms. Chim. Ther 1967, 2, 424-9. 22. Nagarajaπ, K.; Kulkarni, C. L.; Venkateswarlu, A.; Shah, R. K. Condensed heterotricycles. Dibenz[b,f][1 ,4]oxazepin-1 K10H)- thiones, 1 1 -substituted dibenz[b,f][1 ,4]oxazepines, and dibenz[b,f][1 ,4]thiazepine analogs. Indian J. Chem 1974, 12, 258-62. 23. Schmutz, J.; Hunziker, F.; Fischer, E. Patent PI ZA 7106668 730405, Al ZA
71 -6668 71 1005: Diazepine and thiazepine derivatives. S. African Patent 1971. 24. Schmutz, J.; Hunziker, F. Patent PI CH 476753 690930, Al CH 620608:
Analgesic and sedative 1 1 -(substituted-amino)dibenzo[b,f][1 ,4]thiaze pines. Swiss Patent 1962. 25. Howell, C. F.; Hardy, R. A. J.; Quinones, N. Q. Patent: PI FR 1508536 680105, PRAI US 6601 17: 1 1 -Tertiarγ-aminodibenz[b,f][1 ,4]oxazepines and thiazepines. Fr. Patent 1966. 26. Schmutz, J.; Hunziker, F.; Fischer, E. Patent PI CH 422793 670429, Al CH 610720: Dibenzo[b,f][1 ,4]thiazepines. Swiss Patent 1961. 27. Sonesson, C; Lin, C. H.; Hansson, L.; Waters, N.; Svensson, K.; Carlsson, A.;
Smith, M. W.; Wikstrom, H. Substituted (S)-Phenγlpiperidines and Rigid Congeners as Preferential Dopamine Autoreceptor Antagonists: Synthesis and Structure- Activity Relationships. J. Med. Chem. 1994, 37, 2735-53.
28. Haadsma-Svensson, S. R.; Smith, M. W.; Lin, C.-H.; Duncan, J. N.; Sonesson, C; Wikstrom, H.; Waters, N.; Carlsson, A.; Svensson, K. Synthesis and biological activity of C-5 modified derivatives of ( + )-AJ76 and ( + )-UH232: increased dopamine D3 receptor preference and improved pharmacokinetic properties. Bioorg. Med. Chem. Lett 1994, 4, 689-94.
29. Sonesson, C; Boije, M.; Svensson, K.; Ekman, A.; Carlsson, A.; Romero, A. G.; Martin, I. J.; Duncan, J. N.; King, L. J.; Wikstrom, H. Orally active central dopamine and serotonin receptor ligands: 5-, 6-, 7-, and 8- [[(trifluoromethyl)sulfonyl]oxy]-2-(di-n- propylaminoltetralins and the formation of active metabolites in vivo. J. Med. Chem. 1993, 36, 3409-16.
30. Sonesson, C; Waters, N.; Svensson, K.; Carlsson, A.; Smith, M. W.; Piercey, M. F.; Meier, E.; Wikstrom, H. Substituted 3-phenylpiperidines: new centrally acting dopamine autoreceptor antagonists. J. Med. Chem. 1993, 36, 3188-96.
31. Svensson, K. A. I.; Wikstrom, H. V.; Carlsson, P. A. E.; Boije, A. M. P.; Waters, R. N.; Sonesson, C. A.; Stjernlόf, N. P.; Andersson, B. R.; Hansson, L. O. Patent PRAI US 91 -686799 910417: Centrally acting substituted phenylazacycloalkanes. Pet Int. Appl 1991.
32. Wikstrom, H. Patent PRAI SE 91 -3745 91 1218: Preparation of (hetero)aryl triflates as nervous system agents. Pet Int. Appl 1991. 33. Schmutz, J.; Kuenzle, G.; Hunziker, F.; Gauch, R. Heterocycles with 7-membered rings. IX. 1 1 - Amino substituted dibenzo[b,f]-1 ,4-thiazepines and -oxazepines. Helv. Chim. Acta 1967, 50, 245-54.
34. Schmutz, J.; Hunziker, F.; Stille, G.; Lauener, H. Chemical structure and pharmacological activity of a new group of tricyclic neuroleptics. XI.
Heterocycles with seven atoms. Chim. Ther. 1967, 2, 424-9.
35. Brewster, K.; Clarke, R. J.; Harrison, J. M.; Inch, T. D.; Utley, D. Preparation of the eight monohydroxydibenz[b,f][ 1 ,4]oxazepin-1 1 (10H)-ones. J. Chem. Soc, Perkin Trans 1976, 7, 1286-90. 36. Bennett, 0. F.; Johnson, J.; Tramondozzi, J. Synthesis of 2- methoxydibenzo[b,f]( 1 ,4)-thiazepin-1 1 (10H)-one 5,5-dioxide. Org. Prep. Proced. Int 1974, 6, 287-93. 37. Schmutz, J.; Hunziker, F.; Schindler, 0.; Kuenzle, F. M. Patent PI US 3367930 680206, Al US 640924: Preparation of dibenzo[b,f][ 1 ,4]thiazepines and dibenz[b,f][ 1 ,4)oxazepines. U.S. Patent 1964.
38. Pelz, K.; Jirkovsky, I.; Adlerova, E.; Metysova, J.; Protiva, M. Collect. Czech.
Chem. Commun. 1968, 33(6), 1895-1910. 39. Olofson, R. A.; Martz, J. T.; Senet, J.-P.; Piteau, M.; Malfroot, T. A New Reagent for the Selective, High-Yield N-Dealkylation of Tertiary Amines: Improved Syntheses of naltrexone and Nalbuphine. J. Org. Chem. 1964, 2081 -2082.

Claims

C L A I M S
1. Compounds of Formula (1):
Figure imgf000029_0001
or pharmaceutically acceptable acid addition salts thereof, wherein R- is II, (C Cg) alkyl, haloalkyl, hydroxyalkyl, alkenyl, alkynyl, cyclopropylalkyl, aryl, arylalkyl, hetero- aryl, heteroarylalkyl; R2 is H, (C--Cg) alkyl, alkenyl, alkynyl, cyclopropylalkyl or (Cι-Cg) haloalkyl, hydroxyalkyl, hydroxyalkyloxyalkyl or 1-(alkyl-2-imidazolidinone) ; X is NH,
Figure imgf000029_0002
2. A compound according to claim 1, wherein Rj is selected from (C1-C3) alkyl and haloalkyl, R2 is H or CH3 and X is NH, O or S.
3. A compound according to claim 2, wherein R| is CII-j or
4. A compound according to claim 1, which is selected from
2-trifluoromethanesulfonyloxy-ll-(4-methyl-l-piperazinyl) -511- dibenzo[b,e] [1, 4]diazepine,
2-methanesulfonyloxy-ll-(4-methyl-l-piperazinyl) -5H-dibenzo [b,e] [1, ]diazepine, 2. o
2-trifluoro ethanesulfonyloxy-ll-(4-methyl-l-pιperazιno)- dibenzo[b, f] [1,4Joxazepine,
2-methanesulfonyloxy-ll-(4-methyl-l-piperazinyl)-dibenzo [b,e] [l,4]oxazepine,
2-trifluoromethanesulfonyloxy-ll-(4-methyl-l-piperazino)- dibenzo[b, f] [1, ]thiazepine,
2-methanesulfonyloxy-ll-(4-methyl-l-piperazino) -dibenzo- [b,e] [1,4]thiazepine,
2-(4-methylphenylsulfonyloxy) -11-(4-methyl-l-piperazinyl) - dibenzo[b,e] [1, ]diazepine,
2-phenylsulfonyloxy-ll-(4-methyl-l-piperazinyl) -5H-dibenzo- [b,e] [1,4]diazepine,
2-(2-thienylsulfonyloxy)-ll-(4-methyl-l-piperazinyl) -5H- dibenzo[b,e] [1, 4]diazepine and
the compound having the formula
Figure imgf000030_0001
5. Process for the preparation of a compound having the general formula (I) or a pharmaceutically acceptable acid addition salt thereof
Figure imgf000031_0001
wherein R( is H, (C|-C8) alkyl, haloalkyl, hydroxyalkyl, alkenyl, alkynyl, cyclopropylalkyl, aryl, arylalkyl, hetero- aryl or heteroarylalkyl; R2 is H, (C Cg) alkyl, alkenyl, alkynyl, cyclopropylalkyl, (C--Cg) haloalkyl, hydroxyalkyl, hydroxyalkyloxyalkyl or l-alkyl-2-imidazolidinone; X is NH, NRj, O, S, SO, SO2, wherein Rj is as defined above, which process comprises a) reacting a compound of the general formula (II)
Figure imgf000031_0002
with al) a compound of the general formula (III)
RιS02Cl (III) wherein R| is as defined above, to the formation of a compound of the general formula (I) above; or with
a2) N-phenyltrifluoromethanesulphonimide to the formation of a compound of formula (I) , wherein Rj is trifluoromethyl; or
b) reacting a compound of the general formula (IV)
Figure imgf000032_0001
wherein R[ and X are as defined above, with a compound of the general formula (V)
Figure imgf000032_0002
wherein R2 is as defined above, to the formation of a compound of the general formula (I) ; or
c) demethylating a compound of the general formula (VI)
Figure imgf000032_0003
wherein Rj and X are as defined above, using 1-chloro- ethyl chloroformate and then alkylating with a compound of formula (VIII) Y - R2
wherein R2 is as defined above and Y is leaving group to the formation of a compound of formula (I) , wherein R*, R and X are as defined above;
and if, desired, converting a compound of formula (I) into another compound of formula (I) , and/or, if desired, converting a compound of formula (I) into a pharmaceutically acceptable salt thereof and/or, if desired, converting a salt into a free compound, and/or if desired, resolving a mixture of isomers of compounds of formula (I) into the single iso ers.
6. A pharmaceutical composition containing a suitable carrier and/or diluent and, as an active principle, a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof.
7. A compound of formula (I) or a therapeutically acceptable salt thereof as defined in claim 1 for use in therapy.
8. Use of a compound of formula (I) or a therapeutically acceptable salt thereof as defined in claim 1, in the manufacture of medicament for use in treating psychotic/ schizophrenic disorders in one or several receptor systems of the central nervous system.
9. A method for treating psychotic/schizophrenic disorders in one or several receptor systems of the central nervous system in mammals, including man, which method comprises administering to the mammal of a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 1.
10. The method according to claim 9, wherein said compound of formula (I) or the pharmaceutically acceptable salt thereof is administered in an amount of from about 0,1 to about 2000 mg oral daily dose, or from 0,01 to about 100 mg parenteral daily dose.
PCT/SE1996/000344 1995-03-19 1996-03-19 NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS WO1996029316A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU51305/96A AU5130596A (en) 1995-03-19 1996-03-19 New sulfone ester analogues of iso-clozapine and related structures: atypical neuroleptics

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SE9500998A SE9500998D0 (en) 1995-03-19 1995-03-19 New sulfone ester analogues of iso-clozapine and related structures: atypical neuroleptics
SE9500998-1 1995-03-19

Publications (1)

Publication Number Publication Date
WO1996029316A1 true WO1996029316A1 (en) 1996-09-26

Family

ID=20397615

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE1996/000344 WO1996029316A1 (en) 1995-03-19 1996-03-19 NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS

Country Status (3)

Country Link
AU (1) AU5130596A (en)
SE (1) SE9500998D0 (en)
WO (1) WO1996029316A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998004289A2 (en) * 1996-07-26 1998-02-05 Smithkline Beecham Plc Pharmaceutical composition containing a 5ht2c antagonist and a d2 antagonist
WO2002042304A2 (en) * 2000-11-03 2002-05-30 Wyeth Cyclopenta[b][1,4] diazepino[6,7,1-hi]indoles as 5ht2c antagonists
US6759405B2 (en) 2000-11-03 2004-07-06 Wyeth Cycloocta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6777407B2 (en) 2000-11-03 2004-08-17 Wyeth Cyclopenta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6784172B2 (en) 2000-11-03 2004-08-31 Wyeth Processes for preparation of cyclopenta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
WO2004087124A1 (en) * 2003-03-29 2004-10-14 Mitsubishi Pharma Corporation Compounds having serotonin 5-ht17 receptor antagonist activity and muscarinic m4 receptor agonist activity and their use in the treatment of psychotic disorders
US6858604B2 (en) 2000-11-03 2005-02-22 Wyeth Cyclohepta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6916922B2 (en) 2000-11-03 2005-07-12 Wyeth Process for the preparation of 1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta [B] [1,4] diazepino- [6,7,1-hi] indole derivatives
WO2005063254A2 (en) * 2003-12-22 2005-07-14 Acadia Pharmaceuticals Inc. Amino substituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders
US7012089B2 (en) 2002-04-25 2006-03-14 Wyeth [1,4]Diazocino[7,8,1-hi]indole derivatives as antipsychotic and antiobesity agents
US7071185B2 (en) 2002-04-25 2006-07-04 Wyeth 1,2,3,4,7,8-hexahydro-6H-[1,4]diazepino[6,7,1-ij]quinoline derivatives as antipsychotic and antiobesity agents
US7129237B2 (en) 2002-04-25 2006-10-31 Wyeth [1,4]Diazepino[6,7,1-ij]quinoline derivatives as antipsychotic and antiobesity agents
US7141563B2 (en) 2000-11-03 2006-11-28 Wyeth Process for the preparation of 1, 2, 3, 4, 8, 9, 10, 10a-octahydro-7bH-cyclopenta[b] [1, 4]diazepino[6, 7, 1-hi] indole derivatives
WO2007004234A1 (en) * 2005-07-04 2007-01-11 Usv Limited A PROCESS FOR THE PREPARATION OF 2-[2-(4-DIBENZO[b,f] [L,4] THIAZEPIN-11-yl-1- PIPERAZINYL)ETHOXY] ETHANOL FUMARATE
WO2007053618A1 (en) * 2005-10-31 2007-05-10 Acadia Pharmaceuticals Inc. Prodrugs of muscarinic agonists and methods of treatment of neuropsychiatric disorders
CZ301061B6 (en) * 1999-07-09 2009-10-29 Astrazeneca Uk Limited Process for preparing dibenzothiazepine derivatives
US7671196B2 (en) 2005-07-26 2010-03-02 Wyeth Llc Diazepinoquinolines, synthesis thereof, and intermediates thereto
US7781427B2 (en) 2004-11-05 2010-08-24 Wyeth Llc Process for preparing quinoline compounds and products obtained therefrom
CN1913900B (en) * 2003-12-22 2010-11-24 阿卡蒂亚药品公司 Amino bustituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH422793A (en) * 1961-07-20 1966-10-31 Wander Ag Dr A Process for the preparation of 11-basic substituted dibenzo (b, f) (1,4) thiazepines
GB1216523A (en) * 1967-03-13 1970-12-23 Wander S A A Basically substituted dibenzoxazepines, dibenzothiazepines and dibenzodiazepines
WO1995000478A1 (en) * 1993-06-18 1995-01-05 Lundbeck & Co As H Aryltriflates and related compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH422793A (en) * 1961-07-20 1966-10-31 Wander Ag Dr A Process for the preparation of 11-basic substituted dibenzo (b, f) (1,4) thiazepines
GB1216523A (en) * 1967-03-13 1970-12-23 Wander S A A Basically substituted dibenzoxazepines, dibenzothiazepines and dibenzodiazepines
WO1995000478A1 (en) * 1993-06-18 1995-01-05 Lundbeck & Co As H Aryltriflates and related compounds

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Volume 93, No. 17, 27 October 1980, (Columbus, Ohio, USA), PROTIVA MIROSLAV et al., "Procataleptogenic 5H-dibenzol(b,e)-1,4-diazepine Derivative", page 685, Abstract No. 168316b; & CS,A,179 793, (15-07-79). *
COLLECTION CZECHOSLOV. CHEM. COMMUN., Volume 44, 1979, KAREL SINDELAR et al., "Potential Metabolites of the Neuroleptic Agents Noroxyclothepin and Oxyclothepin; Synthesis of 8-CHLORO-3-HYDROXY-10-(4-(2-HYDROXYETHYL)PI PERAZINO)- and -10-(4-(3-HYDROXYPROPYL)PIPERAZINO)-10, 11-DIHYDRODIBENZO.....", pages 3614-3626. *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998004289A3 (en) * 1996-07-26 1998-03-19 Smithkline Beecham Plc Pharmaceutical composition containing a 5ht2c antagonist and a d2 antagonist
WO1998004289A2 (en) * 1996-07-26 1998-02-05 Smithkline Beecham Plc Pharmaceutical composition containing a 5ht2c antagonist and a d2 antagonist
CZ301061B6 (en) * 1999-07-09 2009-10-29 Astrazeneca Uk Limited Process for preparing dibenzothiazepine derivatives
US6784172B2 (en) 2000-11-03 2004-08-31 Wyeth Processes for preparation of cyclopenta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6759405B2 (en) 2000-11-03 2004-07-06 Wyeth Cycloocta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6777407B2 (en) 2000-11-03 2004-08-17 Wyeth Cyclopenta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US7141563B2 (en) 2000-11-03 2006-11-28 Wyeth Process for the preparation of 1, 2, 3, 4, 8, 9, 10, 10a-octahydro-7bH-cyclopenta[b] [1, 4]diazepino[6, 7, 1-hi] indole derivatives
WO2002042304A3 (en) * 2000-11-03 2002-08-29 Wyeth Corp Cyclopenta[b][1,4] diazepino[6,7,1-hi]indoles as 5ht2c antagonists
US6858604B2 (en) 2000-11-03 2005-02-22 Wyeth Cyclohepta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US6916922B2 (en) 2000-11-03 2005-07-12 Wyeth Process for the preparation of 1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta [B] [1,4] diazepino- [6,7,1-hi] indole derivatives
US7351839B2 (en) 2000-11-03 2008-04-01 Wyeth Process for the preparation of 1,2,3,4,8,9,10, 10a-octahydro-7bH-cyclopenta[b][1,4]diazepino-[6,7,1-hi]indole derivatives
US7271162B2 (en) 2000-11-03 2007-09-18 Wyeth Cycloocta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US7271163B2 (en) 2000-11-03 2007-09-18 Wyeth Cyclopenta[b][1,4]diazepino[6,7,1-hi]indoles and derivatives
US7271164B2 (en) 2000-11-03 2007-09-18 Wyeth Cyclohepta[b][1,4]diazepino[6,7,1,-hi]indoles and derivatives
WO2002042304A2 (en) * 2000-11-03 2002-05-30 Wyeth Cyclopenta[b][1,4] diazepino[6,7,1-hi]indoles as 5ht2c antagonists
US7129237B2 (en) 2002-04-25 2006-10-31 Wyeth [1,4]Diazepino[6,7,1-ij]quinoline derivatives as antipsychotic and antiobesity agents
US7071185B2 (en) 2002-04-25 2006-07-04 Wyeth 1,2,3,4,7,8-hexahydro-6H-[1,4]diazepino[6,7,1-ij]quinoline derivatives as antipsychotic and antiobesity agents
US7012089B2 (en) 2002-04-25 2006-03-14 Wyeth [1,4]Diazocino[7,8,1-hi]indole derivatives as antipsychotic and antiobesity agents
US7687620B2 (en) 2002-04-25 2010-03-30 Wyeth Llc [1,4]diazepino[6,7,1-IJ]quinoline derivatives as antipsychotic and antiobesity agents
WO2004087124A1 (en) * 2003-03-29 2004-10-14 Mitsubishi Pharma Corporation Compounds having serotonin 5-ht17 receptor antagonist activity and muscarinic m4 receptor agonist activity and their use in the treatment of psychotic disorders
WO2005063254A3 (en) * 2003-12-22 2005-09-15 Acadia Pharm Inc Amino substituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders
WO2005063254A2 (en) * 2003-12-22 2005-07-14 Acadia Pharmaceuticals Inc. Amino substituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders
CN1913900B (en) * 2003-12-22 2010-11-24 阿卡蒂亚药品公司 Amino bustituted diaryl[a,d]cycloheptene analogs as muscarinic agonists and methods of treatment of neuropsychiatric disorders
KR101157881B1 (en) 2003-12-22 2012-07-06 아카디아 파마슈티칼스 인코포레이티드 AMINO SUBSTITUTED DIARYL[a,d]CYCLOHEPTENE ANALOGS AS MUSCARINIC AGONISTS AND METHODS OF TREATMENT OF NEUROPSYCHIATRIC DISORDERS
US7781427B2 (en) 2004-11-05 2010-08-24 Wyeth Llc Process for preparing quinoline compounds and products obtained therefrom
WO2007004234A1 (en) * 2005-07-04 2007-01-11 Usv Limited A PROCESS FOR THE PREPARATION OF 2-[2-(4-DIBENZO[b,f] [L,4] THIAZEPIN-11-yl-1- PIPERAZINYL)ETHOXY] ETHANOL FUMARATE
US7671196B2 (en) 2005-07-26 2010-03-02 Wyeth Llc Diazepinoquinolines, synthesis thereof, and intermediates thereto
WO2007053618A1 (en) * 2005-10-31 2007-05-10 Acadia Pharmaceuticals Inc. Prodrugs of muscarinic agonists and methods of treatment of neuropsychiatric disorders

Also Published As

Publication number Publication date
SE9500998D0 (en) 1995-03-19
AU5130596A (en) 1996-10-08

Similar Documents

Publication Publication Date Title
WO1996029316A1 (en) NEW SULFONE ESTER ANALOGUES OF iso-CLOZAPINE AND RELATED STRUCTURES: ATYPICAL NEUROLEPTICS
RU2129547C1 (en) Condensed heterocyclic compounds and squalene-synthetase inhibitors
KR900005697B1 (en) Process for preparing 8-chloro-1,5 - benzothiazepine derivatives
Lowe III et al. 5-Phenyl-3-ureidobenzazepin-2-ones as cholecystokinin-B receptor antagonists
JP2008533088A (en) N-hydroxyamide substituted at the ω position with a three-membered cyclic group as a histone deacetylase inhibitor, its preparation method, and use in a pharmaceutical composition
WO2008023847A1 (en) P2x4 receptor antagonist
MXPA02010892A (en) Azabicyclic carbamates and their use as alpha 7 nicotinic acetylcholine receptor agonists.
PL190960B1 (en) Substituted tetracyclic derivatives of tetrahydrofurane
US4585768A (en) 1,5-benzothiazepine derivatives and processes for preparing the same
WO2013026455A1 (en) Permanently positively charged antidepressants
EP0256888B1 (en) Benzothiazepine vasodilators having aralkyl substitution
PL116941B1 (en) Process for manufacturing novel derivatives of cyproheptadine
IL89035A (en) 1,4-Benzoxazine and 1,4-benzothiazine derivatives, their preparation and pharmaceutical compositions containing them
HU180999B (en) Process for producing new 11-substituted 5,11-dihydro-6h-pyrido-square bracket-2,3-b-square bracket closed-square bracket-1,4-square bracket closed-benzodiazepin-6-ones
AU639742B2 (en) New bicyclic amino-substituted compounds
Phillips et al. Binding of 5H-Dibenzo [b, e][1, 4] diazepine and Chiral 5H-Dibenzo [a, d] cycloheptene Analogs of Clozapine to Dopamine and Serotonin Receptors
TW589316B (en) Antidepressant azaheterocyclylmethyl derivatives of 1,4,5-trioxa-phenanthrene
US4729994A (en) Benzothiazepine vasodilators having aralkyl substitution
US4672064A (en) 1,5-benzoxathiepin derivatives, their production and use
CA1285561C (en) Naphthoxazines
AU778796B2 (en) Novel fluorinated imidazoline benzodioxane, preparation and therapeutic uses thereof
AU762021B2 (en) Pyrrolo(2,1-B)(1,3)benzothiazepines with atypical antipsychotic activity
AU2001258469A1 (en) New polycyclic indanylimidazoles with alpha2 adrenergic activity
WO2001085698A1 (en) New polycyclic indanylimidazoles with alpha2 adrenergic activity
JP2002537288A (en) Dopamine D1 receptor agonist compounds

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN

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

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: CA