WO2007122591A2 - Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists - Google Patents

Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists Download PDF

Info

Publication number
WO2007122591A2
WO2007122591A2 PCT/IB2007/051522 IB2007051522W WO2007122591A2 WO 2007122591 A2 WO2007122591 A2 WO 2007122591A2 IB 2007051522 W IB2007051522 W IB 2007051522W WO 2007122591 A2 WO2007122591 A2 WO 2007122591A2
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
phenyl
ethyl
pyrazolo
tetrahydro
Prior art date
Application number
PCT/IB2007/051522
Other languages
French (fr)
Other versions
WO2007122591A3 (en
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 AT07735643T priority Critical patent/ATE496051T1/en
Priority to CA002647077A priority patent/CA2647077A1/en
Priority to DE602007012072T priority patent/DE602007012072D1/en
Priority to JP2009507230A priority patent/JP2009535324A/en
Priority to EP07735643A priority patent/EP2013209B1/en
Priority to US12/298,369 priority patent/US7834028B2/en
Publication of WO2007122591A2 publication Critical patent/WO2007122591A2/en
Publication of WO2007122591A3 publication Critical patent/WO2007122591A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • 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
    • A61P25/20Hypnotics; 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel compounds of formula (I) and/or (Ia) 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 (I) and/or (Ia), 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 such as dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apnea; narcolepsy; idiopathic insomnias; parasomnias; benign prostatic hypertrophy; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders; and other diseases related to general orexin system dysfunctions.
  • pathologies such as dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apne
  • the present invention provides substituted 1 ,2,3,4-tetrahydro-pyrrolo[1 ,2- a]pyrazine and 5,6,7,8-tetrahydro-imidazo[1 ,5-a]pyrazine derivatives, which are non-peptide antagonists of human orexin receptors, in particular 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.
  • a first aspect of the invention consists of a compound of the general formula I
  • R 1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of halogen and trifluoromethyl;
  • 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.
  • the compounds of the general formula I and/or Ia contain one or more asymmetric carbon atoms and may be prepared in form of optically pure enantiomers, mixtures of enantiomers such as racemates, diastereomers, mixtures of diastereomers, diastereomeric racemates, mixtures of diastereomeric racemates, or meso-forms.
  • halogen means fluorine, chlorine, bromine or iodine and preferably fluorine or chlorine. In a further preferred embodiment of the invention the term “halogen” means fluorine.
  • (Ci -4 )alkyl 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; in analogy the term means a straight-chain or branched-chain alkyl group with 2 to 4 carbon atoms; the term means a methyl or ethyl group.
  • Preferred are methyl and ethyl.
  • (Ci -4 )alkyl preferably means methyl.
  • (Ci -4 )alkyl preferably means methyl, ethyl or butyl.
  • aryl alone or in combination, means a phenyl or a naphthyl group.
  • aryl group may also be independently mono-, di-
  • substituents are independently selected from the group consisting of halogen, cyano, trifluoromethyl, trifluoromethoxy, NR 8 R 9 , N(R 8 )C(O)R 9 , and C(O)NR 8 R 9 .
  • Preferred substituents are independently selected from the group consisting of (Ci -4 )alkyl, halogen and trifluoromethyl.
  • Examples are trifluoromethyl-phenyl (eg 4-trifluoromethyl-phenyl), chloro-phenyl (2-chloro-phenyl, 3-chloro-phenyl and 4-chloro-phenyl), methyl- phenyl (eg 2-methyl-phenyl, 3-methyl-phenyl, 4-methyl-phenyl), dimethyl-phenyl
  • R 1 is preferably substituted by methyl, ethyl, isopropyl, trifluoromethyl, fluorine, chlorine, or methoxy. More preferred R 1 is substituted by methyl, ethyl, trifluoromethyl, fluorine or chlorine. Said groups are preferred substituents for the aryl group.
  • aryl preferably means phenyl.
  • the expression pharmaceutically acceptable salts encompasses either salts with inorganic acids or organic acids like hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, phosphorous acid, nitrous acid, citric acid, formic acid, acetic acid, oxalic acid, maleic acid, lactic acid, tartaric acid, fumaric acid, benzoic acid, mandelic acid, cinnamic acid, pamoic acid, stearic acid, glutamic acid, aspartic acid, methanesulfonic acid, ethanesulfonic acid, ethanedisulfonic acid, p-toluenesulfonic acid, salicylic acid, succinic acid, trifluoroacetic acid, and the like that are non toxic to living organisms or, in case the compound of formula (I) and/
  • Salt-forming groups are groups having basic or acidic properties. Compounds having at least one basic group, for example amino, a secondary amino group not forming a peptide bond or a pyridyl group, may form acid addition salts, for example with inorganic acids. When several basic groups are present mono- or poly-acid addition salts may be formed.
  • Compounds having acidic groups may form metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethylamine or tri-(2-hydroxyethyl)-amine, or heterocyclic bases, for example ⁇ /-ethyl-piperidine or ⁇ /, ⁇ /'-dimethylpiperazine. Mixtures of salts are possible.
  • the present invention encompasses all these forms. Mixtures can be separated in a manner known per se, e.g. by column chromatography, thin layer chromatography (TLC), high performance liquid chromatography (HPLC), or crystallization.
  • the present invention encompasses also solvation complexes of compounds of general formula I and/or Ia.
  • the solvation can be effected in the course of the manufacturing process or can take place separately, e.g. as a consequence of hygroscopic properties of an initially anhydrous compound of general formula I and/or Ia.
  • the compounds specifically mentioned above contain two centers of chirality. The more preferred stereoisomers exhibit the chirality as depicted below (see general formula Ia):
  • a further embodiment of the invention are compounds of formula I and/or Ia wherein
  • Y represents -CH 2 -CH 2 -;
  • R 1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of halogen and trifluoromethyl;
  • R 2 represents (Ci -4 )alkyl;
  • R 3 represents (Ci -4 )alkyl;
  • R 4 represents a phenyl group;
  • R 5 represents (Ci -4 )alkyl.
  • a further embodiment of the invention are compounds of formula I and/or Ia wherein
  • Y represents -CH 2 -CH 2 -;
  • R 1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of methyl, ethyl, fluorine, chlorine and trifluoromethyl;
  • R 2 represents methyl
  • R 3 represents ethyl
  • R 4 represents a phenyl group
  • R 5 represents methyl
  • the compounds of the general formula I and/or Ia 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 I and/or Ia.
  • a further aspect of the present invention relates to pharmaceutical compositions comprising a compound of formula I and/or Ia and a pharmaceutically acceptable carrier material. These pharmaceutical compositions may be used for the treatment or prevention of the above-mentioned diseases.
  • the pharmaceutical compositions can be used for enteral, parenteral, or topical administration. They can be administered, for example, peroral, e.g. in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions or suspensions, nasal, e.g.
  • the invention also relates to the use of a compound of general formula I and/or Ia for the preparation of pharmaceutical compositions for the treatment and/or prevention of the above-mentioned diseases.
  • 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) by bringing the described compounds of general formula I and/or Ia 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.
  • the compounds of general formula I and/or Ia or the above-mentioned pharmaceutical compositions may also be used in combination with one or more other therapeutically useful substances.
  • the present invention also relates to pro-drugs of a compound of general formula I and/or Ia that convert in vivo to the compound of general formula I and/or Ia as such. Any reference to a compound of general formula I and/or Ia is therefore to be understood as referring also to the corresponding pro-drugs of the compound of general formula I and/or Ia, as appropriate and expedient.
  • Compounds as described above have IC 50 values below 150 nM at least on one of the orexin receptors, which have been determined with the FLIPR (Fluorometric Imaging Plates Reader) method described in the experimental section. Preferred compounds are active against both, the OXi and OX 2 receptors.
  • the compounds according to formula (I) and/or (Ia) are useful in the preparation of a medicament for the prevention or treatment of diseases selected from the group consisting of dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apnea; narcolepsy; idiopathic insomnias; parasomnias; benign prostatic hypertrophy; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders; and other diseases related to general orexin system dysfunctions.
  • diseases selected from the group consisting of dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnia
  • Compounds of formula (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders selected from the group consisting of eating or drinking disorders, all types of sleep disorders, or cognitive dysfunctions in the healthy population and in psychiatric and neurologic 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 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.
  • 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.
  • the compounds of the general formula I and/or Ia of the present invention may be prepared according to the procedures described herein, especially as described in the experimental part.
  • the compounds according to general formula (I) and/or (Ia) are useful in 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
  • Compounds of general formula (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 of general formula (I) and (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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.
  • R 1 , R 2 , R 3 , R 4 , R 5 and Y are as defined in general formula I 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 F is descirbed in WO05/1 18548.
  • Intermediate A is reacted with the unsymmetrical hydrazine B to give via a 6-step sequence (see experimental part) the 2,5-di-substituted pyrazole-3-yl-ethylamine C in a regioselective manner.
  • Y shall represent -CH2-CH2- in the final compounds
  • the intermediate C is reacted with an aldehyde-derivative D under Pictet-Spengler conditions and microwave irradiation to give precursor E which can be N-alkylated by compounds F to give final orexin receptor antagonists G.
  • the intermediate C is reacted with carboxylic acid derivatives under amide-bond forming conditions to give the amide-intermediates ⁇ which are transformed to the cyclic imines J under Bischler-Napieralski conditions followed by imine reduction to give the amine precursors E.
  • the final orexin receptor antagonists G could be obtained.
  • the Bischler-Napieralski pathway is as well suitable for the preparation of compounds wherein Y represents -CH 2 - CH 2 -;
  • the Pictet-Spengler pathway is not suitable for the preparation of compounds wherein Y represents -CH 2 - or -CH 2 -O- as the aldehyde intermediates D comprising these structural features are not stable enough to react via the desired pathway under the conditions applied in the Pictet-Spengler reaction to prepare intermediates E).
  • the compounds of the formula A, B, D, F and JH are either commercially available, or can be synthesized according to methods known in the art.
  • R 1 , R 2 , R 3 , R 4 , R 5 and Y are as defined in general formula I above, X represents OH or Cl and Z represents Br or OTs;
  • X represents OH or Cl
  • Z represents Br or OTs;
  • intermediate E is reacted with ester derivatives K (instead of amide derivatives F) to give intermediate L, which can either be directly transformed into the final compounds G by reacting the ester with an amine derivative U or which can first be hydrolyzed to the acid intermediate M followed by reaction with an amine N to give final compounds G.
  • ester derivatives K instead of amide derivatives F
  • intermediate L which can either be directly transformed into the final compounds G by reacting the ester with an amine derivative U or which can first be hydrolyzed to the acid intermediate M followed by reaction with an amine N to give final compounds G.
  • the compounds of the formula t ⁇ and K are either commercially available, or can be synthesized according to methods known in the art.
  • Nickel(ll)-chloride in methanol in the presence of BoC 2 O Deprotection to the free amine hydrochloride was achieved via a standard procedure with 4M HCI in dioxane to give, after suspending it in diethylether, filtering and drying, 10 as a white powder.
  • the nitrile precursor 8 was reduced to the amine-intermediate by LAH.
  • the crude product was dissolved in a mixture of diethylether / THF followed by the addition of an aqueous base (e.g.
  • Scheme 1.2 summarizes the preparation of further regioselectively unsymmethcally substituted pyrazole-3-yl-ethylamines.
  • the synthesis again starts from the regioselective condensation of precursor 5 with an unsymmethcal hydrazine-derivative (e.g. benzylhydrazine 13) to give the pyrazole-intermediate 14 which after hydrolysis / decarboxylation resulted in the ester 15.
  • Reduction of the ester 15 with LAH gave the alcohol 16, which after deprotonation with sodium hydride in DMF was transformed into the tosylate-intermediate and followed by the addition of potassium phthalimide (Gabriel synthesis) to give the amine- precursor 18.
  • Hydrazine cleavage of the phthalimide resulted in the formation of the 2-benzyl substituted pyrazole-3-yl-ethylamine derivative 19.
  • Compounds 20 to 23 were prepared according to the same
  • Scheme 5 summarizes the sequence applied for the preparation of 4-"phenethyl"- substituted examples.
  • the pyrazol-ethylamine derivative 12 can be reacted with the phenyl-propionaldehyde 26 in ethanol in the presence of 2 equivalents of acetic acid in the microwave oven for 6 minutes (100 Watt; 130°C, 14 bar) to give after HPLC purification the secondary amine-intermediates 72.
  • S N 2-reaction with the tosylate 73 (described in WO 2005/118548) in methyl-iso- butyl-ketone at 90°C for 24 h under inert conditions led to the final compound 74 (Example 1 ).
  • Examples 2 to 42 could be prepared according to the sequence described in Scheme 5.
  • the following examples illustrate the invention but do not limit the scope thereof. All temperatures are stated in °C.
  • HPLC conditions Analytic: Zorbax 59 SB Aqua column, 4.6 x 50 mm from Agilent Technologies.
  • the isocratic mixture may vary, depending on the compounds.
  • meldrum's acid 50 g, 347 mmol
  • DCM 300 ml
  • TEA 35.1 g, 347 mmol
  • the reaction mixture was stirred for 2 h at r.t. cooled again to 0°C and 1 M hydrochloric acid solution (700 ml) was slowly added.
  • LAH (806 mg, 21.22 mmol) was suspended in anhydrous diethylether (105 ml) at 0°C in an inert atmosphere.
  • Step 1
  • Step2 To the solution of 5-(2-benzyl-5-methyl-2H-pyrazol-3-yl)-2,2-dimethyl- [1 ,3]dioxane-4,6-dione obtained in step 1 , pTsOH (3.74 g, 19.4 mmol) was added and stirring was continued for 2 h at 60°C. The reaction mixture was cooled to 0°C and TEA (1.96 g, 19.4 mmol) was added drop by drop. Stirring was continued for 15 minutes.
  • LAH (250 mg, 6.6 mmol) was suspended in THF (18 ml) in an inert atmosphere and cooled to 0°C followed by the dropwise addition of a solution of (2-benzyl-5- methyl-2H-pyrazol-3-yl)-acetic acid methyl ester (1.07 g, 4.4 mmol) in THF (4 ml). Stirring was continued for 2 h at rt. At 0°C, water (0.25 ml), 15% aqueous NaOH- solution (0.25 ml) and again water (0.75 ml) were subsequently added. Stirring was continued for several hours followed by the addition of sodium sulfate and filtration.
  • 3-(4-Thfluoromethyl-phenyl)-propionic acid (10 g, 45.8 mmol) was dissolved in anhydrous THF (250 ml) and cooled to 0°C followed by the addition of a 1 M solution of borane in THF (69 ml, 69 mmol). Stirring was continued at 0°C for 1 h and 16 h at rt followed by slow addition of methanol (100 ml) and water (100 ml). The organic solvents were evaporated under reduced pressure.
  • Example 1 2- ⁇ 1 -Ethyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpyndin-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 8 2- ⁇ 1 -Ethyl-3-methyl-4-[2-(3-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpyndin-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 12 2- ⁇ 1-Ethyl-4-[2-(3-fluoro-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo ⁇ S-cJpyridin-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 14 2- ⁇ 4-[2-(2,3-Difluoro-phenyl)-ethyl]-1-ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 17 2- ⁇ 4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 28 2- ⁇ 1 -Ethyl-4-[2-(2-methoxy-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 29 N-Methyl-2- ⁇ 3-methyl-1 -phenethyl-4-[2-(4-trifluoromethyl-phenyl)- ethyl]-1 ,4,6,7-tetrahydro-pyrazolo[4,3-c]pyridin-5-yl ⁇ -2-phenyl-acetamide:
  • Example 32 2- ⁇ 1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 33 2- ⁇ 4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • Example 38 2- ⁇ 1 -Ethyl-3-methyl-4-[2-(2,3,4-trifluoro-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo4,3c]pyridin-5-yl ⁇ -N-methyl-2-phenyl-acetamide:
  • Example 40 2- ⁇ 1 -Ethyl-3-methyl-4-[2-(2-methyl-5-t ⁇ fluoromethyl-phenyl)-ethyl]- 1,4,6,7-tetrahydro-pyrazolo[4,3-, c]pyridin-5-yl - ⁇ N-methyl-2-phenyl-acetamide:
  • Example 42 2- ⁇ 4-[2-(3,5-Bis-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ⁇ J-tetrahydro-pyrazolo ⁇ -clpy ⁇ din-S-ylJ-N-methyl ⁇ -phenyl-acetamide:
  • the orexin receptor antagonistic activity of the compounds of formula (I) and/or (Ia) is determined in accordance with the following experimental method.
  • 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.
  • 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 OXi and OX 2 receptors. Selected compounds are displayed in Table 1. Table 1:

Abstract

The invention relates to novel pyrazolo-tetrahydropyridines compounds and their use as orexin receptor antagonists.

Description

Novel pyrazolo-tetrahydropyridine derivatives
The present invention relates to novel compounds of formula (I) and/or (Ia) 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 (I) and/or (Ia), 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 such as dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apnea; narcolepsy; idiopathic insomnias; parasomnias; benign prostatic hypertrophy; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders; and other diseases related to general orexin system dysfunctions. The present invention provides substituted 1 ,2,3,4-tetrahydro-pyrrolo[1 ,2- a]pyrazine and 5,6,7,8-tetrahydro-imidazo[1 ,5-a]pyrazine derivatives, which are non-peptide antagonists of human orexin receptors, in particular 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 OX1 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 (WO02/051838), and 1 ,2,3,4-tetrahydroisoquinoline derivatives (WO01/68609, WO2004/085403) are disclosed by Actelion Pharmaceuticals Ltd. 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 I
General Formula I
Figure imgf000003_0001
wherein Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of
Figure imgf000003_0002
halogen and trifluoromethyl; 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.
The compounds of the general formula I and/or Ia (see below) contain one or more asymmetric carbon atoms and may be prepared in form of optically pure enantiomers, mixtures of enantiomers such as racemates, diastereomers, mixtures of diastereomers, diastereomeric racemates, mixtures of diastereomeric racemates, or meso-forms.
The compounds of the general formula I and/or Ia (see below) also encompass the pharmaceutically acceptable salts thereof. In the present description the term "halogen" means fluorine, chlorine, bromine or iodine 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; in analogy the term
Figure imgf000004_0001
means a straight-chain or branched-chain alkyl group with 2 to 4 carbon atoms; the term
Figure imgf000004_0002
means a methyl or ethyl group. Preferred are methyl and ethyl.
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.
The term "aryl", alone or in combination, means a phenyl or a naphthyl group.
Preferred is a phenyl group. The aryl group may also be independently mono-, di-
, or trisubstituted wherein the substituents are independently selected from the group consisting of
Figure imgf000004_0003
halogen, cyano, trifluoromethyl, trifluoromethoxy, NR8R9, N(R8)C(O)R9, and C(O)NR8R9. Preferred substituents are independently selected from the group consisting of (Ci-4)alkyl, halogen and trifluoromethyl. Examples are trifluoromethyl-phenyl (eg 4-trifluoromethyl-phenyl), chloro-phenyl (2-chloro-phenyl, 3-chloro-phenyl and 4-chloro-phenyl), methyl- phenyl (eg 2-methyl-phenyl, 3-methyl-phenyl, 4-methyl-phenyl), dimethyl-phenyl
(eg 2,3-dimethyl-phenyl, 2,4-dimethyl-phenyl), dimethoxy-phenyl (eg 2,5- dimethoxy-phenyl, 2,4-dimethoxy-phenyl), fluoro-methoxy-phenyl (eg 3-fluoro-4- methoxy-phenyl), dichloro-phenyl (eg 2,4-dichloro-phenyl), difluoro-phenyl (eg 3,4- difluoro-phenyl).
R1 is preferably substituted by methyl, ethyl, isopropyl, trifluoromethyl, fluorine, chlorine, or methoxy. More preferred R1 is substituted by methyl, ethyl, trifluoromethyl, fluorine or chlorine. Said groups are preferred substituents for the aryl group.
For the substituent R4 the term "aryl" preferably means phenyl. The expression pharmaceutically acceptable salts encompasses either salts with inorganic acids or organic acids like hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, phosphorous acid, nitrous acid, citric acid, formic acid, acetic acid, oxalic acid, maleic acid, lactic acid, tartaric acid, fumaric acid, benzoic acid, mandelic acid, cinnamic acid, pamoic acid, stearic acid, glutamic acid, aspartic acid, methanesulfonic acid, ethanesulfonic acid, ethanedisulfonic acid, p-toluenesulfonic acid, salicylic acid, succinic acid, trifluoroacetic acid, and the like that are non toxic to living organisms or, in case the compound of formula (I) and/or (Ia) is acidic in nature, with an inorganic base like an alkali or earth alkali base, e.g. sodium hydroxide, potassium hydroxide, calcium hydroxide and the like. For other examples of pharmaceutically acceptable salts, reference can be made notably to "Salt selection for basic drugs", Int. J. Pharm. (1986), 33, 201- 217.
Salt-forming groups are groups having basic or acidic properties. Compounds having at least one basic group, for example amino, a secondary amino group not forming a peptide bond or a pyridyl group, may form acid addition salts, for example with inorganic acids. When several basic groups are present mono- or poly-acid addition salts may be formed.
Compounds having acidic groups, such as a carboxyl group or a phenolic hydroxy group, may form metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethylamine or tri-(2-hydroxyethyl)-amine, or heterocyclic bases, for example Λ/-ethyl-piperidine or Λ/,Λ/'-dimethylpiperazine. Mixtures of salts are possible.
Compounds having both acidic and basic groups can form internal salts / zwitterions.
For the purposes of isolation or purification, as well as in the case of compounds that represent intermediates, it is as well possible to use pharmaceutically unacceptable salts, e.g. the picrates. However, only pharmaceutically acceptable, non-toxic salts may be used for therapeutic purposes, and are therefore preferred.
The present invention encompasses all these forms. Mixtures can be separated in a manner known per se, e.g. by column chromatography, thin layer chromatography (TLC), high performance liquid chromatography (HPLC), or crystallization. The present invention encompasses also solvation complexes of compounds of general formula I and/or Ia. The solvation can be effected in the course of the manufacturing process or can take place separately, e.g. as a consequence of hygroscopic properties of an initially anhydrous compound of general formula I and/or Ia. The compounds specifically mentioned above contain two centers of chirality. The more preferred stereoisomers exhibit the chirality as depicted below (see general formula Ia):
R-chirality
General Formula Ia
Figure imgf000006_0001
S-chirality A further embodiment of the invention are compounds of formula I and/or Ia wherein
Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of
Figure imgf000007_0001
halogen and trifluoromethyl; R2 represents (Ci-4)alkyl; R3 represents (Ci-4)alkyl; R4 represents a phenyl group; R5 represents (Ci-4)alkyl.
A further embodiment of the invention are compounds of formula I and/or Ia wherein
Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of methyl, ethyl, fluorine, chlorine and trifluoromethyl;
R2 represents methyl;
R3 represents ethyl; R4 represents a phenyl group;
R5 represents methyl.
Especially preferred compounds are listed below:
2-{1 -Ethyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(4-ethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1 -Ethyl-4-[2-(2-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,3-Difluoro-4-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1-Ethyl-3-methyl-4-[2-(2,3,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-[1-Ethyl-3-methyl-4-(2-p-tolyl-ethyl)-1 ,4,6,7-tetrahydro-pyrazolo[4,3- c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(4-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-3-methyl-4-[2-(3-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-[4-(2-Benzo[1 ,3]dioxol-5-yl-ethyl)-1 -ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(4-isopropyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(3-fluoro-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,3-Difluoro-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(4-Chloro-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1 ,3-Dimethyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1-Ethyl-3-methyl-4-[2-(4-methyl-3-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyndin-S-ylJ-N-methyl^-phenyl-acetamide;
2-[1-Ethyl-3-methyl-4-(2-m-tolyl-ethyl)-1 ,4,6,7-tetrahydro-pyrazolo[4,3- c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide; 2-[1 -Ethyl-3-methyl-4-(2-o-tolyl-ethyl)-1 ,4,6,7-tetrahydro-pyrazolo[4,3- c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide;
2-{1-Butyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(3-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-3-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Benzyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-methoxy-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
N-Methyl-2-{3-methyl-1-phenethyl-4-[2-(4-tπfluoromethyl-phenyl)-ethyl]- 1 ,4,6,7-tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-2-phenyl-acetamide;
N-Methyl-2-{3-methyl-1-(2,2,2-tπfluoro-ethyl)-4-[2-(4-trifluoromethyl- phenyO-ethyO-I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-yl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{4-[2-(2,4-Dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,4-Difluoro-3-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Ethyl-3-methyl-4-[2-(2,4,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-3-methyl-4-[2-(2,4,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-3-methyl-4-[2-(2,3,4-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-3-methyl-4-[2-(2-methyl-5-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(3-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{4-[2-(3,5-Bis-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,3-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{4-[2-(2,6-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3-methyl-
I ^^J-tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide; Out of this group of compounds, especially preferred compounds are:
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1-Ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo[ 4,3-,c ]pyrdin-5-yl}-N- -methyl-2phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- - tetrahydro-pyrazolo [4,3--c] pyridin -5-yl -N-methyl-2--phenyl-acetamide; 2-{4-[2-(2,3-Difluoro-4-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo[4,3- c]pyridin-5-yl}N-methyl-2phenyl-acetamide;
2-{1-Ethyl-4-[2-(4-ethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1-ethyl-3-methyl- 1,4,6,7-tetrahydro-pyrazolo[4 , 3--c] pyridin -5- yl}--N-methyl-2phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-methyl-phenyl)-ethyl]-1-ethy-l-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo[ 4,3--c]pyridin-5-yl}-N-methyll-2--phenyl-acetamide;
The compounds of the general formula I and/or Ia 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 I and/or Ia. A further aspect of the present invention relates to pharmaceutical compositions comprising a compound of formula I and/or Ia and a pharmaceutically acceptable carrier material. These pharmaceutical compositions may be used for the treatment or prevention of the above-mentioned diseases. The pharmaceutical compositions can be used for enteral, parenteral, or topical administration. They can be administered, for example, peroral, e.g. in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions or suspensions, nasal, e.g. in the form of sprays, rectal, e.g. in the form of suppositories, parenteral, e.g. in the form of injection solutions or infusion solutions, or topical, e.g. in the form of ointments, creams or oils. The invention also relates to the use of a compound of general formula I and/or Ia for the preparation of pharmaceutical compositions for the treatment and/or prevention of the above-mentioned diseases.
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) by bringing the described compounds of general formula I and/or Ia 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. The compounds of general formula I and/or Ia or the above-mentioned pharmaceutical compositions may also be used in combination with one or more other therapeutically useful substances.
The present invention also relates to pro-drugs of a compound of general formula I and/or Ia that convert in vivo to the compound of general formula I and/or Ia as such. Any reference to a compound of general formula I and/or Ia is therefore to be understood as referring also to the corresponding pro-drugs of the compound of general formula I and/or Ia, as appropriate and expedient. Compounds as described above have IC50 values below 150 nM at least on one of the orexin receptors, which have been determined with the FLIPR (Fluorometric Imaging Plates Reader) method described in the experimental section. Preferred compounds are active against both, the OXi and OX2 receptors.
The compounds according to formula (I) and/or (Ia) are useful in the preparation of a medicament for the prevention or treatment of diseases selected from the group consisting of dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apnea; narcolepsy; idiopathic insomnias; parasomnias; benign prostatic hypertrophy; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders; and other diseases related to general orexin system dysfunctions.
Compounds of formula (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders selected from the group consisting of eating or drinking disorders, all types of sleep disorders, or cognitive dysfunctions in the healthy population and in psychiatric and neurologic 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 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.
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. The compounds of the general formula I and/or Ia of the present invention may be prepared according to the procedures described herein, especially as described in the experimental part.
The compounds according to general formula (I) and/or (Ia) are useful in 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. Compounds of general formula (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 of general formula (I) and (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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 (I) and/or (Ia) are particularly suitable for use in the treatment of diseases or disorders 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.
General methods for the preparation of compounds of general formula I and/or Ia:
Figure imgf000018_0001
R1, R2, R3, R4, R5 and Y are as defined in general formula I above, X represents OH or Cl and Z represents Br or OTs; The preparation of an example for intermediates F is descirbed in WO05/1 18548.
Intermediate A is reacted with the unsymmetrical hydrazine B to give via a 6-step sequence (see experimental part) the 2,5-di-substituted pyrazole-3-yl-ethylamine C in a regioselective manner. In case Y shall represent -CH2-CH2- in the final compounds, the intermediate C is reacted with an aldehyde-derivative D under Pictet-Spengler conditions and microwave irradiation to give precursor E which can be N-alkylated by compounds F to give final orexin receptor antagonists G. In case Y shall represent -CH2- or -CH2-O- in the final compounds G, the intermediate C is reacted with carboxylic acid derivatives under amide-bond forming conditions to give the amide-intermediates \ which are transformed to the cyclic imines J under Bischler-Napieralski conditions followed by imine reduction to give the amine precursors E. By N-alkylation with derivatives F, the final orexin receptor antagonists G could be obtained. (The Bischler-Napieralski pathway is as well suitable for the preparation of compounds wherein Y represents -CH2- CH2-; The Pictet-Spengler pathway is not suitable for the preparation of compounds wherein Y represents -CH2- or -CH2-O- as the aldehyde intermediates D comprising these structural features are not stable enough to react via the desired pathway under the conditions applied in the Pictet-Spengler reaction to prepare intermediates E).
The compounds of the formula A, B, D, F and JH are either commercially available, or can be synthesized according to methods known in the art.
Figure imgf000019_0001
R1, R2, R3, R4, R5 and Y are as defined in general formula I above, X represents OH or Cl and Z represents Br or OTs; For general methods see also WO 00/168609 and WO 02/051838 and WO 04/085403
In a slightly different route the intermediate E is reacted with ester derivatives K (instead of amide derivatives F) to give intermediate L, which can either be directly transformed into the final compounds G by reacting the ester with an amine derivative U or which can first be hydrolyzed to the acid intermediate M followed by reaction with an amine N to give final compounds G. The compounds of the formula t± and K are either commercially available, or can be synthesized according to methods known in the art.
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 I and/or Ia:
Figure imgf000020_0001
MeOH P-TsOH
Figure imgf000020_0002
Scheme 1.1 : Synthesis of pyrazole precursors The preparation of the pyrazole-3-yl-ethylamine-derivatives started by the coupling of diketene (1) and Meldrum's acid (2) in a solvent like DCM in the presence of a base like TEA to give after acidc work-up the intermediate 3. Reaction of compound 3 with an appropriate hydrazine derivative (e.g. ethylhydrazine (4)) in solvent like methanol in the presence of a base like TEA resulted in the formation of the pyrazole-derivative 5. Hydrolysis under acidic conditions (with e.g. p-TsOH in methanol) resulted in the formation of the methylester 6 which was efficiently transformed to the primary amide 7 by stirring it in methanolic ammonia (7M) for 20 h. Dehydration of 7 to prepare the nitrile 8 was accomplished by reaction with TFAA in dioxane in the presence of pyridine. Reduction of the nitirle 8 to the Boc-protected pyrazole-3-yl-ethylamine-dehvative 9 was accomplished by the use of sodium-borohydride and catalytic amounts of
Figure imgf000022_0001
The following compounds were prepared along the same sequence
Figure imgf000022_0002
20 21 22 23
Scheme 1.2: Preparation of 2-substituted 5-methyl-pyrazol-3-yl-ethylamines
Nickel(ll)-chloride in methanol in the presence of BoC2O. Deprotection to the free amine hydrochloride was achieved via a standard procedure with 4M HCI in dioxane to give, after suspending it in diethylether, filtering and drying, 10 as a white powder. In order to obtain the free amine 12, the nitrile precursor 8 was reduced to the amine-intermediate by LAH. After work-up according to the "Corey-proceudre" the crude product was dissolved in a mixture of diethylether / THF followed by the addition of an aqueous base (e.g. 1 M sodium bicarbonate) and benzyl chloroformiate to give the Cbz-protected amine 11 which could then be purified by column chromatography and after purification deprotected by hydrogenolysis to give the desired compound 12. Whether compound 10 or compound 12 were prepared was dependent on the subsequent transformations intended.
Scheme 1.2 summarizes the preparation of further regioselectively unsymmethcally substituted pyrazole-3-yl-ethylamines. The synthesis again starts from the regioselective condensation of precursor 5 with an unsymmethcal hydrazine-derivative (e.g. benzylhydrazine 13) to give the pyrazole-intermediate 14 which after hydrolysis / decarboxylation resulted in the ester 15. Reduction of the ester 15 with LAH gave the alcohol 16, which after deprotonation with sodium hydride in DMF was transformed into the tosylate-intermediate and followed by the addition of potassium phthalimide (Gabriel synthesis) to give the amine- precursor 18. Hydrazine cleavage of the phthalimide resulted in the formation of the 2-benzyl substituted pyrazole-3-yl-ethylamine derivative 19. Compounds 20 to 23 were prepared according to the same sequence of transformations.
Figure imgf000024_0001
According to this 2 step procedure the following phenyl-propanal derivatives were prepared:
Figure imgf000024_0002
Scheme 2: Preparation of substituted 3-phenyl-propionaldehyde precursors
In case the hydrocinnamic acid derivatives were commercially available the 3- phenyl-propionaldehydes were prepared by the two step sequence depicted in Scheme 2; e.g. 4-trifluoromethyl-hydrocinnamic acid (24) was reduced to the 3- (4-Trifluoromethyl-phenyl)-propan-1-ol (25) according to standard protocols with borane in THF (1 M). Oxidation to the aldehyde 26 was achieved by PCC in DCM according to a standard protocol developed by Corey et al. The substituted 3- phenyl-propionaldehydes 27 to 39 were synthesized according to the exact same procedure. Due to the fact that the 3-phenylpropananal-compounds were only of limited stability, they were usually prepared just before their use in the subsequent Pictet-Spengler-reacύon. 41
Figure imgf000025_0001
Tho following compounds were prepared according to the same sequence:
Figure imgf000025_0002
Figure imgf000025_0003
Figure imgf000025_0004
Figure imgf000025_0005
Figure imgf000025_0006
Scheme 3: Preparation of substituted phenyl-propionaldehydes In case neither the cinnamic acids nor the hydrocinnamic acids were available, the phenylpropionaldehyde derivatives were prepared according to the procedure depicted in Scheme 3 and as for example starting with a Knoevenagel condensation of a 3-thfluoromethyl-benzaldehyde 40 with malonic acid (41 ) in pyridine in the presence of piperidine to give, after acidic work-up, the cinnamic acid derivative 42. Catalytic hydrogenation under standard conditions (1 bar H2; Pd-C 10%; methanol; rt; 3h) led to the hydrocinnamic acid compound 43. The acid function was subsequently reduced by BH3 in THF to give the alcohol 44 followed by PCC-oxidation to give the desired aldehyde 45. Compounds 46 to 64 can be prepared according to the same synthetic sequence.
Figure imgf000026_0001
68 69
The following compounds were prepared according to the same sequence:
Figure imgf000026_0002
Scheme 4: Preparation of further substituted phenyl-propionaldehydes
In Scheme 4 an alternative pathway for the preparation of substituted phenylpropionylaldehydes is depicted. 2-Fluoro-3-methylbenzylbromide (65) is reacted with diethylmalonate to give intermediate 66. Esterhydrolysis under basic conditions followed by decarboxylation under acidic conditions resulted in the hydrocinnamic acid intermediate 67. Subsequent borane-reduction (-> alcohol 68) and PCC-oxidation led to the desired substituted phenyl-propionaldehyde 69. Compounds 70 and 71 can be prepared according to the same synthetic sequence.
Figure imgf000027_0001
Scheme 5: Synthesis of final compounds (e.g. Example 1 )
Scheme 5 summarizes the sequence applied for the preparation of 4-"phenethyl"- substituted examples. For example the pyrazol-ethylamine derivative 12 can be reacted with the phenyl-propionaldehyde 26 in ethanol in the presence of 2 equivalents of acetic acid in the microwave oven for 6 minutes (100 Watt; 130°C, 14 bar) to give after HPLC purification the secondary amine-intermediates 72. SN2-reaction with the tosylate 73 (described in WO 2005/118548) in methyl-iso- butyl-ketone at 90°C for 24 h under inert conditions led to the final compound 74 (Example 1 ). Examples 2 to 42 could be prepared according to the sequence described in Scheme 5. The following examples illustrate the invention but do not limit the scope thereof. All temperatures are stated in °C.
Abbreviations (as used herein) aq. aqueous
Boc tert.-butyloxycarbonyl
DCM dichloromethane
DMF dimethylformamide
DMSO dimethylsulfoxide
EDC N-(3-dimethylaminopropyl)-N'-ethyl-carbodiimid
ELSD Evaporative Light-Scattering Detection
Et ethyl
EtOAc ethyl acetate h hour(s)
HPLC High Performance Liquid Chromatography
HV High Vacuum
LAH lithium aluminumhydride
LC-MS Liquid Chromatography - Mass Spectroscopy min minute(s)
MS Mass Spectroscopy
PBS phosphate buffered saline
PyBOP benzothazole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate
PCC pyridinium chlorochromate
P-TsOH para-toluenesulfonic acid rt room temperature sat. saturated
TBTU O-benzotriazol-1 -yl-Λ/,Λ/,Λ/',Λ/-tetramethyluronium tetrafluoroborate
TEA triethylamine
TFA trifluoroacetic acid
TFAA trifluoroacetic anhydride THF tetrahydrofurane
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 + 0.5% TFA. Gradient: 90% B → 5% B over 2 min. Flow: 1 mL/min. Detection: UV/Vis + MS.
Preparative: Zorbax SB Aqua column, 20 x 500 mm from Agilent Technologies.
Eluent: A: Acetonitrile; B: H2O + 0.05% ammonium hydroxide (25% aq.). Gradient: 80% B → 10% B over 6 min. Flow: 40 mL/min. Detection: UV + MS, or UV + ELSD.
Chiral, analytic: Regis Whelk column, 4.6 x 250 mm, 10 μm. Eluent A: EtOH +
0.05% EtβN. Eluent B: hexane. lsocratic conditions, usually 60% B, over 40 min,
1 mL/min. The isocratic mixture may vary, depending on the compounds. Chiral, preparative: As analytical conditions, but on a Regis Whelk 01 column,
50x250 mm and a flow of 100 mL/min.
Preparation of precursors and intermediates:
Figure imgf000029_0001
In an inert atmosphere, meldrum's acid (50 g, 347 mmol) was dissolved in DCM (300 ml) and TEA (35.1 g, 347 mmol) was added. The resulting mixture was cooled to 0°C followed by drop by drop addition of diketene (35 g, 416.3 mmol). The reaction mixture was stirred for 2 h at r.t. cooled again to 0°C and 1 M hydrochloric acid solution (700 ml) was slowly added. The layers were separated and the organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 79.36 g (quantitative yield) of the product 5-(1 -hydroxy-3-oxo-butylidene)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione as an orange solid. LC-MS: tR = 0.55 min; [M+H]+ = no ionisation.
Figure imgf000030_0001
5-(1-Hydroxy-3-oxo-butylidene)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione (10 g, 44 mmol) was dissolved in anhydrous methanol (140 ml) and cooled to 0°C followed by drop by drop addition of a solution of ethylhydrazine oxalate (7.24 g, 48.2 mmol) and TEA (9.75 g, 96.4 mmol) in anhydrous methanol (60 ml) with the help of a dropping funnel within 15 minutes. Upon completion of the addition, the mixture was slowly warmed to 60°C and stirring was continued for 90 minutes. The reaction mixture was concentrated to dryness under reduced pressure and further dried at HV for 15 minutes to give 11.5 g (quantitative yield) of the product 5-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione as an orange oil which was used in the next step without further purification. LC-MS: tR = 0.52 min; [M+H]+ = no ionisation.
Figure imgf000030_0002
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 drop by drop over a period of 20 minutes. The resulting mixture was concentrated under reduced pressure. The residue was taken into DCM (200 ml) and washed with water (2 x 75 ml), dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (silicagel, DCM / methanol = 30 / 1 ) to give 5.56 g(70%) of (2-ethyl-5-methyl-2H-pyrazol-3-yl)-acetic acid methyl ester. LC-MS: tR = 0.62 min; [M+H]+ = 183.18.
Figure imgf000031_0001
(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-acetic acid methyl ester (4.64 g, 25.46 mmol) was dissolved in 7N ammonia in methanol (100 ml, 700 mmol NH3). The reaction mixture was stirred at rt for 2Oh. The solvent was removed under reduced pressure to give 4.25 g (quant, yield) of 2-(2-ethyl-5-methyl-2H-pyrazol-3-yl)- acetamide. LC-MS: tR = 0.33 min; [M+H]+ = no ionisation.
Figure imgf000031_0002
2-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-acetamide (2.0 g, 11.96 mmol) was dissolved in dioxane (180 ml) followed by the addition of pyridine (1.89 g, 23.9 mmol). The resulting solution was cooled to 0°C and TFAA (2.76 g, 13.16 mmol) was added drop by drop. The suspension was then slowly warmed to rt and stirring was continued in total for 4 h. The volatiles were removed under reduced pressure, the residue was dissolved in DCM (150 ml), washed with water (2 x 100 ml), dried over magnesium sulfate, filtered and concentrated under reduced pressure. After flash-chromatography (silicagel; DCM / methanol = 40 / 1 ), 1.58 g (88%) of (2-ethyl-5-methyl-2H-pyrazol-3-yl)-acetonitrile was obtained. LC-MS: tR = 0.65 min; [M+H]+ = 150.18.
Figure imgf000032_0001
LAH (806 mg, 21.22 mmol) was suspended in anhydrous diethylether (105 ml) at 0°C in an inert atmosphere. (2-Ethyl-5-methyl-2H-pyrazol-3-yl)-acetonithle (1.58 g, 10.6 mmol) dissolved in diethylether (10 ml) was added dropwise at 0°C over a period of 11 minutes followed by the addition of anhydrous THF (115 ml). Stirring was continued for 2.5 h followed by careful addition of water (0.8 ml), 15% aqueous sodium hydroxide solution (0.8 ml) and water (2.4 ml). Stirring was continued at rt for 10 minutes. The mixture was filtered, the filter cake was washed with diethylether and the filtrate was concentrated under reduced pressure. The crude 2-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-ethylamine was further transformed to the Z-protected amine in the next step without prior purification. LC-MS: tR = 0.2 min (broad peak); [M+H]+ = no ionization.
Figure imgf000032_0002
The crude primary amine 2-(2-ethyl-5-methyl-2H-pyrazol-3-yl)-ethylamine
(theoretical amount: 1.62 g, 10.6 mmol) was dissolved in diethylether (115 ml), THF (115 ml) and water (55 ml) at 0°C followed by the addition of sodium bicarbonate (2.22 g, 26.5 mmol) and drop by drop addition of benzyl chloroformate (1.99 g, 11.7 mmol). Stirring was continued for 30 min. The layers were separated and the aqueous layer was extracted with diethylether (55 ml). The combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. Purification was accomplished by flash chromatography (DCM / methanol = 50 / 1 ) to give 881 mg (29%) of [2-(2-ethyl-5- methyl-2H-pyrazol-3-yl)-ethyl]-carbamic acid benzyl ester. LC-MS: tR = 0.76 min; [M+H]+ = 288.24.
Figure imgf000033_0001
[2-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-ethyl]-carbamic acid benzyl ester (818 mg, 2.85 mmol) was dissolved in methanol (8 ml) and added to a suspension of Pd-C (10%, 163 mg) in methanol (8 ml), put under an atmosphere of hydrogen (1 bar) and stirring at rt was continued for 45 minutes. The reaction mixture was filtered over a pad of celite and the filtrate was concentrated under reduced pressure to give 408 mg (93.7%) of 2-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-ethylamine. LC-MS: tR = 0.20 min, broad peak; [M+H]+ = no ionization.
Figure imgf000033_0002
Step 1 :
5-(1-Hydroxy-3-oxo-butylidene)-2,2-dimethyl-[1 ,3]dioxane-4,6-dione (1 g, 4.4 mmol) was dissolved in anhydrous methanol (8.1 ml) and cooled to 0°C followed by the addition of a solution of benzylhydrazine dihydrochloride (885 mg, 4.4 mmol) and TEA (979 mg, 9.7 mmol) in anhydrous methanol (6 ml). The reaction mixture was heated to 60°C for 2 h. Product was detected by LC-MS. Step 2 was performed without any prior work-up. LC-MS: tR = 0.66 min; [M+H]+ = 315.4. Step2: To the solution of 5-(2-benzyl-5-methyl-2H-pyrazol-3-yl)-2,2-dimethyl- [1 ,3]dioxane-4,6-dione obtained in step 1 , pTsOH (3.74 g, 19.4 mmol) was added and stirring was continued for 2 h at 60°C. The reaction mixture was cooled to 0°C and TEA (1.96 g, 19.4 mmol) was added drop by drop. Stirring was continued for 15 minutes. The mixture was concentrated under reduced pressure, the residue taken up into DCM (30 ml), washed with water (2 x 15 ml), dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 1.03 g (96%) of (2-benzyl-5-methyl-2H-pyrazol-3-yl)-acetic acid methyl ester which was used in the next step without further purification. LC-MS: tR = 0.83 min; [M+H]+ = 245.16.
Figure imgf000034_0001
LAH (250 mg, 6.6 mmol) was suspended in THF (18 ml) in an inert atmosphere and cooled to 0°C followed by the dropwise addition of a solution of (2-benzyl-5- methyl-2H-pyrazol-3-yl)-acetic acid methyl ester (1.07 g, 4.4 mmol) in THF (4 ml). Stirring was continued for 2 h at rt. At 0°C, water (0.25 ml), 15% aqueous NaOH- solution (0.25 ml) and again water (0.75 ml) were subsequently added. Stirring was continued for several hours followed by the addition of sodium sulfate and filtration. The filtrate was concentrated under reduced pressure to give 850 mg (89%) of 2-(2-benzyl-5-methyl-2H-pyrazol-3-yl)-ethanol directly used in the next step without further purification. LC-MS: tR = 0.66 min; [M+H]+ = 217.15.
Figure imgf000035_0001
2-(2-Benzyl-5-methyl-2H-pyrazol-3-yl)-ethanol (848 mg, 3.92 mmol) was dissolved in DMF (21 ml) and cooled to -20°C followed by the addition of sodium hydride (218 mg, 5.02 mmol). Stirring was continued at rt for 30 minutes. The reaction mixture was again cooled to -20°C and toluene-4-suofonyl chloride (pTs-CI, 966 mg, 5.02 mmol) was added. The cooling bath was removed and stirring was continued for 2 h at rt and 30 minutes at 50°C followed by the addition of potassium phthalimid (1.7 g, 9.2 mmol). Stirring at 50°C was continued for 2h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was taken up in DCM (30 ml) filtered again and the filtrate was washed with water (30 ml). The phases were separated. The organic phase was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative HPLC to give 80 mg (6%) of 2-[2-(2-benzyl-5-methyl-2H-pyrazol-3- yl)-ethyl]-isoindole-1 ,3-dione. LC-MS: tR = 0.95 min; [M+H]+ = 346.31.
Figure imgf000035_0002
2-[2-(2-benzyl-5-methyl-2H-pyrazol-3-yl)-ethyl]-isoindole-1 ,3-dione (80 mg, 0.233 mmol) was dissolved in ethanol (2 ml) followed by the addition of an 1 M solution of hydrazine in methanol (0.34 ml, 0.34 mmol). The reaction mixture was stirred at 90°C for 16 h, cooled to rt and DCM (3 ml) was added and the suspension was filtered. The filtrate was concentrated under reduced pressure to give 40 mg (80%) 2-(2-benzyl-5-methyl-2H-pyrazol-3-yl)-ethylamine. LC-MS: tR = 0.58 min; [M+H]+ = 216.38.
According to the procedure described above for the preparation of 2-(2-benzyl-5- methyl-2H-pyrazol-3-yl)-ethylamine, the following four derivatives could be prepared:
LC-MS: LC-MS: LC-MS: LC-MS: tR = 0.60 min tR = 0.48 min tR = 0.44 min tR = 0.33 min
[M+H]+ = 230.31 [M+H]+ = 182.34 [M+H]+ = 208.28 [M+H]+ = 140.11
Figure imgf000036_0002
In an inert atmosphere, 3-(4-Thfluoromethyl-phenyl)-propionic acid (10 g, 45.8 mmol) was dissolved in anhydrous THF (250 ml) and cooled to 0°C followed by the addition of a 1 M solution of borane in THF (69 ml, 69 mmol). Stirring was continued at 0°C for 1 h and 16 h at rt followed by slow addition of methanol (100 ml) and water (100 ml). The organic solvents were evaporated under reduced pressure. The remaining water phase was extracted with DCM (3 x 100 ml) and the combined organic layers were washed with brine (100 ml), dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by flash chromatography (silicagel, DCM / methanol = 9/1 to give 9.25 g (99%) of 3-(4-Trifluoromethyl-phenyl)-propan-1-ol. LC-MS: tR = 0.89 min; [M+H]+ = no ionisation.
Figure imgf000037_0001
In an inert atmosphere, 3-(4-trifluoromethyl-phenyl)-propan-1-ol (1.02 g, 5 mmol) was dissolved in DCM (10 ml) and slowly added to a suspension of PCC (1.61 g, 7.5 mmol) in anhydrous DCM (15 ml). Stirring was continued for 4 h at rt. Anhydrous heptane (10 ml) was added to the reaction mixture followed by filtration over a pad of silicagel (15 g). The filtrate was evaporated to give 850 mg of 3-(4-trifluoromethyl-phenyl)-propionaldehyde. LC-MS: tR = 0.94 min; [M+H]+ = no ionisation.
According to the procedure described for the preparation of 3-(4-trifluoromethyl- phenyl)-propionaldehyde, the following substituted 3-phenyl-propionaldehyde derivatives were prepared:
Figure imgf000038_0001
tR = 0.82 tR = 0.95 tR = 0.97
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000038_0002
tR = 0.91 tR = 0.94 tR = 0.88
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000038_0003
tR = 0.92 tR = 0.85 tR = 0.89
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000038_0004
tR = 0.88 tR = 0.84 tR = 0.85
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
= no inoization
Figure imgf000038_0005
Figure imgf000039_0001
In an inert atmosphere, 3-(trifluoromethyl)benzaldehyde (13.26 g, 76.15 mmol) was dissolved in pyridine (58 ml) and malonic acid (15 g, 145 mmol) was added and the mixture was warmed to 50°C followed by slow addition (over 5 minutes) of piperidine (5.8 ml, 58.6 mmol). The resulting suspension was warmed to 75°C and stirring was continued for 3.5 h. The reaction mixture was cooled to 0°C and poured onto an ice-cold solution of concentrated hydrochloric acid (12 M, 32 ml) in water (400 ml). The precipitated product was filtered off and dried at HV to give 14.21 g (68%) of 3-(3-trifluoromethyl-phenyl)-acrylic acid. LC-MS: tR = 0.88 min; [M+H]+ = no ionisation.
Figure imgf000039_0002
In an inert atmosphere, Pd-C (10%, 2.84 g)was suspended in methanol (130 ml) followed by the addition of a solution of 3-(3-trifluoromethyl-phenyl)-acrylic acid (14.21 g, 65.7 mmol). The reaction mixture was put under an atmosphere of hydrogen (1 bar) and vigorously stirred for 2.5 h. The reaction mixture was filtered over a pad of celite and concentrated under reduced pressure to give 12.39 g (86 %) of 3-(3-trifluoromethyl-phenyl)-propionic acid. LC-MS: tR = 0.87 min; [M+H]+ = no ionisation.
Figure imgf000039_0003
3-(3-Trifluoromethyl-phenyl)-propionic acid (12.4 g, 57 mmol) was dissolved in THF (160 ml) in an inert atmosphere and cooled to 0°C followed by slow addition of borane in THF (85 ml, 85 mmol). Stirring was continued at 0°C for 1 h and at rt for 24 h. The mixture was cooled to 0°C and methanol (100 ml) and water (100 ml) was carefully added. The organic solvents were removed under reduced pressure. The remaining aqueous layer was extracted with DCM (3 x 100 ml). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (silicagel, DCM / methanol = 9 / 1 ) to give 10.97 g (94%) of 3-(3-trifluoromethyl-phenyl)-propan-1-ol. LC-MS: tR = 0.87 min; [M+H]+ = no ionisation.
Figure imgf000040_0001
According to the procedure described above for the preparation of 3-(4- trifluoromethyl-phenyl)-propionaldehyde by PCC-oxidation, 132 mg (44%) of 3-(3- trifluoromethyl-phenyl)-propionaldehyde {LC-MS: tR = 0.89 min; [M+H]+ = no ionization} was obtained from 3-(3-thfluoromethyl-phenyl)-propan-1-ol (300 mg, 1.5 mmol).
According to the procedure described for the preparation of 3-(3-trifluoromethyl- phenyl)-propionaldehyde, the following substituted 3-phenyl-propionaldehyde derivatives were prepared:
Figure imgf000041_0001
tR = 0.86 tR = 0.98 tR = 0.93
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0002
tR = 0.88 tR = 0.93 tR = 0.96
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0003
tR = 0.95 tR = 0.94 tR = 0.96
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0004
tR = 0.96 tR = 0.95 tR = 0.87
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0005
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0006
tR = 0.94 tR = 0.93 IR = 1 .01
[M+H]+ = no inoization [M+H]+ = no inoization [M+H]+ = no inoization
Figure imgf000041_0007
= no inoization
Figure imgf000042_0001
To a suspension of sodium hydride (215 mg, 4.92 mmol) in dimethoxyethane (3 ml) was added dropwise a solution of diethyl malonate (868 mg, 5.42 mmol) in dimethoxyethane (2 ml) in an inert atmosphere at rt. Stirring was continued for 1 h followed by the addition of a solution of 2-fluoro-3-methyl-benzylbromide (1 g, 4.92 mmol) in dimethoxyethane (15 ml). The reaction mixture was refluxed for 90 minutes, cooled again to rt and water (5 ml) was carefully added. The diemthoxyethane was removed under reduced pressure, DCM (100 ml) was added and the organic layer was washed with water (100 ml), dried over magenisum sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography (silicagel, DCM / heptane = 4 / 1 ) to give 973 mg (70%) of 2-(2-fluoro-3-methyl-benzyl)-malonic acid diethyl ester. LC-MS: tR = 1.03 min; [M+H]+ = 283.22.
Figure imgf000042_0002
Potassium hydroxide (387 mg, 6.9 mmol) was dissolved in water (8 ml) followed by the addition of 2-(2-fluoro-3-methyl-benzyl)-malonic acid diethyl ester (973 mg, 3.44 mmol). The mixture was refluxed for 5 h followed by removal of the ethanol under reduced pressure. Concentrated sulfuric acid (0.59 ml) was added to the remaining aqueous solution and refluxing was continued for 18 h. The reaction mixture was cooled to 0°C and the precipitated product was filtered off, washed with water and dried at HV to give 433 mg (69%) of 3-(2-fluoro-3-methyl-phenyl)- propionic acid. LC-MS: tR = 0.84 min; [M+H]+ = 183.20.
Figure imgf000043_0001
According to procedures described above, 3-(2-fluoro-3-methyl-phenyl)-propionic acid (2.39 g, 13.12 mmol) was reduced by borane in THF to give 1.82 g (82.5%) of 3-(2-fluoro-3-methyl-phenyl)-propan-1-ol. LC-MS: tR = 0.86 min; [M+H]+ = no ionization.
Figure imgf000043_0002
According to procedures described above, 3-(2-fluoro-3-methyl-phenyl)-propan- 1-ol (300 mg, 1.78 mmol) was oxidized by PCC in DCM to give 178 mg (60%) of 3-(2-fluoro-3-methyl-phenyl)-propionaldehyde. LC-MS: tR = 0.86 min; [M+H]+ = no ionization.
According to the procedure described for the preparation of 3-(2-fluoro-3-methyl- phenyl)-propionaldehyde, the following substituted 3-phenyl-propionaldehyde derivatives were prepared:
Figure imgf000043_0003
LC-MS: LC-MS: tR = 0.95 IR = 1 .02
[M+H]+ = no ionization [M+H]+ = no ionization Preparation of final compounds:
Example 1 : 2-{1 -Ethyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyndin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000044_0001
2-(2-Ethyl-5-methyl-2H-pyrazol-3-yl)-ethylamine (148 mg, 0.97 mmol) was dissolved in ethanol (3 ml) and acetic acid (116.5 mg. 1.94 mmol) was added followed by the addition of 3-(4-trifluoromethyl-phenyl)-propionaldehyde (197 mg, 0.97 mmol). The mixture was sealed and put in the microwave oven (100 Watts; 130°C, 14 bar, 6 minutes). The solvents were removed under reduced pressure and the residue was purified by prep. HPLC to give 113 mg (34.5%) of 1 -ethyl-3- methyl-4-[2-(4-thfluoromethyl-phenyl)-ethyl]-4,5,6,7-tetrahydro-1 H-pyrazolo[4,3- φyridine. LC-MS: tR = 0.75 min; [M+H]+ = 338.22.
Figure imgf000044_0002
In an inert atmosphere, 1-ethyl-3-methyl-4-[2-(4-thfluoromethyl-phenyl)-ethyl]- 4,5,6,7-tetrahydro-1 H-pyrazolo[4,3-c]pyridine (51 mg, 0.15 mmol) was dissolved in 3-methyl-2-butyl-ketone (MIBK) (1 ml) followed by the addition of DIPEA (21.3 mg, 0.165 mmol) and toluene-4-sulfonic acid methylcarbamoyl-phenyl-methyl ester (71.8 mg, 0.225 mmol). The reaction mixture was heated to 90°C for 24 h, concentrated in vacuo and the crude product was purified by prep. HPLC to give 70 mg (95%) of 2-{1-ethyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide. LC-MS: tR = 0.84 min; [M+H]+ = 485.28.
According to the procedure described for the preparation of example 1 the following examples 2 to 9 could be prepared:
Example 2: 2-{1-Ethyl-4-[2-(4-ethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide:
Figure imgf000045_0001
22 5 mg yield Example 2
153 mg 162 mg 0 076 mmol 2 mg yield 1 mmol 1 mmol tR = 0 75 min 0 0045 mmol [M+H]+ = 298 21 tR = 0 85 min [M+H]+ = 445 26
Example 3: 2-{1 -Ethyl-4-[2-(2-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000046_0001
884 mg yield Example 3
153 mg 220 mg 0 25 mmol 27 mg yield 1 mmol 1 mmol tR = 0 76 min O 053 mmol [M+H]+ = 356 22 tR = 0 85 min [M+H]+ = 503 22
Example 4: 2-{4-[2-(2,3-Difluoro-4-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyndin-S-ylJ-N-methyl^-phenyl-acetamide :
Figure imgf000046_0002
87 9 mg yield Example 4
153 mg 184 mg 0 28 mmol 16 mg yield 1 mmol 1 mmol tR = 0 74 min 0 084 mmol [M+H]+ = 320 17 tR = 0 84 min [M+H]+ = 467 26
Example 5: 2-{1 -Ethyl-3-methyl-4-[2-(2,3,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000047_0001
77 5 mg yield Example 5
153 mg 188 mg O 24 mmol 50 mg yield 1 mmol 1 mmol tR = 0 72 min O 106 mmol [M+H]+ = 324 42 tR = 0 82 min [M+H]+ = 471 27
Example 6: 2-[1-Ethyl-3-methyl-4-(2-p-tolyl-ethyl)-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide:
Figure imgf000047_0002
10 mg yield Example 6
153 mg 149 mg 0 035 mmol 14 mg yield 1 mmol 1 mmol tR = 0 71 min 0 033 mmol [M+H]+ = 284 27 tR = 0 81 min [M+H]+ = 431 32
Example 7: 2-{1 -Ethyl-4-[2-(4-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000048_0001
29 mg yield Example 7
153 mg 166 mg O 096 mmol 29 2 mg yield 1 mmol 1 mmol tR = 0 73 min 0 065 mmol [M+H]+ = 302 21 tR = 0 82 min [M+H]+ = 449 27
Example 8: 2-{1 -Ethyl-3-methyl-4-[2-(3-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyndin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000048_0002
17 3 mg yield Example 8
153 mg 202 mg 0 051 mmol 16 2 mg yield 1 mmol 1 mmol tR = 0 75 min 0 033 mmol [M+H]+ = 338 22 tR = 0 84 min [M+H]+ = 485 28
Example 9: 2-[4-(2-Benzo[1 ,3]dioxol-5-yl-ethyl)-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo[4,3-c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide:
Figure imgf000049_0001
88 6 mg yield Example 9
153 mg 178 mg O 28 mmol 40 mg yield 1 mmol 1 mmol tR = O 68 min O 086 mmol [M+H]+ = 314 142 tR = O 78mιn [M+H]+ = 461 24
According to the sequences described above, the following examples were prepared:
Example 10: 2-{1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide:
Figure imgf000049_0002
Example 11 : 2-{1-Ethyl-4-[2-(4-isopropyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide:
Figure imgf000050_0001
Example 12: 2-{1-Ethyl-4-[2-(3-fluoro-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo^S-cJpyridin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000050_0002
Example 13: 2-{1 -Ethyl-4-[2-(2-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000050_0003
Example 14: 2-{4-[2-(2,3-Difluoro-phenyl)-ethyl]-1-ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000051_0001
Example 15: 2-{1 -Ethyl-4-[2-(2-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000051_0002
Example 16: 2-{4-[2-(4-Chloro-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide:
Figure imgf000051_0003
Example 17: 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Example 27: 2-{1 -Benzyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000055_0002
Example 28: 2-{1 -Ethyl-4-[2-(2-methoxy-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000055_0003
Example 29: N-Methyl-2-{3-methyl-1 -phenethyl-4-[2-(4-trifluoromethyl-phenyl)- ethyl]-1 ,4,6,7-tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-2-phenyl-acetamide:
Figure imgf000056_0001
Example 32: 2-{1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000057_0001
Example 33: 2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000057_0002
Example 34: 2-{1 -Ethyl-4-[2-(2-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
.43
Figure imgf000057_0003
Example 35: 2-{4-[2-(2,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo[4,3-c] pyridin-5-n-- yll}-N-methyl-2-phenyl-acetamide:
Figure imgf000058_0001
Example 38: 2-{1 -Ethyl-3-methyl-4-[2-(2,3,4-trifluoro-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo4,3c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide:
Example 38 51 mg yield tR = 0.83min [M+H]+ = 471.43
Figure imgf000059_0003
Example 39: 2-{1 -Ethyl-3-methyl-4-[2-(2,3,4-trifluoro-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo[4,3- c]pyridin-5- yl-}N-methyl-2-phenyl-acetamide:
Example 39 24mg yield tR = 0.86min [M+H]+ = 503.38
Figure imgf000059_0002
Example 40: 2-{1 -Ethyl-3-methyl-4-[2-(2-methyl-5-tπfluoromethyl-phenyl)-ethyl]- 1,4,6,7-tetrahydro-pyrazolo[4,3-, c]pyridin-5-yl -}N-methyl-2-phenyl-acetamide:
Example 40 70mg yield tR = O.δOmin [M+H]+ = 499.43
Figure imgf000059_0001
Example 41 : 2-{1-Ethyl-4-[2-(3-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000060_0001
Example 42: 2-{4-[2-(3,5-Bis-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyπdin-S-ylJ-N-methyl^-phenyl-acetamide:
Figure imgf000060_0002
Example 43: 2-{4-[2-(3,5-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3- methyl-1 ,4,6,7-tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl- acetamide:
.24
Figure imgf000060_0003
Example 44: 2-{4-[2-(3,5-Difluoro-4-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I,4,6,7-tetrahydro-pyrazolo[4,3-- c]pyridin -5-yl}-N-methyl-2-phenyl-acetamide: -
Figure imgf000061_0001
Example 47: 2-{4-[2-(2,6-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3- methyl-I AβJ-tetrahydro-pyrazolo^S-clpyridin-S-ylJ-N-methyl^-phenyl- acetamide:
Figure imgf000062_0001
Biological assays
In vitro assay
The orexin receptor antagonistic activity of the compounds of formula (I) and/or (Ia) 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 OXi and OX2 receptors. Selected compounds are displayed in Table 1. Table 1:
Figure imgf000064_0001

Claims

Claims
1. Compounds of the general formula I
General Formula I
Figure imgf000065_0001
wherein Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of
Figure imgf000065_0002
halogen and trifluoromethyl; 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 optically pure enantiomers, mixtures of enantiomers such as racemates, diastereomers, mixtures of diastereomers, diastereomehc racemates, mixtures of diastereomeric racemates, or meso-forms and pharmaceutically acceptable salts thereof.
2. Compounds of the general formula Ia according to claim 1 with the chirality as depicted below
R-chirality
General Formula Ia
Figure imgf000065_0003
S-chirality
3. Compounds according to anyone of claims 1 to 2, wherein
Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of
Figure imgf000066_0001
halogen and trifluoromethyl; R2 represents (Ci-4)alkyl; R3 represents (Ci-4)alkyl; R4 represents a phenyl group; R5 represents (Ci-4)alkyl.
4. Compounds according to anyone of claims 1 to 3, wherein
Y represents -CH2-CH2-;
R1 represents 1 ,3-benzodioxole or a phenyl group, wherein the phenyl group can be mono-, di-, or th-substituted, wherein the substituents are independently selected from the group consisting of methyl, ethyl, fluorine, chlorine and trifluoromethyl;
R2 represents methyl;
R3 represents ethyl; R4 represents a phenyl group;
R5 represents methyl.
5. Compounds according to any one of claims 1 to 4 selected from the group consisting of: 2-{1 -Ethyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1 -Ethyl-4-[2-(4-ethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1 -Ethyl-4-[2-(2-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
Figure imgf000067_0001
2-{1 ,3-Dimethyl-4-[2-(4-tπfluoromethyl-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-3-methyl-4-[2-(4-methyl-3-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; 2-[1 -Ethyl-3-methyl-4-(2-m-tolyl-ethyl)-1 ,4,6,7-tetrahydro-pyrazolo[4,3- c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide;
2-[1-Ethyl-3-methyl-4-(2-o-tolyl-ethyl)-1 ,4,6,7-tetrahydro-pyrazolo[4,3- c]pyridin-5-yl]-N-methyl-2-phenyl-acetamide;
2-{1-Butyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1-Ethyl-4-[2-(3-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(2-fluoro-3-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Benzyl-3-methyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-methoxy-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
N-Methyl-2-{3-methyl-1-phenethyl-4-[2-(4-trifluoromethyl-phenyl)-ethyl]- 1 ,4,6,7-tetrahydro-pyrazolo[4,3-c]pyridin-5-yl}-2-phenyl-acetamide;
N-Methyl-2-{3-methyl-1-(2,2,2-trifluoro-ethyl)-4-[2-(4-tnfluoromethyl- phenyO-ethyO-I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-yl^-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(2-fluoro-3-methyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,4-Dimethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{4-[2-(2,4-Difluoro-3-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1 -Ethyl-3-methyl-4-[2-(2,4,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1 -Ethyl-3-methyl-4-[2-(2,4,5-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyndin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1 -Ethyl-3-methyl-4-[2-(2,3,4-trifluoro-phenyl)-ethyl]-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{1 -Ethyl-3-methyl-4-[2-(2-methyl-5-trifluoromethyl-phenyl)-ethyl]-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(3-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; and
2-{4-[2-(3,5-Bis-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-methyl-phenyl)-ethyl]-1 -ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,3-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide; 2-{4-[2-(3,4-Dichloro-phenyl)-ethyl]-1 -ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyridin-5-yl}-N-methyl-2-phenyl-acetamide;
2-{4-[2-(2,6-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl- I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
6. Compounds according to any one of claims 1 to 5 selected from the group consisting of:
2-{4-[2-(3,4-Dimethyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{1 -Ethyl-4-[2-(3-fluoro-4-methyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-4-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{1-Ethyl-4-[2-(2-fluoro-5-trifluoromethyl-phenyl)-ethyl]-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(2,3-Difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 ,4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide; and
2-{1-Ethyl-4-[2-(4-ethyl-phenyl)-ethyl]-3-methyl-1 ,4,6,7-tetrahydro- pyrazolo[4,3-c]pyhdin-5-yl}-N-methyl-2-phenyl-acetamide; 2-{4-[2-(3,5-Difluoro-4-trifluoromethyl-phenyl)-ethyl]-1 -ethyl-3-methyl-
I ^^J-tetrahydro-pyrazolo^^-clpyridin-S-ylJ-N-methyl^-phenyl-acetamide;
2-{4-[2-(3,5-Difluoro-4-methyl-phenyl)-ethyl]-1-ethyl-3-methyl-1 , 4,6,7- tetrahydro-pyrazolo^^-clpyhdin-S-ylJ-N-methyl^-phenyl-acetamide;
7. Compounds according to anyone of claims 1 to 6 for use as medicaments.
8. Use of a compound according to any of claims 1 to 6 for the preparation of a medicament for the prevention or treatment of diseases selected from the group consisting of dysthymic, mood, psychotic and anxiety disorders; diabetes and appetite, taste, eating, or drinking disorders; hypothalamic diseases; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; insomnias related to psychiatric disorders; sleep apnea; narcolepsy; idiopathic insomnias; parasomnias; benign prostatic hypertrophy; all dementias and cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders; and other diseases related to general orexin system dysfunctions.
PCT/IB2007/051522 2006-04-26 2007-04-25 Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists WO2007122591A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AT07735643T ATE496051T1 (en) 2006-04-26 2007-04-25 PYRAZOLOTETRAHYDROPYRIDINE DERIVATIVES AS OREXIN RECEPTOR ANTAGONISTS
CA002647077A CA2647077A1 (en) 2006-04-26 2007-04-25 Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists
DE602007012072T DE602007012072D1 (en) 2006-04-26 2007-04-25 ORANTAGONISTEN
JP2009507230A JP2009535324A (en) 2006-04-26 2007-04-25 Novel pyrazolo-tetrahydropyridine derivatives
EP07735643A EP2013209B1 (en) 2006-04-26 2007-04-25 Pyrazolo-tetrahydropyridine derivatives as orexin receptor antagonists
US12/298,369 US7834028B2 (en) 2006-04-26 2007-04-25 Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IB2006051300 2006-04-26
IBPCT/IB2006/051300 2006-04-26

Publications (2)

Publication Number Publication Date
WO2007122591A2 true WO2007122591A2 (en) 2007-11-01
WO2007122591A3 WO2007122591A3 (en) 2008-01-10

Family

ID=38541955

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2007/051522 WO2007122591A2 (en) 2006-04-26 2007-04-25 Pyrazolo-tetrahydro pyridine derivatives as orexin receptor antagonists

Country Status (8)

Country Link
US (1) US7834028B2 (en)
EP (1) EP2013209B1 (en)
JP (1) JP2009535324A (en)
CN (1) CN101432285A (en)
AT (1) ATE496051T1 (en)
CA (1) CA2647077A1 (en)
DE (1) DE602007012072D1 (en)
WO (1) WO2007122591A2 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008122513A1 (en) * 2007-04-04 2008-10-16 F. Hoffmann-La Roche Ag Heterocycles as orexin 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
WO2009104155A1 (en) * 2008-02-21 2009-08-27 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[2.2.1]heptane derivatives
US7763638B2 (en) 2004-03-01 2010-07-27 Actelion Pharmaceuticals Ltd. Substituted 1,2,3,4-tetrahydroisoquinoline derivatives
WO2011076744A1 (en) * 2009-12-21 2011-06-30 Novartis Ag Disubstituted heteroaryl-fused pyridines
US8188082B2 (en) 2006-12-22 2012-05-29 Actelion Pharmaceuticals Ltd. 5,6,7,8-tetrahydro-imidazo[1,5-α]pyrazine derivatives
US8247560B2 (en) 2007-12-28 2012-08-21 Actelion Pharmaceuticals Ltd. Trisubstituted 3,4-dihydro-1H-isoquinolin compound, process for its preparation, and its use
WO2012114252A1 (en) 2011-02-21 2012-08-30 Actelion Pharmaceuticals Ltd Novel indole and pyrrolopyridine amides
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
US9156819B2 (en) 2011-10-19 2015-10-13 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
WO2016074696A1 (en) * 2014-11-10 2016-05-19 Givaudan Ag Perfume ingredients and perfume preparations containing same
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 (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101511833A (en) * 2006-08-28 2009-08-19 埃科特莱茵药品有限公司 1,4,5,6,7,8-hexahydro-1,2,5-triaza-azulene derivatives as orexin receptor antagonists
ES2367341T3 (en) * 2008-02-12 2011-11-02 F. Hoffmann-La Roche Ag PIPERIDINE-SULFONAMIDE DERIVATIVES.
EP2508511A1 (en) 2011-04-07 2012-10-10 Laboratoire Biodim Inhibitors of viral replication, their process of preparation and their therapeutical uses
GB201308236D0 (en) 2013-05-08 2013-06-12 Givaudan Sa Improvements in or relating to organic compounds
WO2014205782A1 (en) 2013-06-28 2014-12-31 Givaudan Sa Organic compounds
MX2017005862A (en) 2014-11-10 2017-06-26 Givaudan Sa Improvements in or relating to organic compounds.
CN111494376A (en) * 2020-06-15 2020-08-07 牡丹江医学院 Medicine for treating gingivitis and preparation method thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE60108236T2 (en) 2000-03-14 2005-12-22 Actelion Pharmaceuticals Ltd. 1,2,3,4-tetrahydroisoquinoline DERIVATIVES
CA2408343A1 (en) * 2000-05-11 2002-11-07 Banyu Pharmaceutical Co., Ltd. N-acyltetrahydroisoquinoline derivatives
WO2002051232A2 (en) * 2000-12-27 2002-07-04 Actelion Pharmaceuticals Ltd. Novel benzazepines and related heterocyclic derivatives
KR100681585B1 (en) 2002-04-26 2007-02-09 에프. 호프만-라 로슈 아게 Isoquinoline derivatives
US6709392B1 (en) * 2002-10-10 2004-03-23 Koninklijke Philips Electronics N.V. Imaging ultrasound transducer temperature control system and method using feedback
RU2345985C2 (en) * 2003-03-26 2009-02-10 Актелион Фармасьютиклз Лтд. Derivatives of tetrahydroisoquinolylacetamide as orexin receptor antagonists
CA2557163C (en) 2004-03-01 2011-08-16 Actelion Pharmaceuticals Ltd Substituted 1,2,3,4-tetrahydroisoquinoline derivatives
FR2874011B1 (en) 2004-08-03 2007-06-15 Sanofi Synthelabo SULFONAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
KR101065239B1 (en) * 2006-03-15 2011-09-16 액테리온 파마슈티칼 리미티드 Tetrahydroisoquinoline derivatives to enhance memory function

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7763638B2 (en) 2004-03-01 2010-07-27 Actelion Pharmaceuticals Ltd. Substituted 1,2,3,4-tetrahydroisoquinoline derivatives
US8188082B2 (en) 2006-12-22 2012-05-29 Actelion Pharmaceuticals Ltd. 5,6,7,8-tetrahydro-imidazo[1,5-α]pyrazine derivatives
US8067598B2 (en) 2007-04-04 2011-11-29 Hoffman-La Roche Inc. Heterofused piperidines as orexin antagonists
JP2010523518A (en) * 2007-04-04 2010-07-15 エフ.ホフマン−ラ ロシュ アーゲー Heterocycles as orexin antagonists
WO2008122513A1 (en) * 2007-04-04 2008-10-16 F. Hoffmann-La Roche Ag Heterocycles as orexin 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
US8242121B2 (en) 2007-05-23 2012-08-14 Merck Sharp & Dohme Corp. 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
US8247560B2 (en) 2007-12-28 2012-08-21 Actelion Pharmaceuticals Ltd. Trisubstituted 3,4-dihydro-1H-isoquinolin compound, process for its preparation, and its use
WO2009104155A1 (en) * 2008-02-21 2009-08-27 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[2.2.1]heptane derivatives
CN101965343A (en) * 2008-02-21 2011-02-02 埃科特莱茵药品有限公司 2-aza-bicyclo[2.2.1]heptane derivatives
US8236801B2 (en) 2008-02-21 2012-08-07 Actelion Pharmaceuticals Ltd. 2-aza-bicyclo[2.2.1]heptane derivatives
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
CN102762560A (en) * 2009-12-21 2012-10-31 诺瓦提斯公司 Disubstituted heteroaryl-fused pyridines
US8742106B2 (en) 2009-12-21 2014-06-03 Novartis Ag Disubstituted heteroaryl-fused pyridines
WO2011076744A1 (en) * 2009-12-21 2011-06-30 Novartis Ag Disubstituted heteroaryl-fused pyridines
WO2012114252A1 (en) 2011-02-21 2012-08-30 Actelion Pharmaceuticals Ltd Novel indole and pyrrolopyridine amides
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
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
US9896452B2 (en) 2012-02-07 2018-02-20 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
WO2016074696A1 (en) * 2014-11-10 2016-05-19 Givaudan Ag Perfume ingredients and perfume preparations containing same
US11434236B2 (en) 2016-02-12 2022-09-06 Astrazeneca Ab Halo-substituted piperidines as orexin receptor modulators
US10894789B2 (en) 2016-02-12 2021-01-19 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
US7834028B2 (en) 2010-11-16
CN101432285A (en) 2009-05-13
CA2647077A1 (en) 2007-11-01
JP2009535324A (en) 2009-10-01
EP2013209A2 (en) 2009-01-14
DE602007012072D1 (en) 2011-03-03
ATE496051T1 (en) 2011-02-15
WO2007122591A3 (en) 2008-01-10
EP2013209B1 (en) 2011-01-19
US20090099228A1 (en) 2009-04-16

Similar Documents

Publication Publication Date Title
EP2013209B1 (en) Pyrazolo-tetrahydropyridine derivatives as orexin receptor antagonists
EP2059520B1 (en) 1,4,5,6,7,8-hexahydro-i,2,5-triaza-azulene 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
US8133901B2 (en) 3-heteroaryl (amino or amido)-1-(biphenyl or phenylthiazolyl) carbonylpiperidine derivatives as orexin receptor inhibitors
EP2318367B1 (en) Piperidine and pyrrolidine compounds
US20110086889A1 (en) Tetrazole compounds as orexin receptor antagonists
JP2010540429A (en) Pyrrolidines and piperidines as orexin receptor antagonists
WO2008087611A2 (en) Pyrrolidine- and piperidine- bis-amide derivatives
NZ581986A (en) Quinoline compounds suitable for treating disorders that respond to modulation of the serotonin 5-HT6 receptor
CA2726834A1 (en) 5,6,7,8-tetrahydro-imidazo[1,5-a]pyrazine compounds
CA3120037A1 (en) Heteroaromatic compounds as vanin inhibitors
CN103298811A (en) Diaza-spiro[5.5]undecanes useful as orexin receptor antagonists
TW202304872A (en) Taar1 and serotonin modulators, and pharmaceutical compositions, and methods of use thereof

Legal Events

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

Ref document number: 2007735643

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2647077

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009507230

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 12298369

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 200780015202.1

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE