WO2008026149A1 - 1,4,5,6, 7,8-hexahydro-i^1s-triaza-azulene derivatives as orexin receptor antagonists - Google Patents

1,4,5,6, 7,8-hexahydro-i^1s-triaza-azulene derivatives as orexin receptor antagonists Download PDF

Info

Publication number
WO2008026149A1
WO2008026149A1 PCT/IB2007/053417 IB2007053417W WO2008026149A1 WO 2008026149 A1 WO2008026149 A1 WO 2008026149A1 IB 2007053417 W IB2007053417 W IB 2007053417W WO 2008026149 A1 WO2008026149 A1 WO 2008026149A1
Authority
WO
WIPO (PCT)
Prior art keywords
disorders
methyl
phenyl
ethyl
pain
Prior art date
Application number
PCT/IB2007/053417
Other languages
French (fr)
Inventor
Hamed Aissaoui
Christoph Boss
Markus Gude
Ralf Koberstein
Thierry Sifferlen
Original Assignee
Actelion Pharmaceuticals Ltd
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 Actelion Pharmaceuticals Ltd filed Critical Actelion Pharmaceuticals Ltd
Priority to EP07826141A priority Critical patent/EP2059520B1/en
Priority to CA002659352A priority patent/CA2659352A1/en
Priority to DE602007004999T priority patent/DE602007004999D1/en
Priority to US12/439,303 priority patent/US20100029617A1/en
Priority to JP2009526234A priority patent/JP2010502587A/en
Priority to AT07826141T priority patent/ATE458740T1/en
Publication of WO2008026149A1 publication Critical patent/WO2008026149A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the present invention relates to novel compounds of formula (II) and their use as pharmaceuticals.
  • the invention also concerns related aspects including processes for the preparation of the compounds, pharmaceutical compositions containing one or more compounds of formula (II), and especially their use as orexin receptor antagonists.
  • Orexins are novel neuropeptides found in 1998 by two research groups, orexin A is a 33 amino acid peptide and orexin B is a 28 amino acid peptide (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexins are produced in discrete neurons of the lateral hypothalamus and bind to the G- protein-coupled receptors (OXi and OX 2 receptors).
  • the orexin-1 receptor (OXi) is selective for OX-A
  • the orexin-2 receptor (OX 2 ) is capable to bind OX-A as well as OX-B.
  • Orexins are found to stimulate food consumption in rats suggesting a physiological role for these peptides as mediators in the central feedback mechanism that regulates feeding behaviour (Sakurai T. et al., Cell, 1998, 92, 573-585). On the other hand, it was also observed that orexins regulate states of sleep and wakefulness opening potentially novel therapeutic approaches to narcolepsy as well as insomnia and other sleep disorders (Chemelli R.M. et al., Cell, 1999, 98, 437-451 ).
  • Orexin receptors are found in the mammalian brain and may have numerous implications in pathologies as known from the literature.
  • the present invention provides substituted 1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza- azulene derivatives, which are non-peptide antagonists of human orexin OXi and OX 2 receptors. These compounds are in particular of potential use in the treatment of e.g. eating disorders, drinking disorders, sleep disorders, or cognitive dysfunctions in psychiatric and neurologic disorders. Up to now, some low molecular weight compounds are known having a potential to antagonise either specifically OXi or OX 2 , or both receptors at the same time. In WO01/85693, Banyu Pharmaceuticals claimed N-acyltetrahydroisoquinoline derivatives. Other orexin receptor antagonists such as novel benzazepine derivatives are disclosed in WO02/051838.
  • a first aspect of the invention consists of a compound of the general formula II, wherein the chirality is as depicted below, and
  • Y represents -CH 2 -CH 2 -;
  • R 1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl, halogen and thfluoromethyl;
  • R 2 represents (Ci -4 )alkyl;
  • R 3 represents (Ci -4 )alkyl;
  • R 4 represents a phenyl group, wherein the phenyl group is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl and halogen; R 5 represents (Ci -4 )alkyl.
  • Also part of the invention are compounds of the formula (II) and pharmaceutically acceptable salts thereof.
  • halogen means fluorine, chlorine or bromine and preferably fluorine or chlorine. In a further preferred embodiment of the invention the term “halogen” means fluorine.
  • (Ci -4 )alkyl preferably means methyl, ethyl or butyl and more preferably ethyl.
  • the phenyl group as described in the present invention may be independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl, halogen, and trifluoromethyl.
  • substituents are independently selected from the group consisting of (Ci -4 )alkyl, halogen, and trifluoromethyl. Examples are phenyl, 3,5-difluoro-4-trifluoromethyl-phenyl, dimethyl-phenyl (e.g. 2,3- dimethyl-phenyl, 2,4-dimethyl-phenyl, 3,4-dimethyl-phenyl), 3,5-difluoro-4-methyl- phenyl, and 2-fluoro-4-trifluoromethyl-phenyl.
  • R 1 preferably represents a phenyl group which is independently mono-, di-, or trisubstituted (preferred di-, or trisubstituted) wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl, halogen, and trifluoromethyl.
  • the substituents are independently selected from the group consisting of methyl, ethyl, isopropyl, trifluoromethyl, fluorine and chlorine. More preferred, the substituents are independently selected from the group consisting of methyl, ethyl, trifluoromethyl, fluorine, and chlorine. Examples are 3,5-difluoro-4-trifluoromethyl-phenyl, dimethyl-phenyl (e.g.
  • R 4 represents a phenyl group which is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl, and halogen.
  • phenyl preferably means unsubstituted phenyl.
  • salts refers to non-toxic, inorganic or organic acid and/or base addition salts. Reference can be made to "Salt selection for basic drugs", Int. J. Pharm. (1986), 33, 201-217.
  • a further preferred embodiment of the invention are compounds of formula Il wherein
  • Y represents -CH 2 -CH 2 -;
  • R 1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from methyl, ethyl, fluorine, chlorine and thfluoromethyl;
  • R 2 represents (Ci -4 )alkyl;
  • R 3 represents (Ci -4 )alkyl;
  • R 4 represents a phenyl group, wherein the phenyl group is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci -4 )alkyl and halogen; R 5 represents (Ci -4 )alkyl.
  • Y represents -CH 2 -CH 2 -;
  • R 1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from the group consisting of methyl, ethyl, fluorine, chlorine and thfluoromethyl;
  • R 2 represents methyl;
  • R 3 represents ethyl;
  • R 4 represents a phenyl group;
  • R 5 represents methyl.
  • the compounds of Formula Il may contain one or more stereogenic or asymmetric centers, such as one or more asymmetric carbon atoms.
  • the compounds of Formula Il may thus be present as mixtures of stereoisomers or preferably as pure stereoisomers. Mixtures of stereoisomers may be separated in a manner known to a person skilled in the art.
  • the compounds specifically mentioned above contain two centers of chirality.
  • the compounds of the general formula Il exhibit the following chirality: (R)-2' and (S)-4 or (R)-2' and (R)-4.
  • the more preferred stereoisomers exhibit the chirality as depicted below:
  • the compounds of formula Il and their pharmaceutically acceptable salts can be used as medicaments, e.g. in the form of pharmaceutical compositions for enteral or parental administration.
  • the compounds of the general formula Il are useful for the treatment and/or prevention of the diseases mentioned herein.
  • the invention relates to a method for the treatment and/or prevention of the diseases mentioned herein, said method comprising administering to a subject a pharmaceutically active amount of a compound of general formula II.
  • the compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of dysthymic disorders including major depression and cyclothymia, affective neurosis, all types of manic depressive disorders, delirium, psychotic disorders, schizophrenia, catatonic schizophrenia, delusional paranoia, adjustment disorders and all clusters of personality disorders; schizoaffective disorders; anxiety disorders including generalized anxiety, obsessive compulsive disorder, posttraumatic stress disorder, panic attacks, all types of phobic anxiety and avoidance; separation anxiety; all psychoactive substance use, abuse, seeking and reinstatement; all types of psychological or physical addictions, dissociative disorders including multiple personality syndromes and psychogenic amnesias; sexual and reproductive dysfunction; psychosexual dysfunction and addiction; tolerance to
  • the compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of all types of sleep disorders, of stress- related syndromes, of psychoactive substance use and abuse, of cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders, of eating or drinking disorders.
  • Eating disorders may be defined as comprising metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
  • Pathologically modified food intake may result from disturbed appetite (attraction or aversion for food); altered energy balance (intake vs.
  • Drinking disorders include polydipsias in psychiatric disorders and all other types of excessive fluid intake.
  • Sleep disorders include all types of parasomnias, insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias; restless leg syndrome; sleep apneas; jet-lag syndrome; shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders.
  • Insomnias are defined as comprising sleep disorders associated with aging; intermittent treatment of chronic insomnia; situational transient insomnia (new environment, noise) or short-term insomnia due to stress; grief; pain or illness.
  • Insomnia also include stress-related syndromes including post-traumatic stress disorders as well as other types and subtypes of anxiety disorders such as generalized anxiety, obsessive compulsive disorder, panic attacks and all types of phobic anxiety and avoidance; psychoactive substance use, abuse, seeking and reinstatement are defined as all types of psychological or physical addictions and their related tolerance and dependence components.
  • Cognitive dysfunctions include deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
  • compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders.
  • diseases selected from the group consisting of sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders.
  • compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
  • diseases selected from the group consisting of cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
  • compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of eating disorders that comprise metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
  • compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of psychoactive substance use and abuse that comprise all types of psychological or physical addictions and their related tolerance and dependence components.
  • compositions can be effected in a manner which will be familiar to any person skilled in the art (see for example Mark Gibson, Editor, Pharmaceutical Preformulation and Formulation, IHS Health Group, Englewood, CO, USA, 2001 ; Remington, The Science and Practice of Pharmacy, 20th Edition, Philadelphia College of Pharmacy and Science; or Remington, The Science and Practice of Pharmacy, 21st Edition (2005), Part 5, "Pharmaceutical Manufacturing” [published by Lippincott Williams & Wilkins]) by bringing the described compounds of Formula (II) and their pharmaceutically acceptable salts, optionally in combination with other therapeutically valuable substances, into a galenical administration form together with suitable, non-toxic, inert, therapeutically compatible solid or liquid carrier materials and, if desired, usual pharmaceutical adjuvants.
  • R 1 , R 2 , R 3 , R 4 , R 5 and Y are as defined in general formula Il above, X represents OH or Cl and Z represents Br or OTs;
  • X represents OH or Cl and Z represents Br or OTs;
  • the preparation of an example for intermediates IH is described in WO05/1 18548.
  • the acylated Meldrum's acid derivative A reacted with the mono-substituted hydrazine B to give via a multi-step sequence (see Schemel and experimental part) the 3-(2,5-di-substituted-2/-/-pyrazol-3-yl)-propylamine C in a highly regioselective manner.
  • Primary amine C was then acylated with carboxylic acid derivative D to give the acyclic amide E which was transformed into the cyclic seven-membered imine F via Bischler-Napieralski reaction.
  • Subsequent imine reduction gave the secondary amine precursor G which was finally ⁇ /-alkylated with electrophile IH in order to obtain the final orexin receptor antagonists M.
  • R 1 , R 2 , R 3 , R 4 , R 5 and Y are as defined in general formula Il above, and Z represents Br or OTs;
  • Z represents Br or OTs;
  • the secondary amine G could be ⁇ /-alkylated with ester derivative J (instead of amide derivative IH) to give intermediate K which can either be directly transformed into the final compounds Il by reaction with an amine derivative L or which can be first hydrolyzed to the corresponding carboxylic acid M followed by reaction with an amine L to give final compounds M.
  • 3-(2-ethyl-5- methyl-2/-/-pyrazol-3-yl)-propylamine 10 can be acylated with the hydrocinnamic acid derivative 14 in presence of PyBOP giving the amide derivative 15.
  • the resulting solution was further stirred at 0°C, under nitrogen, for 1 h15. After concentration to dryness under reduced pressure, dichloromethane (50 ml), and water (50 ml) were successively added. The aqueous layer was further extracted with dichloromethane (25 ml). The mixed organic extracts were dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. The resulting crude was purified by flash chromatography (silicagel; DCM / MeOH / 25% aq.
  • reaction mixture was heated to 70°C, under nitrogen, for 23h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure.
  • reaction mixture was heated to 70°C, under nitrogen, for 23h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure.
  • reaction mixture was heated to 70°C, under nitrogen, for 24h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (50 ml) was added, and the solution was washed with brine (50 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure.
  • reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure.
  • the orexin receptor antagonistic activity of the compounds of formula (II) is determined in accordance with the following experimental method.
  • Chinese hamster ovary (CHO) cells expressing the human orexin-1 receptor and the human orexin-2 receptor, respectively, are grown in culture medium (Ham F- 12 with L-Glutamine) containing 300 ⁇ g/ml G418, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin and 10 % inactivated fetal calf serum (FCS).
  • FCS fetal calf serum
  • the cells are seeded at 80'0OO cells / well into 96-well black clear bottom sterile plates (Costar) which have been precoated with 1 % gelatine in Hanks' Balanced Salt Solution (HBSS). All reagents are from Gibco BRL.
  • the seeded plates are incubated overnight at 37°C in 5% CO 2 .
  • Human orexin-A as an agonist is prepared as 1 mM stock solution in methanol: water (1 :1 ), diluted in HBSS containing 0.1 % bovine serum albumin (BSA) and 2 mM HEPES for use in the assay at a final concentration of 10 nM.
  • BSA bovine serum albumin
  • Antagonists are prepared as 10 mM stock solution in DMSO, then diluted in 96- well plates, first in DMSO, then in HBSS containing 0.1 % bovine serum albumin (BSA) and 2 mM HEPES. On the day of the assay, 100 ⁇ l of loading medium (HBSS containing 1 % FCS, 2 mM HEPES, 5 mM probenecid (Sigma) and 3 ⁇ M of the fluorescent calcium indicator fluo-3 AM (1 mM stock solution in DMSO with 10% pluronic acid) (Molecular Probes) is added to each well.
  • BSA bovine serum albumin
  • the 96-well plates are incubated for 60 min at 37° C in 5% CO 2 .
  • the loading solution is then aspirated and cells are washed 3 times with 200 ⁇ l HBSS containing 2.5 mM probenecid, 0.1 % BSA, 2 mM HEPES. 100 ⁇ l of that same buffer is left in each well.
  • antagonists are added to the plate in a volume of 50 ⁇ l, incubated for 20 min and finally 100 ⁇ l of agonist is added. Fluorescence is measured for each well at 1 second intervals, and the height of each fluorescence peak is compared to the height of the fluorescence peak induced by 10 nM orexin-A with buffer in place of antagonist. For each antagonist, IC 50 value (the concentration of compound needed to inhibit 50 % of the agonistic response) is determined. Antagonistic activities of compounds are in the nanomolar range with respect to OX1 and OX 2 receptors. Selected compounds are displayed in Table 1.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Psychiatry (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Addiction (AREA)
  • Pain & Pain Management (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Psychology (AREA)
  • Reproductive Health (AREA)
  • Gynecology & Obstetrics (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Hospice & Palliative Care (AREA)
  • Pulmonology (AREA)
  • Anesthesiology (AREA)
  • Child & Adolescent Psychology (AREA)

Abstract

The invention relates to 1,4,5,6,7,8-hexahydro-1,2,5-triaza-azulene derivatives of formula (II), wherein the chirality is as depicted below, (II) and their use as orexin receptor antagonists.

Description

1,4,5,6,7,8-HEXAHYDRO-I^1S-TRIAZA-AZULENE DERIVATIVES AS OREXIN RECEPTOR ANTAGONISTS
The present invention relates to novel compounds of formula (II) and their use as pharmaceuticals. The invention also concerns related aspects including processes for the preparation of the compounds, pharmaceutical compositions containing one or more compounds of formula (II), and especially their use as orexin receptor antagonists.
Orexins (orexin A or OX-A and orexin B or OX-B) are novel neuropeptides found in 1998 by two research groups, orexin A is a 33 amino acid peptide and orexin B is a 28 amino acid peptide (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexins are produced in discrete neurons of the lateral hypothalamus and bind to the G- protein-coupled receptors (OXi and OX2 receptors). The orexin-1 receptor (OXi) is selective for OX-A, and the orexin-2 receptor (OX2) is capable to bind OX-A as well as OX-B. Orexins are found to stimulate food consumption in rats suggesting a physiological role for these peptides as mediators in the central feedback mechanism that regulates feeding behaviour (Sakurai T. et al., Cell, 1998, 92, 573-585). On the other hand, it was also observed that orexins regulate states of sleep and wakefulness opening potentially novel therapeutic approaches to narcolepsy as well as insomnia and other sleep disorders (Chemelli R.M. et al., Cell, 1999, 98, 437-451 ).
Orexin receptors are found in the mammalian brain and may have numerous implications in pathologies as known from the literature.
The present invention provides substituted 1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza- azulene derivatives, which are non-peptide antagonists of human orexin OXi and OX2 receptors. These compounds are in particular of potential use in the treatment of e.g. eating disorders, drinking disorders, sleep disorders, or cognitive dysfunctions in psychiatric and neurologic disorders. Up to now, some low molecular weight compounds are known having a potential to antagonise either specifically OXi or OX2, or both receptors at the same time. In WO01/85693, Banyu Pharmaceuticals claimed N-acyltetrahydroisoquinoline derivatives. Other orexin receptor antagonists such as novel benzazepine derivatives are disclosed in WO02/051838.
Furthermore, the use of solution-phase chemistry for the lead optimization of 1 ,2,3,4-tetrahydroisoquinoline derivatives as potential orexin receptor antagonists has been reported (Chimia, 2003, 57, 5, 270-275).
A first aspect of the invention consists of a compound of the general formula II, wherein the chirality is as depicted below, and
R-chirality
Formula Il
Figure imgf000003_0001
S-chirality wherein
Y represents -CH2-CH2-;
R1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl, halogen and thfluoromethyl; R2 represents (Ci-4)alkyl; R3 represents (Ci-4)alkyl;
R4 represents a phenyl group, wherein the phenyl group is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl and halogen; R5 represents (Ci-4)alkyl.
Also part of the invention are compounds of the formula (II) and pharmaceutically acceptable salts thereof.
In the present description the term "halogen" means fluorine, chlorine or bromine and preferably fluorine or chlorine. In a further preferred embodiment of the invention the term "halogen" means fluorine. The term "(Ci-4)alkyl", alone or in combination, means a straight-chain or branched-chain alkyl group with 1 to 4 carbon atoms. Examples of (Ci-4)alkyl groups are methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert- butyl. For the substituent R2 or R5 the term "(Ci-4)alkyl" preferably means methyl.
For the substituent R3, the term "(Ci-4)alkyl" preferably means methyl, ethyl or butyl and more preferably ethyl.
The phenyl group as described in the present invention may be independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl, halogen, and trifluoromethyl. Examples are phenyl, 3,5-difluoro-4-trifluoromethyl-phenyl, dimethyl-phenyl (e.g. 2,3- dimethyl-phenyl, 2,4-dimethyl-phenyl, 3,4-dimethyl-phenyl), 3,5-difluoro-4-methyl- phenyl, and 2-fluoro-4-trifluoromethyl-phenyl. R1 preferably represents a phenyl group which is independently mono-, di-, or trisubstituted (preferred di-, or trisubstituted) wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl, halogen, and trifluoromethyl. Preferred, the substituents are independently selected from the group consisting of methyl, ethyl, isopropyl, trifluoromethyl, fluorine and chlorine. More preferred, the substituents are independently selected from the group consisting of methyl, ethyl, trifluoromethyl, fluorine, and chlorine. Examples are 3,5-difluoro-4-trifluoromethyl-phenyl, dimethyl-phenyl (e.g. 2,3-dimethyl-phenyl, 2,4-dimethyl-phenyl, 3,4-dimethyl-phenyl), 3,5-difluoro-4-methyl-phenyl, and 2- fluoro-4-trifluoromethyl-phenyl. R4 represents a phenyl group which is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl, and halogen. For the substituent R4 the term "phenyl" preferably means unsubstituted phenyl.
The term "pharmaceutically acceptable salts" refers to non-toxic, inorganic or organic acid and/or base addition salts. Reference can be made to "Salt selection for basic drugs", Int. J. Pharm. (1986), 33, 201-217. A further preferred embodiment of the invention are compounds of formula Il wherein
Y represents -CH2-CH2-;
R1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from methyl, ethyl, fluorine, chlorine and thfluoromethyl; R2 represents (Ci-4)alkyl; R3 represents (Ci-4)alkyl;
R4 represents a phenyl group, wherein the phenyl group is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl and halogen; R5 represents (Ci-4)alkyl.
A further preferred embodiment of the invention are compounds of formula Il wherein
Y represents -CH2-CH2-;
R1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from the group consisting of methyl, ethyl, fluorine, chlorine and thfluoromethyl; R2 represents methyl; R3 represents ethyl; R4 represents a phenyl group; R5 represents methyl.
The compounds of Formula Il may contain one or more stereogenic or asymmetric centers, such as one or more asymmetric carbon atoms. The compounds of Formula Il may thus be present as mixtures of stereoisomers or preferably as pure stereoisomers. Mixtures of stereoisomers may be separated in a manner known to a person skilled in the art. The compounds specifically mentioned above contain two centers of chirality. In a preferred embodiment, the compounds of the general formula Il exhibit the following chirality: (R)-2' and (S)-4 or (R)-2' and (R)-4. The more preferred stereoisomers exhibit the chirality as depicted below:
R-chirality
Formula Il
Figure imgf000006_0001
S-chirality
Particularly preferred compounds are listed below:
(R)-2'-{(S)-4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl- 4,6,7,8-tetrahydro-1/-/-1 ,2,5-thaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide;
(R)-2'-{(S)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8- tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide;
(R)-2'-{1-ethyl-(S)-4-[2-(2-fluoro-4-thfluoromethyl-phenyl)-ethyl]-3-methyl- 4,6,7,8-tetrahydro-1/-/-1 ,2,5-thaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide;
(R)-2'-{(S)-4-[2-(3,5-difluoro-4-thfluoromethyl-phenyl)-ethyl]-1-ethyl-3- methyl-4,6,7,8-tetrahydro-1H-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl- acetamide;
The compounds of formula Il and their pharmaceutically acceptable salts can be used as medicaments, e.g. in the form of pharmaceutical compositions for enteral or parental administration.
The compounds of the general formula Il are useful for the treatment and/or prevention of the diseases mentioned herein.
In one embodiment, the invention relates to a method for the treatment and/or prevention of the diseases mentioned herein, said method comprising administering to a subject a pharmaceutically active amount of a compound of general formula II. The compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of dysthymic disorders including major depression and cyclothymia, affective neurosis, all types of manic depressive disorders, delirium, psychotic disorders, schizophrenia, catatonic schizophrenia, delusional paranoia, adjustment disorders and all clusters of personality disorders; schizoaffective disorders; anxiety disorders including generalized anxiety, obsessive compulsive disorder, posttraumatic stress disorder, panic attacks, all types of phobic anxiety and avoidance; separation anxiety; all psychoactive substance use, abuse, seeking and reinstatement; all types of psychological or physical addictions, dissociative disorders including multiple personality syndromes and psychogenic amnesias; sexual and reproductive dysfunction; psychosexual dysfunction and addiction; tolerance to narcotics or withdrawal from narcotics; increased anaesthetic risk, anaesthetic responsiveness; hypothalamic-adrenal dysfunctions; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders including neuropathic pain and restless leg syndrome; sleep apnea; narcolepsy; chronic fatigue syndrome; insomnias related to psychiatric disorders; all types of idiopathic insomnias and parasomnias; sleep- wake schedule disorders including jet-lag; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurological disorders; mental dysfunctions of aging; all types of amnesia; severe mental retardation; dyskinesias and muscular diseases; muscle spasticity, tremors, movement disorders; spontaneous and medication-induced dyskinesias; neurodegenerative disorders including Huntington's, Creutzfeld-Jacob's, Alzheimer's diseases and Tourette syndrome; Amyotrophic lateral sclerosis; Parkinson's disease; Cushing's syndrome; traumatic lesions; spinal cord trauma; head trauma; perinatal hypoxia; hearing loss; tinnitus; demyelinating diseases; spinal and cranial nerve diseases; ocular damage; retinopathy; epilepsy; seizure disorders; absence seizures, complex partial and generalized seizures; Lennox- Gastaut syndrome; migraine and headache; pain disorders; anaesthesia and analgesia; enhanced or exaggerated sensitivity to pain such as hyperalgesia, causalgia, and allodynia; acute pain; burn pain; atypical facial pain; neuropathic pain; back pain; complex regional pain syndrome I and II; arthritic pain; sports injury pain; dental pain; pain related to infection e.g. by HIV; post-chemotherapy pain; post-stroke pain; post-operative pain; neuralgia; osteoarthritis; conditions associated with visceral pain such as irritable bowel syndrome; eating disorders; diabetes; toxic and dysmetabolic disorders including cerebral anoxia, diabetic neuropathies and alcoholism; appetite, taste, eating, or drinking disorders; somatoform disorders including hypochondriasis; vomiting/nausea; emesis; gastric dyskinesia; gastric ulcers; Kallman's syndrome (anosmia); impaired glucose tolerance; intestinal motility dyskinesias; hypothalamic diseases; hypophysis diseases; hyperthermia syndromes, pyrexia, febrile seizures, idiopathic growth deficiency; dwarfism; gigantism; acromegaly; basophil adenoma; prolactinoma; hyperprolactinemia; brain tumors, adenomas; benign prostatic hypertrophy, prostate cancer; endometrial, breast, colon cancer; all types of testicular dysfunctions, fertility control; reproductive hormone abnormalities; hot flashes; hypothalamic hypogonadism, functional or psychogenic amenorrhea; urinary bladder incontinence; asthma; allergies; all types of dermatitis, acne and cysts, sebaceous gland dysfunctions; cardiovascular disorders; heart and lung diseases, acute and congestive heart failure; hypotension; hypertension; dyslipidemias, hyperlipidemias, insulin resistance; urinary retention; osteoporosis; angina pectoris; myocardial infarction; arrhythmias, coronary diseases, left ventricular hypertrophy; ischemic or haemorrhagic stroke; all types of cerebrovascular disorders including subarachnoid haemorrhage, ischemic and hemorrhagic stroke and vascular dementia; chronic renal failure and other renal diseases; gout; kidney cancer; urinary incontinence; and other diseases related to general orexin system dysfunctions. The compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of all types of sleep disorders, of stress- related syndromes, of psychoactive substance use and abuse, of cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders, of eating or drinking disorders. Eating disorders may be defined as comprising metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa. Pathologically modified food intake may result from disturbed appetite (attraction or aversion for food); altered energy balance (intake vs. expenditure); disturbed perception of food quality (high fat or carbohydrates, high palatability); disturbed food availability (unrestricted diet or deprivation) or disrupted water balance. Drinking disorders include polydipsias in psychiatric disorders and all other types of excessive fluid intake. Sleep disorders include all types of parasomnias, insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias; restless leg syndrome; sleep apneas; jet-lag syndrome; shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders. Insomnias are defined as comprising sleep disorders associated with aging; intermittent treatment of chronic insomnia; situational transient insomnia (new environment, noise) or short-term insomnia due to stress; grief; pain or illness. Insomnia also include stress-related syndromes including post-traumatic stress disorders as well as other types and subtypes of anxiety disorders such as generalized anxiety, obsessive compulsive disorder, panic attacks and all types of phobic anxiety and avoidance; psychoactive substance use, abuse, seeking and reinstatement are defined as all types of psychological or physical addictions and their related tolerance and dependence components. Cognitive dysfunctions include deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
In a further preferred embodiment of the invention compounds according to formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders. In another preferred embodiment of the invention compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
In another preferred embodiment of the invention compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of eating disorders that comprise metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
In another preferred embodiment of the invention compounds of general formula (II) may be used for the preparation of a medicament, and are suitable, for the prevention or treatment of diseases selected from the group consisting of psychoactive substance use and abuse that comprise all types of psychological or physical addictions and their related tolerance and dependence components.
The production of the pharmaceutical compositions can be effected in a manner which will be familiar to any person skilled in the art (see for example Mark Gibson, Editor, Pharmaceutical Preformulation and Formulation, IHS Health Group, Englewood, CO, USA, 2001 ; Remington, The Science and Practice of Pharmacy, 20th Edition, Philadelphia College of Pharmacy and Science; or Remington, The Science and Practice of Pharmacy, 21st Edition (2005), Part 5, "Pharmaceutical Manufacturing" [published by Lippincott Williams & Wilkins]) by bringing the described compounds of Formula (II) and their pharmaceutically acceptable salts, optionally in combination with other therapeutically valuable substances, into a galenical administration form together with suitable, non-toxic, inert, therapeutically compatible solid or liquid carrier materials and, if desired, usual pharmaceutical adjuvants.
General methods for the preparation of compounds of general formula II:
Steps H2 (described in Schem
Figure imgf000011_0001
Bischler-Napieralski rea ction
Figure imgf000011_0002
Figure imgf000011_0003
Figure imgf000011_0004
R1 , R2, R3, R4, R5 and Y are as defined in general formula Il above, X represents OH or Cl and Z represents Br or OTs; The preparation of an example for intermediates IH is described in WO05/1 18548.
The acylated Meldrum's acid derivative A reacted with the mono-substituted hydrazine B to give via a multi-step sequence (see Schemel and experimental part) the 3-(2,5-di-substituted-2/-/-pyrazol-3-yl)-propylamine C in a highly regioselective manner. Primary amine C was then acylated with carboxylic acid derivative D to give the acyclic amide E which was transformed into the cyclic seven-membered imine F via Bischler-Napieralski reaction. Subsequent imine reduction gave the secondary amine precursor G which was finally Λ/-alkylated with electrophile IH in order to obtain the final orexin receptor antagonists M.
Figure imgf000012_0001
M
R1, R2, R3, R4, R5 and Y are as defined in general formula Il above, and Z represents Br or OTs; For general methods see also WO 00/168609, WO 02/051838, and WO 04/085403
In a slightly different route the secondary amine G could be Λ/-alkylated with ester derivative J (instead of amide derivative IH) to give intermediate K which can either be directly transformed into the final compounds Il by reaction with an amine derivative L or which can be first hydrolyzed to the corresponding carboxylic acid M followed by reaction with an amine L to give final compounds M.
In general, all chemical transformations can be performed according to well- known standard methodologies as described in the literature or as described in the procedures below.
Preparation of compounds of general formula II:
Figure imgf000013_0001
P-TsOH MeOH
LiAIH4 / THF
Figure imgf000013_0003
Figure imgf000013_0002
1 ) 1 N BH3 THF / THF 1 2) CICO2Bn / K2CO3 / THF
Figure imgf000013_0004
10
Scheme 1: Synthesis ofpyrazole precursors.
The preparation of 3-(2,5-disubstituted-2/-/-pyrazol-3-yl)-propylamine derivatives (Scheme 1) started with the acylation of Meldrum's acid 2 with diketene 1 in presence of thethylamine giving intermediate 3. Reaction of a methanolic solution of 3 with an appropriately substituted hydrazine derivative (e.g. ethylhydrazine 4) in presence of triethylamine led regioselectively to the formation of the pyrazolo- derivative 5. Hydrolysis under acidic conditions (p-TsOH in methanol) resulted in the formation of methyl ester 6 which was quantitatively reduced with LiAIH4 to the corresponding alcohol derivative 7. Activation of the primary alcohol 7 as the corresponding mesylate and subsequent nucleophilic displacement mediated by tetrabutylammonium cyanide gave the expected nitrile derivative 8. Borane- mediated reduction of the nitrile 8 followed by protection of the resulting primary amine with a benzyloxycarbonyl moiety gave after flash chromatography the protected compound 9. Finally, deprotection of the amino function by hydrogenolysis using palladium over charcoal gave the expected primary amine 10.
In case neither the cinnamic acids nor the hydrocinnamic acids were commercially available, the synthetic routes depicted in Scheme 2 were followed for their preparation.
Method A for the preparation of hydrocinnamic acid derivatives from benzaldehyde derivatives via Knoevenagel condensation:
Figure imgf000015_0001
Tho following hydrocinnamic acids were prepared according to this sequence:
Figure imgf000015_0002
Method B for the preparation of hydrocinnamic acid derivatives from arylhalides via Heck reaction:
Figure imgf000015_0003
Q R
Hal = Br, or I
Tho following hydrocinnamic acids were prepared according to this sequence:
Figure imgf000015_0004
Scheme 2: Preparation of hydrocinnamic acid derivatives.
According to "method A", a Knoevenagel condensation between benzaldehyde derivatives N and malonic acid, in pyridine and in presence of piperidine gave, after acidic work-up, the cinnamic acid derivatives O. Catalytic hydrogenation under standard conditions (1 atm H2; 10% Pd-C; in methanol; rt) led to the corresponding hydrocinnamic acids P. Compounds 11 and 12 could be prepared according to this synthetic sequence. According to "method B", a Heck coupling between arylhalide Q and butyl acrylate using Pd(OAc)2/DABCO as a catalytic system gave butyl esters of the expected cinnamic acid derivatives R. Subsequent palladium-catalyzed hydrogenation of R followed by saponification of the butyl ester led to the target hydrocinnamic acid derivatives S. Compounds 13 and 14 could be prepared by following this synthetic route.
Scheme 3 summarizes the sequence applied for the preparation of final compounds (example in the case of Y = -CH2CH2-). For example 3-(2-ethyl-5- methyl-2/-/-pyrazol-3-yl)-propylamine 10 can be acylated with the hydrocinnamic acid derivative 14 in presence of PyBOP giving the amide derivative 15. Treatment of 15 under conditions of Bischler-Napieralski reaction (POCI3, in acetonitrile; reflux) led with good yield to the cyclic imine 16 which could be reduced with sodium borohydhde to the corresponding secondary amine 17. SN2- reaction with the tosylate 18 (described in WO 2005/118548) in 2-butanone at 70°C led to an equimolar mixture of diastereoisomers 19. Finally HPLC-mediated purification allowed the isolation of the pure diastereoisomers 20 [with (S; R) configuration] and 21 [with (R;R) configuration].
Figure imgf000017_0001
NaBH4 / MeOH
Figure imgf000017_0002
20 19 17
Figure imgf000017_0003
21
Scheme 3: Synthesis of final compounds (e.g. 20 and 21).
The following examples illustrate the invention but do not limit the scope thereof. All temperatures are stated in °C.
Abbreviations (as used herein)
AcOEt ethyl acetate aq. aqueous
BH3THF borane-tetrahydrofuran complex
Boc tert.-butyloxycarbonyl
CICO2Bn benzyl chloroformate
DABCO 1 ,4-diazabicyclo[2.2.2]octane
DCM dichloromethane
DIPEA N-ethyldiisopropylamine
DMF N,N-dimethylformamide
DMSO dimethylsulfoxide
EDC N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide
ELSD Evaporative Light-Scattering Detection
Et ethyl h hour(s)
HPLC High Performance Liquid Chromatography
HV High Vacuum
K2CO3 potassium carbonate
LiAIH4 lithium aluminum hydride
LC-MS Liquid Chromatography - Mass Spectroscopy
MeCN acetonitrile
MeOH methanol
MeSO2CI methanesulfonyl chloride min. minute(s)
MS Mass Spectroscopy n-Bu4NCN tetrabutylammonium cyanide
NaBH4 sodium borohydride NaOH sodium hydroxide
NEt3 triethylamine
PBS phosphate buffered saline
Pd(C) palladium over charcoal
Pd(OAc)2 palladium (II) acetate
POCI3 phosphorus oxychloride
P-TsOH para-toluenesulfonic acid
PyBOP benzotriazole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate rt room temperature sat. saturated
TEA triethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC Thin Layer Chromatography tR retention time
UV ultra violet
Vis visible
General Procedures and Examples:
HPLC conditions:
Analytic: Zorbax 59 SB Aqua column, 4.6 x 50 mm from Agilent Technologies. Eluents: A: acetonitrile; B: H2O + 0.04% TFA. Gradient: 90% B → 5% B over 2 min. Flow: 4.5 mL/min. Detection: UVA/is + MS.
Preparative: Waters Xterra RP18 (large), 75 x 30 mm. Eluent: A: Acetonitrile; B: H2O + 0.05% ammonium hydroxide (25% aq.). Gradient: 90% B → 10% B over 6.5 min. Flow: 75 mL/min. Detection: UV + ELSD. Preparation of precursors and intermediates:
5-(1-hydroxy-3-oxo-butylidene)-2,2-dimethyl-[1,3]dioxane-4,6-dione
Meldrum's acid (50.0 g; 346.9 mmol) was dissolved in DCM (300 ml) and NEt3 (48.3 ml; 346.9 mmol) was added. The resulting mixture was cooled to 0°C before dropwise addition of diketene (31.9 ml; 416.3 mmol). The reaction mixture was stirred for 2 h at r.t, cooled again to 0°C, and 1 M aqueous hydrochloric acid
(700 ml) was slowly added. The layers were separated, the organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure to give the product 5-(1-hydroxy-3-oxo-butylidene)-2,2- dimethyl-[1 ,3]dioxane-4,6-dione as an orange solid which was further dried under
HV (79.36 g; 100%). LC-MS: tR = 0.55 min.; [M+H]+: no ionisation.
5-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-2,2-dimethyl-[1,3]dioxane-4,6-dione 5-(1-Hydroxy-3-oxo-butylidene)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione (10.0 g; 43.8 mmol) was dissolved in anhydrous methanol (140 ml), and cooled to 0°C before dropwise addition within 15 min. of a solution of ethylhydrazine oxalate (7.24 g; 48.2 mmol) and NEt3 (13.4 ml; 96.4 mmol) in anhydrous methanol (60 ml). Upon completion of the addition, the mixture was slowly warmed to 60°C and stirring was continued for 90 min. The reaction mixture was concentrated to dryness under reduced pressure, and further dried under HV to give the product 5-(2- ethyl-5-methyl-2/-/-pyrazol-3-yl)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione as an orange oil (11.05 g; quantitative yield) which was used in the next step without further purification. LC-MS: tR = 0.52 min.; [M+H]+: no ionisation.
(2-ethyl-5-methyl-2H-pyrazol-3-yl)-acetic acid methyl ester
5-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione (11.05 g; 43.8 mmol) was dissolved in anhydrous methanol (125 ml) followed by the addition of p-TsOH x H2O (36.68 g; 192.85 mmol). The resulting reaction mixture was heated to 60°C for 75 minutes, cooled to 0°C, and thethylamine (26.85 ml; 192.9 mmol) was added dropwise over a period of 20 minutes. The resulting mixture was concentrated under reduced pressure. The residue was taken into DCM (200 ml), washed with water (2 x 75 ml), dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; DCM / MeOH = 30 / 1 ) gave the pure product (2-ethyl- 5-methyl-2/-/-pyrazol-3-yl)-acetic acid methyl ester as a yellow oil which was further dried under HV (5.56 g; 70%). LC-MS: tR = 0.62 min.; [M+H]+ = 183.18 g/mol.
2-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-ethanol To ice-cooled THF (250 ml) was added in one portion lithium aluminum hydride (2.207 g; 58.171 mmol). The resulting grey suspension was further stirred at 0°C, under nitrogen, and a solution of (2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-acetic acid methyl ester (10.0 g; 54.878 mmol) in anhydrous THF (80 ml) was added dropwise in 30 min. The resulting mixture was stirred at 0°C for 1 h. The ice- cooled reaction mixture was then treated sequentially with water (2.2 ml), 15% aqueous NaOH (2.2 ml), and water (6.6 ml). After additional stirring at rt (30 min.), the resulting precipitate was filtered and washed with diethylether. The resulting filtrate was dried over magnesium sulfate, filtered, concentrated to dryness under reduced pressure, and further dried under HV to give the product 2-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-ethanol as a yellow oil (8.45 g; 100%). LC- MS: tR = 0.36 min.; [M+H]+ = 155.16 g/mol.
3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propionitrile
A solution of methanesulfonyl chloride (1.86 ml; 23.950 mmol) in anhydrous THF (10 ml) was added dropwise to an ice-cooled solution of 2-(2-ethyl-5-methyl-2/-/- pyrazol-3-yl)-ethanol (3.358 g, 21.775 mmol) and triethylamine (4.1 ml; 29.390 mmol) in anhydrous THF (100 ml). The resulting mixture was allowed to stir at
0°C, under nitrogen, for 15 min. The reaction mixture was filtered and the solid was washed with anhydrous THF (60 ml). Tetrabutylammonium cyanide (14.611 g; 54.440 mmol) was then added in one portion at rt. The resulting orange solution was heated to 75°C for 1 h. The reaction mixture was allowed to cool to rt before water (30 ml), and diethylether (100 ml) were successively added. After separation, the aqueous layer was further extracted with ethyl acetate (3 x 100 ml). The mixed organic extracts were then dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Flash chromatography (silicagel; DCM / MeOH = 40 / 1 ) gave the pure product 3-(2- ethyl-5-methyl-2/-/-pyrazol-3-yl)-propionitrile as a yellow oil which was further dried under HV (3.07 g; 86%). LC-MS: tR = 0.57 min.; [M+H]+ = 164.39 g/mol.
[3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propyl]-carbamic acid benzyl ester To an ice-cooled solution of 3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propionitrile (3.070 g; 18.809 mmol) in anhydrous THF (100 ml) was added slowly 1 N BH3THF complex solution in THF (75 ml; 75 mmol) The resulting solution was then heated to 75°C for 1 h. After cooling to 0°C, aqueous 2N HCI (115 ml; 230 mmol) was carefully added. The resulting solution was stirred at 50°C for 1 h. After cooling to 0°C, solid K2CO3 (20.794 g; 150.469 mmol) was added portionwise, followed by dropwise addition of benzyl chloroformate (3.07 ml; 20.689 mmol). The resulting mixture was stirred at 0°C for 1 h, then at rt overnight. Diethylether (100 ml) and water (100 ml) were then added, and the aqueous layer was further extracted with diethylether (2 x 100 ml). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; ethyl acetate / heptane = 1 / 1 => ethyl acetate) gave the pure product [3-(2-ethyl-5- methyl-2/-/-pyrazol-3-yl)-propyl]-carbamic acid benzyl ester as a colorless oil which was further dried under HV (4.21 g; 74%). LC-MS: tR = 0.78 min.; [M+H]+ = 302.51 g/mol.
3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propylamine
A mixture of [3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propyl]-carbamic acid benzyl ester (1.400 g; 4.645 mmol) and 10% palladium on activated charcoal (280 mg) was placed under nitrogen before methanol (25 ml) was carefully added. The suspension was placed under vacuum, then under hydrogen (1 atm), and the reaction mixture was stirred at rt for 1 h. After filtration over celite, and additional washing of the celite with methanol, the resulting filtrate was concentrated to dryness under reduced pressure. After further drying under HV, the pure product 3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propylamine (0.774 g; 100%) as a yellow oil could be used for the next step without additional purification. LC-MS: tR = 0.21 min. (broad peak); [M+H]+ = 168.39 g/mol.
3-(3,4-dimethyl-phenyl)-acrylic acid
A suspension of 3,4-dimethylbenzaldehyde (15.000 g; 111.793 mmol) and malonic acid (22.103 g; 212.410 mmol) in pyridine (85 ml) was heated to 500C, under nitrogen. Then piperidine (8.5 ml; 86.079 mmol) was added dropwise (over 5 minutes) and the resulting suspension was heated to 75°C for 2h. The reaction mixture was cooled to 0°C, and poured into an ice-cooled solution of concentrated hydrochloric acid (12 N; 96 ml) in water (1200 ml). The precipitated colorless product was filtered off, and washed with water (3 x 100 ml). Remaining water was evaporated under reduced pressure, then under HV to give the dried product 3-(3,4-dimethyl-phenyl)-acrylic acid as a colorless solid (19.23 g; 98%). LC-MS: tR = 0.88 min; [M+H]+: no ionisation.
3-(3,4-dimethyl-phenyl)-propionic acid
A mixture of 3-(3,4-dimethyl-phenyl)-acrylic acid (19.269 g; 109.355 mmol) and 10% palladium over activated charcoal (1.920 g) was placed under nitrogen before methanol (300 ml) was carefully added. The resulting suspension was placed under vacuum, then under hydrogen (1 atm), and the reaction mixture was vigorously stirred at rt for 4h. The reaction mixture was filtered over a pad of celite, and concentrated under reduced pressure to give the expected product 3- (3,4-dimethyl-phenyl)-propionic acid as a grey solid which was further dried under HV (19.070 g; 98%). LC-MS: tR = 0.85 min; [M+H]+: no ionisation.
3-(2-fluoro-4-trifluoromethyl-phenyl)-acrylic acid A suspension of 2-fluoro-4-(thfluoromethyl)benzaldehyde (5.000 g; 26.027 mmol) and malonic acid (5.145 g; 49.451 mmol) in pyridine (20 ml) was heated to 50°C, under nitrogen. Then piperidine (2 ml; 20.040 mmol) was added dropwise (over 5 minutes), and the resulting suspension was heated to 75°C for 3h20. The reaction mixture was cooled to 0°C and poured into an ice-cooled solution of concentrated hydrochloric acid (12 N; 32 ml) in water (400 ml). The precipitated colorless product was filtered off, and washed with water (3 x 100 ml). Remaining water was evaporated under reduced pressure, then under HV to give the dried product 3-(2-fluoro-4-trifluoromethyl-phenyl)-acrylic acid as a colorless solid (5.030 g; 82.5%). LC-MS: tR = 0.89 min; [M+H]+: no ionisation.
3-(2-fluoro-4-trifluoromethyl-phenyl)-propionic acid
A mixture of 3-(2-fluoro-4-trifluoromethyl-phenyl)-acrylic acid (5.937 g; 25.356 mmol) and 10% palladium over activated charcoal (0.590 g) was placed under nitrogen before methanol (60 ml) was carefully added. The resulting suspension was placed under vacuum, then under hydrogen (1 atm), and the reaction mixture was vigorously stirred at rt for 4h. The reaction mixture was filtered over a pad of celite, and concentrated under reduced pressure to give the expected product 3-(2-fluoro-4-trifluoromethyl-phenyl)-propionic acid as a grey solid which was further dried under HV (4.590 g; 77%). LC-MS: tR = 0.88 min; [M+H]+: no ionisation.
5-bromo-1,3-difluoro-2-methyl-benzene
A solution of methanesulfonyl chloride (4.72 ml; 60.794 mmol) in anhydrous dichloromethane (10 ml) was added dropwise (over 5 min.) to an ice-cooled solution of 4-bromo-2,6-difluorobenzyl alcohol (11.300 g; 50.669 mmol) and triethylamine (14.1 ml; 101.338 mmol) in dichloromethane (200 ml). The resulting solution was stirred at 0°C, under nitrogen, for 30 min. The reaction mixture was diluted with ethyl acetate (200 ml), and water (100 ml) was added. The organic layer was successively washed with 1 N aqueous hydrochloric acid (100 ml), saturated aqueous NaHCU3 (100 ml), and finally with brine (100 ml). The organic layer was then dried over magnesium sulfate, filtered, and concentrated to dryness under reduced pressure to give an orange solid which was additionally dried under HV (15.170 g; 99.5%). LC-MS for the mesylate: tR = 0.92 min.; [M+H]+: no ionisation. To an ice-cooled solution of the obtained mesylate derivative (15.170 g; 50.381 mmol) in anhydrous THF (90 ml) was added dropwise a solution of superhydride LiEt3BH (1 N in THF; 106 ml; 106 mmol). The resulting mixture was stirred at 00C for 5 min., and then at rt for 30 min. The solution was cooled to 0°C before dropwise addition of water (100 ml), and addition of diethylether (200 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and carefully concentrated under reduced pressure (CAUTION ! product with low boiling point, therefore heating bath of the rotary evaporator at 30°C !). Purification by flash chromatography (CH2CI2) gave the pure product 5-bromo-1 ,3-difluoro-2-methyl- benzene as a colorless oil (6.910 g; 66%). LC-MS: tR = 1.00 min.; [M+H]+: no ionisation.
3-(3,5-difluoro-4-methyl-phenyl)-acrylic acid butyl ester
To a solution of 5-bromo-1 ,3-difluoro-2-methyl-benzene (6.910 g; 33.379 mmol) in anhydrous DMF (200 ml) were added successively butyl acrylate (7.15 ml; 50.062 mmol), DABCO (157 mg; 1.333 mmol), potassium carbonate (4.612 g; 33.379 mmol), and palladium acetate (150 mg; 0.669 mmol). The resulting brown suspension was stirred at 120°C for 1 h. The reaction mixture was allowed to cool to rt before diethylether (400 ml) was added. This mixture was then washed with water (2 x 200 ml), and the mixed aqueous layers were further extracted with diethylether (150 ml). The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purification by flash chromatography (silicagel; DCM / heptane = 1/1 => DCM) gave the pure product 3-(3,5-difluoro-4-methyl-phenyl)-acrylic acid butyl ester as a yellow oil which was further dried under HV (4.69 g; 55%). LC-MS: tR = 1.10 min.; [M+H]+: no ionisation. 3-(3,5-difluoro-4-methyl-phenyl)-propionic acid butyl ester
A mixture of 3-(3,5-difluoro-4-methyl-phenyl)-acrylic acid butyl ester (7.651 g;
30.089 mmol) and 10% palladium over activated charcoal (0.760 g) was placed under nitrogen before methanol (100 ml) was carefully added. The resulting suspension was placed under vacuum, then under hydrogen (1 atm), and the reaction mixture was vigorously stirred at rt for 2h45. The reaction mixture was filtered over a pad of celite, and concentrated under reduced pressure to give the expected product 3-(3,5-difluoro-4-methyl-phenyl)-propionic acid butyl ester as a yellow oil which was further dried under HV (6.960 g; 90%). LC-MS: tR = 1.10 min; [M+H]+: no ionisation.
3-(3,5-difluoro-4-methyl-phenyl)-propionic acid
To a solution of 3-(3,5-difluoro-4-methyl-phenyl)-propionic acid butyl ester (6.960 g; 27.157 mmol) in methanol (150 ml) and water (25 ml) was added at rt aqueous 1 N NaOH (68 ml; 68 mmol). The resulting solution was further stirred at rt for 1 h. Methanol was then removed under reduced pressure. Water (25 ml) was added, and the mixture was acidified with aqueous 1 N HCI (68 ml) in order to reach pH = 2. Dichloromethane (150 ml) was added, and the layers were shaken and separated. The aqueous layer was further extracted with dichloromethane (50 ml). The mixed organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The product 3- (3,5-difluoro-4-methyl-phenyl)-propionic acid was obtained as a pale yellow solid which was further dried under HV (5.090 g; 94%). LC-MS: tR = 0.86 min.; [M+H]+: no ionisation.
3-(3,5-difluoro-4-trifluoromethyl-phenyl)-acrylic acid butyl ester
To a solution of 3,5-difluoro-4-(trifluoromethyl)bromobenzene (10.000 g; 38.316 mmol) in anhydrous DMF (235 ml) were added successively butyl acrylate (8.2 ml; 57.474 mmol), DABCO (172 mg; 1.533 mmol), potassium carbonate (5.295 g; 38.316 mmol), and palladium(ll) acetate (172 mg; 0.766 mmol). The resulting brown suspension was stirred at 120°C for 2h30. The reaction mixture was allowed to cool to rt before diethylether (500 ml) was added. This mixture was then washed successively with water (1 x 400 ml; 1 x 200 ml) and with brine (50 ml), and the mixed aqueous layers were further extracted with diethylether (300 ml). The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. Purification by flash chromatography (silicagel; DCM / heptane = 1/1 ) gave the pure product 3- (3,5-difluoro-4-trifluoromethyl-phenyl)-acrylic acid butyl ester as a slightly beige solid which was further dried under HV (10.120 g; 86%). LC-MS: tR = 1.12 min.; [M+H]+: no ionisation.
3-(3,5-difluoro-4-trifluoromethyl-phenyl)-propionic acid butyl ester
A mixture of 3-(3,5-difluoro-4-trifluoromethyl-phenyl)-acrylic acid butyl ester (8.849 g; 28.710 mmol) and 10% palladium over activated charcoal (0.885 g) was placed under nitrogen before methanol (120 ml) was carefully added. The resulting suspension was placed under vacuum, then under hydrogen (1 atm), and the reaction mixture was vigorously stirred at rt for 3h30. The reaction mixture was filtered over a pad of celite, and concentrated under reduced pressure to give the expected product 3-(3,5-difluoro-4-trifluoromethyl-phenyl)- propionic acid butyl ester as a yellow oil which was further dried under HV (8.622 g; 97%). LC-MS: tR = 1.11 min; [M+H]+: no ionisation.
3-(3,5-difluoro-4-trifluoromethyl-phenyl)-propionic acid
To a solution of 3-(3,5-difluoro-4-trifluoromethyl-phenyl)-propionic acid butyl ester (7.658 g; 24.682 mmol) in methanol (168 ml) and water (32 ml) was added at rt aqueous 1 N NaOH (49.4 ml; 49.4 mmol). The resulting solution was further stirred at rt for 45 min. Methanol was then removed under reduced pressure. Water (25 ml) was added, and the mixture was acidified with aqueous 1 N HCI (50 ml) in order to reach pH = 2. Dichloromethane (150 ml) was added, and the layers were shaken and separated. The aqueous layer was further extracted with dichloromethane (50 ml). The mixed organic layers were dried over magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The product 3-(3,5-difluoro-4-trifluoromethyl-phenyl)-propionic acid was obtained as a colorless solid which was further dried under HV (6.216 g; 99%). LC-MS: tR = 0.90 min.; [M+H]+: no ionisation.
3-(3,5-difluoro-4-trifluoromethyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol- 3-yl)-propyl]-propionamide
To a solution of 3-(3,5-difluoro-4-thfluoromethyl-phenyl)-propionic acid (1.031 g; 4.056 mmol) in anhydrous DMF (17 ml) were added successively PyBOP (2.322 g; 4.462 mmol), a solution of 3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propylamine (0.678 g; 4.056 mmol) in anhydrous DMF (5 ml), and finally N- ethyldiisopropylamine (1.74 ml; 10.140 mmol). The resulting solution was further stirred at rt, under nitrogen, for 1 h. Ethyl acetate (100 ml) was added, and the resulting solution was washed with a 1/1 mixture of brine and water (3 x 70 ml). The resulting organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The crude orange oil (2.737 g) was purified by flash chromatography (silicagel; DCM / MeOH = 20 / 1 ) to give the pure product 3-(3,5-difluoro-4-trifluoromethyl-phenyl)-Λ/-[3-(2-ethyl-5- methyl-2/-/-pyrazol-3-yl)-propyl]-propionamide as a beige solid which was further dried under HV (1.384 g; 85%). LC-MS: tR = 0.84 min.; [M+H]+ = 404.53 g/mol.
3-(3,4-dimethyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propyl]- propionamide
To a solution of 3-(3,4-dimethyl-phenyl)-propionic acid (0.796 g; 4.470 mmol) in anhydrous DMF (15 ml) were added successively PyBOP (2.558 g; 4.917 mmol), a solution of 3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propylamine (0.747 g; 4.470 mmol) in anhydrous DMF (10 ml), and finally Λ/-ethyldiisopropylamine (1.91 ml; 11.175 mmol). The resulting solution was further stirred at rt, under nitrogen, for 1 h. Ethyl acetate (100 ml) was added, and the resulting solution was washed with a 1/1 mixture of brine and water (3 x 30 ml), and with water (30 ml). The resulting organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The crude was purified by flash chromatography (silicagel; DCM / MeOH = 15 / 1 ) to give the pure product 3-(3,4- dimethyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propyl]-propionamide as a beige solid which was further dried under HV (1.250 g; 85%). LC-MS: tR = 0.82 min.; [M+H]+ = 328.24 g/mol.
Λ/-[3-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-propyl]-3-(2-fluoro-4-trifluoromethyl- phenyl)-propionamide
To a solution of 3-(2-fluoro-4-thfluoromethyl-phenyl)-propionic acid (0.776 g;
3.286 mmol) in anhydrous DMF (15 ml) were added successively PyBOP (1.880 g; 3.614 mmol), a solution of 3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propylamine (0.549 g; 3.286 mmol) in anhydrous DMF (10 ml), and finally N- ethyldiisopropylamine (1.40 ml; 8.215 mmol). The resulting solution was further stirred at rt, under nitrogen, for 1 h. Ethyl acetate (100 ml) was added, and the resulting solution was washed with a 1/1 mixture of brine and water (3 x 30 ml). The resulting organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The crude was purified by flash chromatography (silicagel; DCM / MeOH = 15 / 1 ) to give the pure product Λ/-[3-(2-Ethyl-5-methyl-2/-/-pyrazol-3-yl)-propyl]-3-(2-fluoro-4-trifluoromethyl- phenyl)-propionamide as a beige solid which was further dried under HV (0.950 g; 75%). LC-MS: tR = 0.83 min.; [M+H]+ = 386.38 g/mol.
3-(3,5-difluoro-4-methyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)- propylj-propionamide
To a solution of 3-(3,5-difluoro-4-methyl-phenyl)-propionic acid (0.928 g; 4.640 mmol) in anhydrous DMF (15 ml) were added successively PyBOP (2.656 g; 5.104 mmol), a solution of 3-(2-ethyl-5-methyl-2/-/-pyrazol-3-yl)-propylamine (0.776 g; 4.640 mmol) in anhydrous DMF (10 ml), and finally N- ethyldiisopropylamine (2.0 ml; 11.600 mmol). The resulting solution was further stirred at rt, under nitrogen, for 1 h. Ethyl acetate (100 ml) was added, and the resulting solution was washed with a 1/1 mixture of brine and water (3 x 30 ml), and with water (30 ml). The resulting organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to dryness under reduced pressure. The crude was purified by flash chromatography (silicagel; DCM / MeOH = 15 / 1 ) to give the pure product 3-(3,5-difluoro-4-methyl-phenyl)-Λ/-[3-(2- 5 ethyl-5-methyl-2/-/-pyrazol-3-yl)-propyl]-propionamide as a colorless solid which was further dried under HV (1.320 g; 81 %). LC-MS: tR = 0.81 min.; [M+H]+ = 350.53 g/mol.
4-[2-(3,5-difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3-methyl- o 1 ,4,5,6,7, 8-hexahydro-1 ,2,5-triaza-azulene
A solution of 3-(3,5-difluoro-4-trifluoromethyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H- pyrazol-3-yl)-propyl]-propionamide (0.919 g; 2.278 mmol) and phosphorus oxychloride (1.05 ml; 11.391 mmol) in anhydrous acetonitrile (25 ml) was heated at reflux (90°C), under nitrogen, for 1 h45. The yellow solution was allowed to cool5 to room temperature, and was then concentrated to dryness under reduced pressure. The oily residue was dissolved in methanol (5 ml), concentrated again to dryness under reduced pressure, and this co-evaporation with methanol was repeated two additional times (2 x 5 ml MeOH). The resulting crude imine (LC- MS: t R = 0.81 min.; [M+H]+: 386.42 g/mol) was reduced to the corresponding0 amine without additional purification. A solution of this crude imine (0.882 g; 2.278 mmol) in anhydrous methanol (18 ml) was cooled to 0°C before portionwise addition of sodium borohydride (0.431 g; 11.390 mmol). The resulting yellow solution was further stirred at 0°C, under nitrogen, for 4h. After concentration to dryness under reduced pressure, dichloromethane (75 ml), and5 water (40 ml) were successively added. The aqueous layer was further extracted with dichloromethane (2 x 20 ml). The mixed organic extracts were dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. The resulting crude (yellow oil; 0.919 g) was purified by flash chromatography (silicagel; DCM / MeOH / 25% aq. NH4OH = 15 / 1 / 0.1 => DCM0 / MeOH / aq. NH4OH = 9 / 1 / 0.1 ) giving the pure product 4-[2-(3,5-difluoro-4- trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,5,6,7,8-hexahydro-1 ,2,5-thaza- azulene as a beige solid which was further dried under HV (0.473 g; 54%). LC- MS: tR = 0.79 min.; [M+1] = 388.46 g/mol.
4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1,4,5,6,7,8-hexahydro- 1,2,5-triaza-azulene
A solution of 3-(3,4-dimethyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)- propyl]-propionamide (1.250 g; 3.817 mmol) and phosphorus oxychloride (1.75 ml; 19.086 mmol) in anhydrous acetonitrile (40 ml) was heated at reflux (90°C), under nitrogen, for 1 h15. The resulting solution was allowed to cool to room temperature, and was then concentrated to dryness under reduced pressure. The oily residue was dissolved in methanol (5 ml), concentrated again to dryness under reduced pressure, and this co-evaporation with methanol was repeated two additional times (2 x 5 ml MeOH). The resulting crude imine (yellow oil; LC- MS: t R = 0.76 min.; [M+H]+: 310.47 g/mol) was reduced to the corresponding amine without additional purification. A solution of this crude imine (1.181 g; 3.817 mmol) in anhydrous methanol (45 ml) was cooled to 0°C before portionwise addition of sodium borohydride (0.752 g; 19.085 mmol). The resulting solution was further stirred at 0°C, under nitrogen, for 1 h15. After concentration to dryness under reduced pressure, dichloromethane (50 ml), and water (50 ml) were successively added. The aqueous layer was further extracted with dichloromethane (25 ml). The mixed organic extracts were dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. The resulting crude was purified by flash chromatography (silicagel; DCM / MeOH / 25% aq. NH4OH = 9 / 1 / 0.1 ) giving the pure product 4-[2-(3,4-dimethyl-phenyl)- ethyl]-1-ethyl-3-methyl-1 ,4,5,6,7,8-hexahydro-1 ,2,5-thaza-azulene as a yellow oil which was further dried under HV (0.980 g; 82%). LC-MS: tR = 0.76 min.; [M+1] = 312.51 g/mol.
1-ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 ,4,5,6,7,8- hexahydro-1 ,2,5-triaza-azulene A solution of Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-propyl]-3-(2-fluoro-4- trifluoromethyl-phenyl)-propionamide (1.020 g; 2.647 mmol) and phosphorus oxychloride (1.21 ml; 13.233 mmol) in anhydrous acetonitrile (40 ml) was heated at reflux (90°C), under nitrogen, for 1 h15. The resulting solution was allowed to cool to room temperature, and was then concentrated to dryness under reduced pressure. The oily residue was dissolved in methanol (5 ml), concentrated again to dryness under reduced pressure, and this co-evaporation with methanol was repeated two additional times (2 x 5 ml MeOH). The resulting crude imine (yellow solid; LC-MS: t R = 0.79 min.; [M+H]+: 368.40 g/mol) was reduced to the corresponding amine without additional purification. A solution of this crude imine (0.972 g; 2.647 mmol) in anhydrous methanol (25 ml) was cooled to 0°C before portionwise addition of sodium borohydride (0.521 g; 13.228 mmol). The resulting solution was further stirred at 0°C, under nitrogen, for 1 h15. After concentration to dryness under reduced pressure, dichloromethane (50 ml), and water (50 ml) were successively added. The aqueous layer was further extracted with dichloromethane (25 ml). The mixed milky organic extracts were directly concentrated under reduced pressure and the resulting crude was purified by flash chromatography (silicagel; DCM / MeOH / 25% aq. NH4OH = 9 / 1 / 0.1 ) giving the pure product 1-ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3- methyl-1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza-azulene as a yellow oil which was further dried under HV (0.453 g; 46%). LC-MS: tR = 0.77 min.; [M+1] = 370.50 g/mol.
4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-1,4,5,6,7,8- hexahydro-1 ,2,5-triaza-azulene
A solution of 3-(3,5-difluoro-4-methyl-phenyl)-Λ/-[3-(2-ethyl-5-methyl-2H-pyrazol- 3-yl)-propyl]-propionamide (1.320 g; 3.778 mmol) and phosphorus oxychloride (1.73 ml; 18.888 mmol) in anhydrous acetonitrile (40 ml) was heated at reflux (90°C), under nitrogen, for 1 h15. The resulting solution was allowed to cool to room temperature, and was then concentrated to dryness under reduced pressure. The oily residue was dissolved in methanol (5 ml), concentrated again to dryness under reduced pressure, and this co-evaporation with methanol was repeated two additional times (2 x 5 ml MeOH). The resulting crude imine (yellow oil; LC-MS: t R = 0.76 min.; [M+H]+: 332.46 g/mol) was reduced to the corresponding amine without additional purification. A solution of this crude imine (1.250 g; 3.772 mmol) in anhydrous methanol (45 ml) was cooled to 0°C before portionwise addition of sodium borohydride (0.743 g; 18.859 mmol). The resulting solution was further stirred at 0°C, under nitrogen, for 1 h15. After concentration to dryness under reduced pressure, dichloromethane (50 ml), and water (50 ml) were successively added. The aqueous layer was further extracted with dichloromethane (25 ml). The mixed milky organic extracts were directly concentrated under reduced pressure, and the resulting crude was purified by flash chromatography (silicagel; DCM / MeOH / 25% aq. NH4OH = 9 / 1 / 0.1 ) giving the pure product 4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3- methyl-1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza-azulene as a yellow solid which was further dried under HV (0.958 g; 76%). LC-MS: tR = 0.75 min.; [M+1] = 334.50 g/mol.
Preparation of final compounds:
Example 1 :
2'-{4-[2-(3,5-difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl -3-methyl- 4,6,7,8-tetrahydro-1H-1,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide
To a solution of 4-[2-(3,5-difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3- methyl-1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza-azulene (0.470 g; 1.213 mmol) in 2- butanone (6 ml) were added successively at rt (S)-toluene-4-sulfonic acid methylcarbamoyl-phenyl-methyl ester (0.426 g; 1.335 mmol), and N- ethyldiisopropylamine (0.41 ml; 2.426 mmol). The reaction mixture was heated to 70°C, under nitrogen, for 23h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; DCM / MeOH = 20 / 1 ) gave the mixture of the two epimers (R)-2'-{(S)-4-[2-(3,5-difluoro-4- trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-4,6,7,8-tetrahydro-1 /-/-1 ,2,5-triaza- azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide and (R)-2'-{(R)-4-[2-(3,5-difluoro-4- trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-4,6,7,8-tetrahydro-1 H-λ ,2,5-triaza- azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide (yellow solid; 0.450 g; 69%). LC-MS: tR = 0.87 min.; [M+H]+: 535.57 g/mol.
The optically pure isomers (R)-2'-{(S)-4-[2-(3,5-difluoro-4-trifluoromethyl-phenyl)- ethyl]-1 -ethyl-3-methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl- 2'-phenyl-acetamide (83 mg; colorless solid; LC-MS: tR = 0.87 min., [M+H]+ = 535.57 g/mol) and (R)-2'-{(R)-4-[2-(3,5-difluoro-4-thfluoromethyl-phenyl)-ethyl]-1- ethyl-3-methyl-4,6,7,8-tetrahydro-1 /-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'- phenyl-acetamide (75 mg; colorless solid; LC-MS: tR = 0.87 min., [M+H]+ = 535.56 g/mol) were then obtained after preparative HPLC-puhfication of this mixture of epimers.
Example 2:
2'-{4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8-tetrahydro-1H-
1,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide To a solution of 4-[2-(3,4-dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 , 4,5,6,7,8- hexahydro-1 ,2,5-triaza-azulene (0.980 g; 3.146 mmol) in 2-butanone (10 ml) were added successively at rt (S)-toluene-4-sulfonic acid methylcarbamoyl- phenyl-methyl ester (1.105 g; 3.461 mmol), and Λ/-ethyldiisopropylamine (1.07 ml; 6.293 mmol). The reaction mixture was heated to 70°C, under nitrogen, for 23h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; DCM / MeOH = 20 / 1 ) gave the mixture of the two epimers (R)-2'-{(S)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-4,6,7,8- tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide and (R)-2'- {(R)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide (yellow solid; 1.100 g; 76%). LC-MS: tR = 0.84 min.; [M+H]+: 459.62 g/mol.
The optically pure isomers (R)-2'-{(S)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3- methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl- acetamide (76 mg; colorless solid; LC-MS: tR = 0.84 min., [M+H]+ = 459.62 g/mol) and (R)-2'-{(R)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8- tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide (70 mg; colorless solid; LC-MS: tR = 0.84 min., [M+H]+ = 459.63 g/mol) were then obtained after preparative HPLC-purification of this mixture of epimers.
Example 3:
2'-{1 -ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl -4,6,7,8- tetrahydro-1H-1,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide To a solution of 1-ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl- 1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza-azulene (0.453 g; 1.226 mmol) in 2-butanone (5 ml) were added successively at rt (S)-toluene-4-sulfonic acid methylcarbamoyl-phenyl-methyl ester (0.430 g; 1.349 mmol), and N- ethyldiisopropylamine (0.42 ml; 2.453 mmol). The reaction mixture was heated to 70°C, under nitrogen, for 24h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (50 ml) was added, and the solution was washed with brine (50 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; DCM / MeOH = 20 / 1 ) gave the mixture of the two epimers (R)-2'-{1-ethyl-(S)-4-[2-(2-fluoro-4- trifluoromethyl-phenyl)-ethyl]-3-methyl-4,6,7,8-tetrahydro-1/-/-1 ,2,5-thaza-azulen- 5-yl}-Λ/-methyl-2'-phenyl-acetamide and (R)-2'-{1 -ethyl-(R)-4-[2-(2-fluoro-4- trifluoromethyl-phenyl)-ethyl]-3-methyl -4,6,7, 8-tetrahydro-1 /-/-1 ,2, 5-thaza-azulen- 5-yl}-Λ/-methyl-2'-phenyl-acetamide (yellow solid; 0.343 g; 54%). LC-MS: tR = 0.86 min.; [M+H]+: 517.62 g/mol. The optically pure isomers (R)-2'-{1-ethyl-(S)-4-[2-(2-fluoro-4-trifluoromethyl- phenyl)-ethyl]-3-methyl-4,6,7,8-tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl- 2'-phenyl-acetamide (82 mg; colorless solid; LC-MS: tR = 0.86 min., [M+H]+ =
517.62 g/mol) and (R)-2'-{1-ethyl-(R)-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)- ethyl]-3-methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'- phenyl-acetamide (90 mg; colorless solid; LC-MS: tR = 0.86 min., [M+H]+ =
517.63 g/mol) were then obtained after preparative HPLC-puhfication of this mixture of epimers.
Example 4:
2'-{4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8- tetrahydro-1H-1,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide
To a solution of 4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl- 1 ,4,5,6,7,8-hexahydro-1 ,2,5-triaza-azulene (0.958 g; 2.873 mmol) in 2-butanone (10 ml) were added successively at rt (S)-toluene-4-sulfonic acid methylcarbamoyl-phenyl-methyl ester (1.009 g; 3.161 mmol), and N- ethyldiisopropylamine (0.98 ml; 5.746 mmol). The reaction mixture was heated to 70°C, under nitrogen, for 23h. Then the reaction mixture was cooled to rt, the solvent was removed under vacuum, dichloromethane (80 ml) was added, and the solution was washed with brine (2 x 80 ml). The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. Purification by flash chromatography (silicagel; DCM / MeOH = 20 / 1 ) gave the mixture of two epimers (R)-2'-{(S)-4-[2-(3,5-difluoro-4-methyl-phenyl)- ethyl]-1 -ethyl-3-methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-thaza-azulen-5-yl}-Λ/-methyl- 2'-phenyl-acetamide and (R)-2'-{(R)-4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1- ethyl-3-methyl-4,6,7,8-tetrahydro-1 H-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'- phenyl-acetamide (yellow solid; 1.09 g; 79%). LC-MS: tR = 0.84 min.; [M+H]+: 481.62 g/mol. The optically pure isomers (R)-2'-{(S)-4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1- ethyl-3-methyl-4,6,7,8-tetrahydro-1 H- 1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'- phenyl-acetamide (83 mg; colorless solid; LC-MS: tR = 0.84 min., [M+H]+ = 481.62 g/mol) and (R)-2'-{(R)-4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3- methyl-4,6,7,8-tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl- acetamide (76 mg; colorless solid; LC-MS: tR = 0.85 min., [M+H]+ = 481.64 g/mol) were then obtained after preparative HPLC-purification of this mixture of epimers.
Biological assays
In vitro assay
The orexin receptor antagonistic activity of the compounds of formula (II) is determined in accordance with the following experimental method.
Experimental method:
• Intracellular calcium measurements:
Chinese hamster ovary (CHO) cells expressing the human orexin-1 receptor and the human orexin-2 receptor, respectively, are grown in culture medium (Ham F- 12 with L-Glutamine) containing 300 μg/ml G418, 100 U/ml penicillin, 100 μg/ml streptomycin and 10 % inactivated fetal calf serum (FCS). The cells are seeded at 80'0OO cells / well into 96-well black clear bottom sterile plates (Costar) which have been precoated with 1 % gelatine in Hanks' Balanced Salt Solution (HBSS). All reagents are from Gibco BRL. The seeded plates are incubated overnight at 37°C in 5% CO2.
Human orexin-A as an agonist is prepared as 1 mM stock solution in methanol: water (1 :1 ), diluted in HBSS containing 0.1 % bovine serum albumin (BSA) and 2 mM HEPES for use in the assay at a final concentration of 10 nM.
Antagonists are prepared as 10 mM stock solution in DMSO, then diluted in 96- well plates, first in DMSO, then in HBSS containing 0.1 % bovine serum albumin (BSA) and 2 mM HEPES. On the day of the assay, 100 μl of loading medium (HBSS containing 1 % FCS, 2 mM HEPES, 5 mM probenecid (Sigma) and 3 μM of the fluorescent calcium indicator fluo-3 AM (1 mM stock solution in DMSO with 10% pluronic acid) (Molecular Probes) is added to each well.
The 96-well plates are incubated for 60 min at 37° C in 5% CO2. The loading solution is then aspirated and cells are washed 3 times with 200 μl HBSS containing 2.5 mM probenecid, 0.1 % BSA, 2 mM HEPES. 100 μl of that same buffer is left in each well.
Within the Fluorescent Imaging Plate Reader (FLIPR, Molecular Devices), antagonists are added to the plate in a volume of 50 μl, incubated for 20 min and finally 100 μl of agonist is added. Fluorescence is measured for each well at 1 second intervals, and the height of each fluorescence peak is compared to the height of the fluorescence peak induced by 10 nM orexin-A with buffer in place of antagonist. For each antagonist, IC50 value (the concentration of compound needed to inhibit 50 % of the agonistic response) is determined. Antagonistic activities of compounds are in the nanomolar range with respect to OX1 and OX2 receptors. Selected compounds are displayed in Table 1.
Table 1 :
Figure imgf000038_0001

Claims

Claims
1. Compounds of formula (II), wherein the chirality is as depicted below, and
R-chirality
Formula Il
Figure imgf000039_0001
S-chirality wherein
Y represents -CH2-CH2-;
R1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl, halogen and thfluoromethyl; R2 represents (Ci-4)alkyl; R3 represents (Ci-4)alkyl;
R4 represents a phenyl group, wherein the phenyl group is unsubstituted or independently mono-, di-, or trisubstituted wherein the substituents are independently selected from the group consisting of (Ci-4)alkyl and halogen; R5 represents (Ci-4)alkyl; and pharmaceutically acceptable salts thereof.
2. Compounds according to claim 1 , wherein
R1 represents a phenyl group, wherein the phenyl group can be mono-, di-, or trisubstituted, wherein the substituents are independently selected from methyl, ethyl, fluorine, chlorine and trifluoromethyl; and pharmaceutically acceptable salts thereof.
3. Compounds according to claim 1 and 2, wherein R2 represents methyl; R3 represents ethyl; R4 represents a phenyl group;
R5 represents methyl; and pharmaceutically acceptable salts thereof.
4. Compounds according to anyone of claims 1 to 3 selected from the group consisting of:
(R)-2'-{(S)-4-[2-(3,5-difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8- tetrahydro-1/-/-1 ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide; (R)-2'-{(S)-4-[2-(3,4-dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-4,6,7,8-tetrahydro- 1 HA ,2,5-thaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide;
(R)-2'-{1-ethyl-(S)-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-4,6,7,8- tetrahydro-1 H-λ ,2,5-triaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide; and (R)-2'-{(S)-4-[2-(3,5-difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- 4,6,7,8-tetrahydro-1/-/-1 ,2,5-thaza-azulen-5-yl}-Λ/-methyl-2'-phenyl-acetamide; and pharmaceutically acceptable salts thereof.
5. Compounds according to anyone of claims 1 to 4 for use as medicaments.
6. Use of a compound according to any of claims 1 to 4 for the preparation of a medicament for the prevention or treatment of diseases selected from the group consisting of dysthymic disorders including major depression and cyclothymia, affective neurosis, all types of manic depressive disorders, delirium, psychotic disorders, schizophrenia, catatonic schizophrenia, delusional paranoia, adjustment disorders and all clusters of personality disorders; schizoaffective disorders; anxiety disorders including generalized anxiety, obsessive compulsive disorder, posttraumatic stress disorder, panic attacks, all types of phobic anxiety and avoidance; separation anxiety; all psychoactive substance use, abuse, seeking and reinstatement; all types of psychological or physical addictions, dissociative disorders including multiple personality syndromes and psychogenic amnesias; sexual and reproductive dysfunction; psychosexual dysfunction and addiction; tolerance to narcotics or withdrawal from narcotics; increased anaesthetic risk, anaesthetic responsiveness; hypothalamic-adrenal dysfunctions; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders including neuropathic pain and restless leg syndrome; sleep apnea; narcolepsy; chronic fatigue syndrome; insomnias related to psychiatric disorders; all types of idiopathic insomnias and parasomnias; sleep-wake schedule disorders including jet-lag; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurological disorders; mental dysfunctions of aging; all types of amnesia; severe mental retardation; dyskinesias and muscular diseases; muscle spasticity, tremors, movement disorders; spontaneous and medication-induced dyskinesias; neurodegenerative disorders including Huntington's, Creutzfeld-Jacob's, Alzheimer's diseases and Tourette syndrome; Amyotrophic lateral sclerosis; Parkinson's disease; Cushing's syndrome; traumatic lesions; spinal cord trauma; head trauma; perinatal hypoxia; hearing loss; tinnitus; demyelinating diseases; spinal and cranial nerve diseases; ocular damage; retinopathy; epilepsy; seizure disorders; absence seizures, complex partial and generalized seizures; Lennox- Gastaut syndrome; migraine and headache; pain disorders; anaesthesia and analgesia; enhanced or exaggerated sensitivity to pain such as hyperalgesia, causalgia, and allodynia; acute pain; burn pain; atypical facial pain; neuropathic pain; back pain; complex regional pain syndrome I and II; arthritic pain; sports injury pain; dental pain; pain related to infection e.g. by HIV; post-chemotherapy pain; post-stroke pain; post-operative pain; neuralgia; osteoarthritis; conditions associated with visceral pain such as irritable bowel syndrome; eating disorders; diabetes; toxic and dysmetabolic disorders including cerebral anoxia, diabetic neuropathies and alcoholism; appetite, taste, eating, or drinking disorders; somatoform disorders including hypochondriasis; vomiting/nausea; emesis; gastric dyskinesia; gastric ulcers; Kallman's syndrome (anosmia); impaired glucose tolerance; intestinal motility dyskinesias; hypothalamic diseases; hypophysis diseases; hyperthermia syndromes, pyrexia, febrile seizures, idiopathic growth deficiency; dwarfism; gigantism; acromegaly; basophil adenoma; prolactinoma; hyperprolactinemia; brain tumors, adenomas; benign prostatic hypertrophy, prostate cancer; endometrial, breast, colon cancer; all types of testicular dysfunctions, fertility control; reproductive hormone abnormalities; hot flashes; hypothalamic hypogonadism, functional or psychogenic amenorrhea; urinary bladder incontinence; asthma; allergies; all types of dermatitis, acne and cysts, sebaceous gland dysfunctions; cardiovascular disorders; heart and lung diseases, acute and congestive heart failure; hypotension; hypertension; dyslipidemias, hyperlipidemias, insulin resistance; urinary retention; osteoporosis; angina pectoris; myocardial infarction; arrhythmias, coronary diseases, left ventricular hypertrophy; ischemic or haemorrhagic stroke; all types of cerebrovascular disorders including subarachnoid haemorrhage, ischemic and hemorrhagic stroke and vascular dementia; chronic renal failure and other renal diseases; gout; kidney cancer; urinary incontinence; and other diseases related to general orexin system dysfunctions.
7. Use of a compound according to any of claims 1 to 4 for the preparation of a medicament for the prevention or treatment of diseases selected from the group consisting of all types of sleep disorders, of stress-related syndromes, of psychoactive substance use and abuse, of cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders, of eating or drinking disorders. Eating disorders may be defined as comprising metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa. Pathologically modified food intake may result from disturbed appetite (attraction or aversion for food); altered energy balance (intake vs. expenditure); disturbed perception of food quality (high fat or carbohydrates, high palatability); disturbed food availability (unrestricted diet or deprivation) or disrupted water balance. Drinking disorders include polydipsias in psychiatric disorders and all other types of excessive fluid intake. Sleep disorders include all types of parasomnias, insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias; restless leg syndrome; sleep apneas; jet-lag syndrome; shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders. Insomnias are defined as comprising sleep disorders associated with aging; intermittent treatment of chronic insomnia; situational transient insomnia (new environment, noise) or short-term insomnia due to stress; grief; pain or illness. Insomnia also include stress-related syndromes including post-traumatic stress disorders as well as other types and subtypes of anxiety disorders such as generalized anxiety, obsessive compulsive disorder, panic attacks and all types of phobic anxiety and avoidance; psychoactive substance use, abuse, seeking and reinstatement are defined as all types of psychological or physical addictions and their related tolerance and dependence components. Cognitive dysfunctions include deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
PCT/IB2007/053417 2006-08-28 2007-08-27 1,4,5,6, 7,8-hexahydro-i^1s-triaza-azulene derivatives as orexin receptor antagonists WO2008026149A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP07826141A EP2059520B1 (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-hexahydro-i,2,5-triaza-azulene derivatives as orexin receptor antagonists
CA002659352A CA2659352A1 (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-hexahydro-1,2,5-triaza-azulene derivatives as orexin receptor antagonists
DE602007004999T DE602007004999D1 (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-HEXAHYDRO-1,2,5-TRIAZA AZULEN DERIVATIVES AS OREXIN RECEPTOR ANTAGONISTS
US12/439,303 US20100029617A1 (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-hexahydro-1,2,5-triaza-azulene derivatives as orexin receptor antagonists
JP2009526234A JP2010502587A (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-Hexahydro-1,2,5-triaza-azulene derivatives as orexin receptor antagonists
AT07826141T ATE458740T1 (en) 2006-08-28 2007-08-27 1,4,5,6,7,8-HEXAHYDRO-1,2,5-TRIAZA-AZULENE DERIVATIVES AS OREXIN RECEPTOR ANTAGONISTS

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IB2006052984 2006-08-28
IBPCT/IB2006/052984 2006-08-28

Publications (1)

Publication Number Publication Date
WO2008026149A1 true WO2008026149A1 (en) 2008-03-06

Family

ID=38951714

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2007/053417 WO2008026149A1 (en) 2006-08-28 2007-08-27 1,4,5,6, 7,8-hexahydro-i^1s-triaza-azulene derivatives as orexin receptor antagonists

Country Status (9)

Country Link
US (1) US20100029617A1 (en)
EP (1) EP2059520B1 (en)
JP (1) JP2010502587A (en)
CN (1) CN101511833A (en)
AT (1) ATE458740T1 (en)
CA (1) CA2659352A1 (en)
DE (1) DE602007004999D1 (en)
ES (1) ES2339822T3 (en)
WO (1) WO2008026149A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008147518A1 (en) 2007-05-23 2008-12-04 Merck & Co., Inc. Pyridyl piperidine orexin receptor antagonists
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
US9156819B2 (en) 2011-10-19 2015-10-13 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
US9440982B2 (en) 2012-02-07 2016-09-13 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
US9499517B2 (en) 2012-02-07 2016-11-22 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists
WO2017194548A1 (en) 2016-05-10 2017-11-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of autoimmune inflammatory diseases
US10221170B2 (en) 2014-08-13 2019-03-05 Eolas Therapeutics, Inc. Difluoropyrrolidines as orexin receptor modulators
US10828302B2 (en) 2016-03-10 2020-11-10 Janssen Pharmaceutica Nv Methods of treating depression using orexin-2 receptor antagonists
US10894789B2 (en) 2016-02-12 2021-01-19 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US11059828B2 (en) 2009-10-23 2021-07-13 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-C]pyrroles as orexin receptor modulators

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010513466A (en) * 2006-12-22 2010-04-30 アクテリオン ファーマシューティカルズ リミテッド 5,6,7,8-tetrahydro-imidazo [1,5-A] pyrazine derivatives
CN102076694A (en) * 2008-06-25 2011-05-25 埃科特莱茵药品有限公司 5, 6, 7, 8-tetrahydro-imidazo[1, 5-a]pyrazine compounds
KR20230043227A (en) * 2016-05-12 2023-03-30 에자이 알앤드디 매니지먼트 가부시키가이샤 Methods of treating circadian rhythm sleep disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002051838A1 (en) * 2000-12-27 2002-07-04 Actelion Pharmaceuticals Ltd. Novel benzazepines and related heterocyclic derivatives which are useful as orexin receptor antagonists
WO2004004733A1 (en) * 2002-07-09 2004-01-15 Actelion Pharmaceuticals Ltd. 7,8,9,10-tetrahydro-6h-azepino, 6,7,8,9-tetrahydro-pyrido and 2,3-dihydro-2h-pyrrolo[2,1-b]-quinazolinone derivatives

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA02008797A (en) * 2000-03-14 2005-09-08 Actelion Pharmaceuticals Ltd 1,2,3,4 tetrahydroisoquinoline derivatives.
WO2001085693A1 (en) * 2000-05-11 2001-11-15 Banyu Pharmaceutical Co., Ltd. N-acyltetrahydroisoquinoline derivatives
EP1637521B1 (en) * 2003-06-23 2013-06-19 Ono Pharmaceutical Co., Ltd. Novel tricyclic heterocycle compound
DK1668014T3 (en) * 2003-09-17 2009-04-20 Janssen Pharmaceutica Nv Condensed heterocyclic compounds as serotonin receptor modulators
WO2007122591A2 (en) * 2006-04-26 2007-11-01 Actelion Pharmaceuticals Ltd Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002051838A1 (en) * 2000-12-27 2002-07-04 Actelion Pharmaceuticals Ltd. Novel benzazepines and related heterocyclic derivatives which are useful as orexin receptor antagonists
WO2004004733A1 (en) * 2002-07-09 2004-01-15 Actelion Pharmaceuticals Ltd. 7,8,9,10-tetrahydro-6h-azepino, 6,7,8,9-tetrahydro-pyrido and 2,3-dihydro-2h-pyrrolo[2,1-b]-quinazolinone derivatives

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8242121B2 (en) 2007-05-23 2012-08-14 Merck Sharp & Dohme Corp. Pyridyl piperidine orexin receptor antagonists
US8569311B2 (en) 2007-05-23 2013-10-29 Merch Sharp & Dohme Corp. Pyridyl piperidine orexin receptor antagonists
WO2008147518A1 (en) 2007-05-23 2008-12-04 Merck & Co., Inc. Pyridyl piperidine orexin receptor antagonists
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
US11059828B2 (en) 2009-10-23 2021-07-13 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-C]pyrroles as orexin receptor modulators
US11667644B2 (en) 2009-10-23 2023-06-06 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-c]pyrroles as orexin receptor modulators
US9156819B2 (en) 2011-10-19 2015-10-13 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
US9896452B2 (en) 2012-02-07 2018-02-20 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
US9499517B2 (en) 2012-02-07 2016-11-22 Eolas Therapeutics, Inc. Substituted prolines / piperidines as orexin receptor antagonists
US9440982B2 (en) 2012-02-07 2016-09-13 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
US10221170B2 (en) 2014-08-13 2019-03-05 Eolas Therapeutics, Inc. Difluoropyrrolidines as orexin receptor modulators
US10894789B2 (en) 2016-02-12 2021-01-19 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US11434236B2 (en) 2016-02-12 2022-09-06 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US10828302B2 (en) 2016-03-10 2020-11-10 Janssen Pharmaceutica Nv Methods of treating depression using orexin-2 receptor antagonists
US11241432B2 (en) 2016-03-10 2022-02-08 Janssen Pharmaceutica Nv Methods of treating depression using orexin-2 receptor antagonists
WO2017194548A1 (en) 2016-05-10 2017-11-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of autoimmune inflammatory diseases

Also Published As

Publication number Publication date
JP2010502587A (en) 2010-01-28
CA2659352A1 (en) 2008-03-06
CN101511833A (en) 2009-08-19
ATE458740T1 (en) 2010-03-15
ES2339822T3 (en) 2010-05-25
US20100029617A1 (en) 2010-02-04
EP2059520A1 (en) 2009-05-20
EP2059520B1 (en) 2010-02-24
DE602007004999D1 (en) 2010-04-08

Similar Documents

Publication Publication Date Title
EP2059520B1 (en) 1,4,5,6,7,8-hexahydro-i,2,5-triaza-azulene derivatives as orexin receptor antagonists
EP2013209B1 (en) Pyrazolo-tetrahydropyridine derivatives as orexin receptor antagonists
EP2125823B1 (en) 5,6,7,8-tetrahydro-imidazo[1,5-a]pyrazine derivatives
RU2478099C2 (en) 2-aza-bicyclo[3,3,0]octane derivatives
US8742106B2 (en) Disubstituted heteroaryl-fused pyridines
US20110086889A1 (en) Tetrazole compounds as orexin receptor antagonists
US20110039857A1 (en) Piperidine and pyroolidine compounds
CA2699328A1 (en) Pyrrolidines and piperidines as orexin receptor antagonists
CA2669060A1 (en) Piperidine compounds
US10738031B2 (en) N-[(heteroaryloxy)propanyl]heteroaryl carboxamides as antagonists of orexin subtype 1 receptor activity
US20110105514A1 (en) 5,6,7,8-tetrahydro-imidazo[1,5-a]pyrazine compounds
WO2017178343A1 (en) Novel n-[(pyrimidinylamino)propanyl]-and n-[(pyridinylamino)propanyl]arylcarboxamides
WO2017178340A1 (en) Novel n-[(pyridyloxy)propanyl]benzamides
EP3442967A1 (en) Novel n-[(pyrimidinyloxy)propanyl]benzamides
WO2017178339A1 (en) Novel n-[(pyrazinyloxy)propanyl]benzamides
EP3442966A1 (en) Noveln

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780032131.6

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07826141

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2007826141

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2659352

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009526234

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 12439303

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU