WO2007126935A2 - Diazepan orexin receptor antagonists - Google Patents

Diazepan orexin receptor antagonists Download PDF

Info

Publication number
WO2007126935A2
WO2007126935A2 PCT/US2007/007738 US2007007738W WO2007126935A2 WO 2007126935 A2 WO2007126935 A2 WO 2007126935A2 US 2007007738 W US2007007738 W US 2007007738W WO 2007126935 A2 WO2007126935 A2 WO 2007126935A2
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
phenyl
compound
substituents selected
Prior art date
Application number
PCT/US2007/007738
Other languages
French (fr)
Other versions
WO2007126935A3 (en
Inventor
Karen M. Brashear
Paul J. Coleman
Christopher D. Cox
Anthony M. Smith
David B. Whitman
Original Assignee
Merck & Co., Inc.
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 Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CA002647678A priority Critical patent/CA2647678A1/en
Priority to AU2007245037A priority patent/AU2007245037A1/en
Priority to US12/225,506 priority patent/US8685961B2/en
Priority to EP07754286.8A priority patent/EP2001485B1/en
Priority to JP2009502989A priority patent/JP2009531447A/en
Publication of WO2007126935A2 publication Critical patent/WO2007126935A2/en
Publication of WO2007126935A3 publication Critical patent/WO2007126935A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Anesthesiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Child & Adolescent Psychology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention is directed to diazepan compounds which are antagonists of orexin receptors, and which are useful in the treatment or prevention of neurological and psychiatric disorders and diseases in which orexin receptors are involved. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.

Description

TITLE OF THE INVENTION
DIAZEPAN OREXIN RECEPTOR ANTAGONISTS
BACKGROUND OF THE INVENTION
The orexins (hypocretins) comprise two neuropeptides produced in the hypothalamus: the orexin A (OX-A) (a 33 amino acid peptide) and the orexin B (OX-B) (a 28 amino acid peptide) (Sakurai T. et al., Cell, 1998, 92, 573-585). 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). Orexins also regulate states of sleep and wakefulness opening potentially novel therapeutic approaches for narcoleptic or insomniac patients (Chemelli R.M. et al., Cell, 1999, 98, 437-451). Two orexin receptors have been cloned and characterized in mammals. They belong to the super family of G-protein coupled receptors (Sakurai T. et al., Cell, 1998, 92, 573- 585): the orexin- 1 receptor (OX or OXlR) is selective for OX-A and the orexin-2 receptor (OX2 or OX2R) is capable to bind OX-A as well as OX-B. The physiological actions in which orexins are presumed to participate are thought to be expressed via one or both of OX 1 receptor and OX 2 receptor as the two subtypes of orexin receptors.
Orexin receptors are found in the mammalian brain and may have numerous implications in pathologies such as depression; anxiety; addictions; obsessive compulsive disorder; affective neurosis; depressive neurosis; anxiety neurosis; dysthymic disorder; behaviour disorder; mood disorder; sexual dysfunction; psychosexual dysfunction; sex disorder; schizophrenia; manic depression; delirium; dementia; severe mental retardation and dyskinesias such as Huntington's disease and Tourette syndrome; eating disorders such as anorexia, bulimia, cachexia, and obesity; cardiovascular diseases; diabetes; appetite/taste disorders; emesis, vomiting, nausea; asthma; cancer; Parkinson's disease; Cushing's syndrome/disease; basophile adenoma; prolactinoma; hyperprolactinemia; hypophysis tumour/adenoma; hypothalamic diseases; inflammatory bowel disease; gastric diskinesia; gastric ulcers; Froehlich's syndrome; adrenohypophysis disease; hypophysis disease; adrenohypophysis hypofunction; adrenohypophysis hyperfunction; hypothalamic hypogonadism; Kallman's syndrome (anosmia, hyposmia); functional or psychogenic amenorrhea; hypopituitarism; hypothalamic hypothyroidism; hypothalamic- adrenal dysfunction; idiopathic hyperprolactinemia; hypothalamic disorders of growth hormone deficiency; idiopathic growth deficiency; dwarfism; gigantism; acromegaly; disturbed biological and circadian rhythms; sleep disturbances associated with diseases such as neurological disorders, neuropathic pain and restless leg syndrome; heart and lung diseases, acute and congestive heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectoris; myocardinal infarction; ischemic or haemorrhagic stroke; subarachnoid haemorrhage; ulcers; allergies; benign prostatic hypertrophy; chronic renal failure; renal disease; impaired glucose tolerance; migraine; hyperalgesia; pain; 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; pain related to infection e.g. HTV, post-chemotherapy pain; post-stroke pain; post-operative pain; neuralgia; emesis, nausea, vomiting; conditions associated with visceral pain such as irritable bowel syndrome, and angina; migraine; urinary bladder incontinence e.g. urge incontinence; tolerance to narcotics or withdrawal from narcotics; sleep disorders; sleep apnea; narcolepsy; insomnia; parasomnia; jet lag syndrome; and neurodegenerative disorders including nosological entities such as disinhibϊtion-dementia- parkinsonism-amyotrophy complex; pallido-ponto-nigral degeneration; epilepsy; seizure disorders and other diseases related to general orexin system dysfunction.
Certain orexin receptor antagonists are disclosed in PCT patent publications WO 99/09024, WO 99/58533, WO 00/47576, WO 00/47577, WO 00/47580, WO 01/68609, WO 01/85693, WO 2002/051232, WO 2002/051838, WO 2003/002559, WO 2003/002561, WO 2003/032991, WO 2003/037847, WO 2003/041711, WO 2003/051872, WO 2003/051873, WO 2004/004733, WO 2004/033418, WO 2004/083218, WO 2004/085403, WO 2005/060959, WO2005/118548. 2-Amino-methylpiperidine derivatives (WO 01/96302), 3-aminomethyl morpholine derivatives (WO 02/44172) and N-aroyl cyclic amines (WO 02/090355, WO 02/089800 and WO 03/051368) are disclosed as orexin receptor antagonists.
SUMMARY OF THE INVENTION
The present invention is directed to diazepan compounds which are antagonists of orexin receptors, and which are useful in the treatment or prevention of neurological and psychiatric disorders and diseases in which orexin receptors are involved. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to compounds of the formula I:
Figure imgf000004_0001
"I wherein: X is selected from -SO2-, -(C=O)-, and -(C=S)-;
Rl is selected from the group consisting of:
(1) phenyl, where the phenyl is substituted with Rl a, Rib and Rlc, and
(2) napthyl, where the napthyl is substituted with Rl a, Rl b and Rl <=;
(3) heteroaryl, where the heteroaryl is substituted with Rla, Rib and Rlc;
R2 is heteroaryl, where the heteroaryl is substituted with R2a5 R2b and R2c;
RIa5 Rlb} RIc5 R2a5 R2b and R2c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -(C=O)1n-On-Cl -6alkyl, where m is O or 1, n is O or 1 (wherein if m is O or n is O, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) -(C=O)rn-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(8) -(C=0)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl 3,
(10) -(C=O)1n-NRl ORI 11 wherein Rl O and Rl 1 are independently selected from the group consisting of: (a) hydrogen, (b) Cl _6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
(c) C3-6alkenyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
(d) cycloalkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
(e) phenyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3, and
(f) heterocycle, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
Figure imgf000005_0001
(12) -S(O)q-Rl2, where q is 0, 1 or 2 and where Rl2 is selected from the definitions of
RlO and Rl I,
(13) -CO2H5
(14) -CN, and
(15) -NO2;
R4 are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -Ci_6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) -O-Cl_6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(6) -phenyl or -napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(7) -NRl ORI 1 , wherein Rl 0 and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Ci_6alkyl. which is unsubstituted or substituted with one or more substituents selected from Rl 3, or R3 and R4 taken together form C=O; Rl 3 is selected from the group consisting of:
(1) halogen,
(2) hydroxyl,
(3) -(C=O)1n-On-C l-6alkyl. where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 4,
(4) -On-(C i-3)perfluoroalkyl,
(5) -(C=0)m-On-C3_6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from R 14,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from Rl 4,
(7) -(C=O)m-On-phenyl or -(C=O)m-On-napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl 4, (S) -(C=O)m-Oπ-heterocycle5 where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl 4,
(9) -(C=O)1n-NRiWRI i,
(10) -S(O)2-NRl0Rl l.
(H) -S(O)q-Rl2,
(12) -CO2H,
(13) -CN, and
(14) -NO2;
Rl 4 is selected from the group consisting of:
(1) hydroxyl,
(2) halogen,
(3) Ci-6alkyl,
(4) -C3-6cycloalkyl,
(5) -O-Ci_6alkyl,
(6) -0(C=O)-Ci -6alkyl,
(7) -NH-Ci-6alkyl,
(8) phenyl,
(9) heterocycle,
(10) -CO2H, and
(H) -CN; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ia:
Figure imgf000007_0001
Ia wherein Rl, R.2, R3 and R4 are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ib:
Ib wherein Rl, R2, R3 and R4 are defined herein; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention includes compounds of the formula Ic:
Figure imgf000007_0003
Ic wherein Rl and R2 are defined herein; or a pharmaceutically acceptable salt thereof.
An embodiment of the present invention includes compounds wherein X is -SO2-.
An embodiment of the present invention includes compounds wherein X is -(C=O)-.
An embodiment of the present invention includes compounds wherein X is -(C=S)-.
An embodiment of the present invention includes compounds wherein Rl is phenyl, which is unsubstituted or substituted with one or more of: (1) halogen, (2) hydroxyl,
(3) -On-Cl_6alkyl, where n is 0 or 1 (wherein if n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(4) -On-phenyl, where the phenyl is unsubstituted or substituted with one or more substiruents selected from Rl 3,
(5) -heterocycle, where the heterocycle is unsubstituted or substituted with one or more substiruents selected from Rl 3,
(6) -NRl ORI 1 , wherein Rl 0 and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Cl-6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
Figure imgf000008_0001
(8) -CO2H5
(9) -CN, and
(10) -NO2-
An embodiment of the present invention includes compounds wherein
Rl is phenyl, which is unsubstituted or substituted with one or more methyl, -CF3, halo, -OCF3, -OCH3, -OCH2CH3, -CO2CH3, -CN, -N(CH3), -NH(CH2CH3), -NO2, triazolyl or phenyl:
An embodiment of the present invention includes compounds wherein
Rl is phenyl, which is unsubstituted or substituted with one or more methyl, -CF3, chloro, fiuro, -OCF3, -OCH3, -OCH2CH3, -CO2CH3, triazolyl or phenyl.
An embodiment of the present invention includes compounds wherein Rl is phenyl, which is unsubstituted or substituted with one or more methyl, -CF3, fluro, -OCF3, -OCH3, -CO2CH3 or phenyl.
An embodiment of the present invention includes compounds wherein Rl is selected from the group consisting of:
(1) phenyl,
(2) biphenyl,
(3) 2,6-dimethoxyphenyl,
(4) 2,4-dichlorophenyl,
(5) 2,6-dichlorophenyl,
(6) 2,3-difluophenyl,
(7) 2,4-difluophenyl, (8) 2,6-difluophenyl,
(9) 2-methoxy-4-methyl-phenyl,
(10) 3 -methoxy-bipheny 1,
(11) 3 -methyl-biphenyl, and
(12) 5-methyl-2-triazolyl-phenyl.
An embodiment of the present invention includes compounds wherein Rl is phenyl.
An embodiment of the present invention includes compounds wherein Rl is 2,6-dimethoxyphenyl.
An embodiment of the present invention includes compounds wherein R2 is heteroaryl, which is unsubstituted or substituted with one or more of:
(1) halogen,
(2) hydroxyl,
(3) -On-Ci_6alkyl, where n is 0 or 1 (wherein if n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected
Figure imgf000009_0001
(4) -On-phenyl, where the phenyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) -heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl 3,
(6) -NRl ORI 1. wherein Rl 0 and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Ci_6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
Figure imgf000009_0002
(8) -CO2H,
(9) -CN, and
(10) -NO2.
An embodiment of the present invention includes compounds wherein
R2 is heteroaryl, which is unsubstituted or substituted with halogen, hydroxyl, Ci_6alkyl, -O-Ci. 6alkyl or phenyl.
An embodiment of the present invention includes compounds wherein R2 is selected from the group consisting of: (1) benzimidazolyl, (2) benzothiazolyl,
(3) benzoxazolyl,
(4) quinazolinyl,
(5) quinolinyl,
(6) thiadiazolyl, which is unsubstituted or substituted with halogen, hydroxyl, Ci_6alkyl, -O-Ci-βalkyl or phenyl.
An embodiment of the present invention includes compounds wherein R2 is selected from the group consisting of:
(1) benzimidazol-2-yl,
(2) l,3-benzothiazol-2-yl,
(3) l,3-benzoxazol-2-yl,
(4) 2-quinazolinyl5
(5) 1 -quinolin-2-yl,
(6) l,2,4-thiadiazol-5-yl, which is unsubstituted or substituted with methyl, fluoro, chloro or phenyl.
An embodiment of the present invention includes compounds wherein R2 is benzothiazolyl, which is unsubstituted or substituted with chloro.
An embodiment of the present invention includes compounds wherein R2 is 6-chloro-benzothiazolyl.
An embodiment of the present invention includes compounds wherein R2 is benzoxazolyl.
An embodiment of the present invention includes compounds wherein R2 is quinazolinyl or quinolinyl.
An embodiment of the present invention includes compounds wherein R3 and R4 are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -Ci_6alkyl,
(5) -O-Ci-6alkyl,
(6) -phenyl,
(7) -NRl ORI 1. wherein RlO and Rl 1 are independently selected from the group consisting of:
(a) hydrogen, (b) Ci-6alkyl, or R3 and R4 taken together form C=O;
An embodiment of the present invention includes compounds wherein R3 and R4 are hydrogen.
An embodiment of the present invention includes compounds wherein R3 is fluoro and R4 is hydrogen.
An embodiment of the present invention includes compounds wherein R3 is hydroxyl and R4 is hydrogen.
An embodiment of the present invention includes compounds wherein R3 is -NH-Ci_6alkyl and R4 is hydrogen.
An embodiment of the present invention includes compounds wherein R3 is -O-Ci_6alkyl and R4 is hydrogen.
An embodiment of the present invention includes compounds wherein R3 and R4 taken together form C=O.
Specific embodiments of the present invention include a compound which is selected from the group consisting of the subject compounds of the Examples herein or a pharmaceutically acceptable salt thereof.
The compounds of the present invention may contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within the ambit of this invention. The present invention is meant to comprehend all such isomeric forms of these compounds. Formula I shows the structure of the class of compounds without specific stereochemistry.
The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diasteromeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art. Alternatively, any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
As appreciated by those of skill in the art, halogen or halo as used herein are intended to include fluoro, chloro, bromo and iodo. Similarly, Ci-6, as in Ci-βalkyl is defined to identify the group as having 1, 2, 3, 4, 5 or 6 carbons in a linear or branched arrangement, such that Ci_8alkyl specifically includes methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert- butyl, pentyl, and hexyl. A group which is designated as being independently substituted with substituents may be independently substituted with multiple numbers of such substituents. The term "heterocycle" as used herein includes both unsaturated and saturated heterocyclic moieties, wherein the unsaturated heterocyclic moieties (i.e. "heteroaryl") include benzoimidazolyl, benzimidazolonyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzothiazolyl, benzotriazolyl, benzothiophenyl, benzoxazepin, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl. triazolyl, and N-oxides thereof, and wherein the saturated heterocyclic moieties include azetidϊnyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyridin-2-onyl, pyrrolidinyl, morpholinyl, tetrahydrofuranyl, thiomoφholinyl, and tetrahydrothienyl, and N- oxides thereof.
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particular embodiments are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2- diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl- moφholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric. gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like. Particular embodiments are the citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids. It will be understood that, as used herein, references to the compounds of Formula I are meant to also include the pharmaceutically acceptable salts.
Exemplifying the invention is the use of the compounds disclosed in the Examples and herein. Specific compounds within the present invention include a compound which selected from the group consisting of the compounds disclosed in the following Examples and pharmaceutically acceptable salts thereof and individual diastereomers thereof.
The subject compounds are useful in a method of antagonizing orexϊn receptor activity in a patient such as a mammal in need of such inhibition comprising the administration of an effective amount of the compound. The present invention is directed to the use of the compounds disclosed herein as antagonists of orexin receptor activity. In addition to primates, especially humans, a variety of other mammals can be treated according to the method of the present invention. The present invention is further directed to a method for the manufacture of a medicament for antagonizing orexin receptor activity or treating the disorders and diseases noted herein in humans and animals comprising combining a compound of the present invention with a pharmaceutical carrier or diluent.
The subject treated in the present methods is generally a mammal, such as a human being, male or female. The term "therapeutically effective amount" means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. It is recognized that one skilled in the art may affect the neurological and psychiatric disorders by treating a patient presently afflicted with the disorders or by prophylactically treating a patient afflicted with the disorders with an effective amount of the compound of the present invention. As used herein, the terms "treatment" and "treating" refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of the neurological and psychiatric disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the mentioned conditions, particularly in- a patient who is predisposed to such disease or disorder. The terms "administration of and or "administering a" compound should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to the individual in need thereof.
The term "composition" as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term in relation to pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
The utility of the compounds in accordance with the present invention as orexin receptor OXlR and/or OX2R antagonists may be readily determined without undue experimentation by methodology well known in the art, including the "FLBPR Ca2+ Flux Assay" (Okumura et al., Biochem. Biophys. Res. Comm. 280:976-981, 2001). In a typical experiment the OXl and 0X2 receptor antagonistic activity of the compounds of the present invention was determined in accordance with the following experimental method. For intracellular calcium measurements, Chinese hamster ovary (CHO) cells expressing the rat orexin- 1 receptor or the human orexin-2 receptor, are grown in Iscove's modified DMEM containing 2 mM L-glutamine, 0.5 g/ml G418, 1% hypoxanthine-thymidine supplement, 100 U/ml penicillin, 100 ug/ml streptomycin and 10 % heat-inactivated fetal calf serum (FCS). The cells are seeded at 20,000 cells / well into Becton-Dickinson black 384-well clear bottom sterile plates coated with poly-D- lysine. All reagents were from GIBCO-Invitrogen Corp. The seeded plates are incubated overnight at 37°C and 5% CO2. Ala6'12 human orexin-A as the agonist is prepared as a 1 mM stock solution in 1% bovine serum albumin (BSA) and diluted in assay buffer (HBSS containing 20 mM HEPES, 0.1% BSA and 2.5mM probenecid, pH7.4) for use in the assay at a final concentration of 7OpM. Test compounds are prepared as 10 mM stock solution in DMSO, then diluted in 384-well plates, first in DMSO, then assay buffer. On the day of the assay, cells are washed 3 times with 100 ul assay buffer and then incubated for 60 min (37° C, 5% CO2) in 60 ul assay buffer containing 1 uM Fluo-4AM ester , 0.02 % pluronic acid, and 1 % BSA: The dye loading solution is then aspirated and cells are washed 3 times with 100 ul assay buffer. 30 ul of that same buffer is left in each well. Within the Fluorescent Imaging Plate Reader (FLIPR5 Molecular Devices), test compounds are added to the plate in a volume of 25 ul , incubated for 5 min and finally 25 ul of agonist is added. Fluorescence is measured for each well at 1 second intervals for 5 minutes and the height of each fluorescence peak is compared to the height of the fluorescence peak induced by 70 pM Ala6'12 orexin-A with buffer in place of antagonist. For each antagonist, IC50 value (the concentration of compound needed to inhibit 50 % of the agonist response) is determined. The intrinsic orexin receptor antagonist activity of a compound which may be used in the present invention may be determined by these assays.
In particular, the compounds of the following examples had activity in antagonizing the rat orexin- 1 receptor and/or the human orexin-2 receptor in the aforementioned assays, generally with an IC50 of less than about 50 μM. Many of the compounds within the present invention had activity in antagonizing the rat orexin- 1 receptor and/or the human orexin- 2 receptor in the aforementioned assays with an IC50 of less than about 100 nM. Such a result is indicative of the intrinsic activity of the compounds in use as antagonists of orexin- 1 receptor and/or the orexin-2 receptor. The present invention also includes compounds within the generic scope of the invention which possess activity as agonists of the orexin- 1 receptor and/or the orexin-2 receptor.
The orexin receptors have been implicated in a wide range of biological functions. This has suggested a potential role for these receptors in a variety of disease processes in humans or other species. The compounds of the present invention have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of neurological and psychiatric disorders associated with orexin receptors, including one or more of the following conditions or diseases: sleep disorders, sleep disturbances, including enhancing sleep quality, improving sleep quality, increasing sleep efficiency, augmenting sleep maintenance; increasing the value which is calculated from the time that a subject sleeps divided by the time that a subject is attempting to sleep; improving sleep initiation; decreasing sleep latency or onset (the time it takes to fall asleep); decreasing difficulties in falling asleep; increasing sleep continuity; decreasing the number of awakenings during sleep; decreasing intermittent wakings during sleep; decreasing nocturnal arousals; decreasing the time spent awake following the initial onset of sleep; increasing the total amount of sleep; reducing the fragmentation of sleep; altering the timing, frequency or duration of REM sleep bouts; altering the timing, frequency or duration of slow wave (i.e. stages 3 or 4) sleep bouts; increasing the amount and percentage of stage 2 sleep; promoting slow wave sleep; enhancing EEG-delta activity during sleep; decreasing nocturnal arousals, especially early morning awakenings; increasing daytime alertness; reducing daytime drowsiness; treating or reducing excessive daytime sleepiness; increasing satisfaction with the intensity of sleep; increasing sleep maintenance; idiopathic insomnia; sleep problems; insomnia, hypersomnia, idiopathic hypersomnia, repeatability hypersomnia, intrinsic hypersomnia, narcolepsy, interrupted sleep, sleep apnea, wakefulness, nocturnal myoclonus, REM sleep interruptions, jet-lag, shift workers' sleep disturbances, dyssomnias, night terror, insomnias associated with depression, emotional/mood disorders, Alzheimer's disease or cognitive impairment, as well as sleep walking and enuresis, and sleep disorders which accompany aging; Alzheimer's sundowning; conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, conditions due to drugs which cause reductions in REM sleep as a side effect; fibromyalgia; syndromes which are manifested by non-restorative sleep and muscle pain or sleep apnea which is associated with respiratory disturbances during sleep; conditions which result from a diminished quality of sleep; increasing learning; augmenting memory; increasing retention of memory; eating disorders associated with excessive food intake and complications associated therewith, compulsive eating disorders, obesity (due to any cause, whether genetic or environmental), obesity-related disorders including overeating and bulimia nervosa, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dyslipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, abnormal heart rhythms and arrythrnias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia, metabolic syndrome, also known as syndrome X, insulin resistance syndrome, reproductive hormone abnormalities, sexual and reproductive dysfunction, such as impaired fertility, infertility, hypogonadism in males and hirsutism in females, fetal defects associated with maternal obesity, gastrointestinal motility disorders, such as obesity-related gastro-esophageal reflux, respiratory disorders, such as obesity-hypoventilation syndrome (Pickwickian syndrome), breathlessness, cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, hyperuricaemia, lower back pain, gallbladder disease, gout, kidney cancer, increased anesthetic risk, reducing the risk of secondary outcomes of obesity, such as reducing the risk of left ventricular hypertrophy; diseases or disorders where abnormal oscillatory activity occurs in the brain, including depression, migraine, neuropathic pain, Parkinson's disease, psychosis and schizophrenia, as well as diseases or disorders where there is abnormal coupling of activity, particularly through the thalamus; enhancing cognitive function; enhancing memory; increasing memory retention; increasing immune response; increasing immune function; hot flashes; night sweats; extending life span; schizophrenia; muscle-related disorders that are controlled by the excitation/relaxation rhythms imposed by the neural system such as cardiac rhythm and other disorders of the cardiovascular system; conditions related to proliferation of cells such as vasodilation or vasorestriction and blood pressure; cancer; cardiac arrhythmia; hypertension; congestive heart failure; conditions of the genital/urinary system; disorders of sexual function and fertility; adequacy of renal function; responsivity to anesthetics; mood disorders, such as depression or more particularly depressive disorders, for example, single episodic or recurrent major depressive disorders and dysthymic disorders, or bipolar disorders, for example, bipolar I disorder, bipolar II disorder and cyclothymic disorder, mood disorders due to a general medical condition, and substance-induced mood disorders; anxiety disorders including acute stress disorder, agoraphobia, generalized anxiety disorder, obsessive-compulsive disorder, panic attack, panic disorder, post-traumatic stress disorder, separation anxiety disorder, social phobia, specific phobia, substance-induced anxiety disorder and anxiety due to a general medical condition; acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, ischemic stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage; Huntington's Chorea; amyotrophic lateral sclerosis; multiple sclerosis; ocular damage; retinopathy; cognitive disorders; idiopathic and drug-induced Parkinson's disease; muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions; cognitive disorders including dementia (associated with Alzheimer's disease, ischemia, trauma, vascular problems or stroke, HIV disease, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeldt- Jacob disease, perinatal hypoxia, other general medical conditions or substance abuse); delirium, amnestic disorders or age related cognitive decline; schizophrenia or psychosis including schizophrenia (paranoid, disorganized, catatonic or undifferentiated), schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, psychotic disorder due to a general medical condition and substance-induced psychotic disorder; substance-related disorders and addictive behaviors (including substance-induced delirium, persisting dementia, persisting amnestic disorder, psychotic disorder or anxiety disorder; tolerance, addictive feeding, dependence or withdrawal from substances including alcohol, amphetamines, cannabis, cocaine, hallucinogens, inhalants, nicotine, opioids, phencyclidine, sedatives, hypnotics or anxiolytics); movement disorders, including akinesias and akinetic-rigid syndromes (including Parkinson's disease, drug-induced parkinsonism, postencephalitic parkinsonism, progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration, parkinsonism- ALS dementia complex and basal ganglia calcification), chronic fatigue syndrome, fatigue, including Parkinson's fatigue, multiple sclerosis fatigue, fatigue caused by a sleep disorder or a circadian rhythm disorder, medication-induced parkinsonism (such as neuroleptic-induced parkinsonism, neuroleptic malignant syndrome, neuroleptic- induced acute dystonia, neuroleptic-induced acute akathisia, neuroleptic-induced tardive dyskinesia and medication-induced postural tremor), Gilles de Ia Tourette's syndrome, epilepsy, and dyskinesias [including tremor (such as rest tremor, essential tremor, postural tremor and intention tremor), chorea (such as Sydenham's chorea, Huntington's disease, benign hereditary chorea, neuroacanthocytosis, symptomatic chorea, drug-induced chorea and hemiballism), myoclonus (including generalised myoclonus and focal myoclonus), tics (including simple tics, complex tics and symptomatic tics), restless leg syndrome and dystonia (including generalised dystonia such as iodiopathic dystonia, drug-induced dystonia, symptomatic dystonia and paroxymal dystonia, and focal dystonia such as blepharospasm, oromandibular dystonia, spasmodic dysphonia, spasmodic torticollis, axial dystonia, dystonic writer's cramp and hemiplegic dystonia); attention deficit/hyperactivity disorder (ADHD); conduct disorder; migraine (including migraine headache); urinary incontinence; substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.); psychosis; schizophrenia; anxiety (including generalized anxiety disorder, panic disorder, and obsessive compulsive disorder); mood disorders (including depression, mania, bipolar disorders); trigeminal neuralgia; hearing loss; tinnitus; neuronal damage including ocular damage; retinopathy; macular degeneration of the eye; emesis; brain edema; pain, including acute and chronic pain states, severe pain, intractable pain, inflammatory pain, neuropathic pain, post-traumatic pain, bone and joint pain (osteoarthritis), repetitive motion pain, dental pain, cancer pain, myofascial pain (muscular injury, fibromyalgia), perioperative pain (general surgery, gynecological), chronic pain, neuropathic pain, post-traumatic pain, trigeminal neuralgia, migraine and migraine headache.
Thus, in specific embodiments the present invention provides methods for: enhancing the quality of sleep; augmenting sleep maintenance; increasing REM sleep; increasing stage 2 sleep; decreasing fragmentation of sleep patterns; treating insomnia; enhancing cognition; increasing memory retention; treating or controlling obesity; treating or controlling depression; treating, controlling, ameliorating or reducing the risk of epilepsy, including absence epilepsy; treating or controlling pain, including neuropathic pain; treating or controlling Parkinson's disease; treating or controlling psychosis; or treating, controlling, ameliorating or reducing the risk of schizophrenia, in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of a compound of the present invention.
The subject compounds are further useful in a method for the prevention, treatment, control, amelioration, or reducation of risk of the diseases, disorders and conditions noted herein. The dosage of active ingredient in the compositions of this invention may be varied, however, it is necessary that the amount of the active ingredient be such that a suitable dosage form is obtained. The active ingredient may be administered to patients (animals and human) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment. The dose will vary from patient to patient depending upon the nature and severity of disease, the patient's weight, special diets then being followed by a patient, concurrent medication, and other factors which those skilled in the art will recognize. Generally, dosage levels of between 0.0001 to 10 mg/kg. of body weight daily are administered to the patient, e.g., humans and elderly humans, to obtain effective antagonism of orexin receptors. The dosage range will generally be about 0.5 mg to 1.0 g. per patient per day which may be administered in single or multiple doses. In one embodiment, the dosage range will be about 0.5 mg to 500 mg per patient per day; in another embodiment about 0.5 mg to 200 mg per patient per day; and in yet another embodiment about 5 mg to 50 mg per patient per day. Pharmaceutical compositions of the present invention may be provided in a solid dosage formulation such as comprising about 0.5 mg to 500 mg active ingredient, or comprising about 1 mg to 250 mg active ingredient. The pharmaceutical composition may be provided in a solid dosage formulation comprising about 1 mg, 5 mg, 10 mg, 25 mg, 50 mg, 100 mg, 200 mg or 250 mg active ingredient. For oral administration, the compositions may be provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, such as 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, such as once or twice per day.
The compounds of the present invention may be used in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of the present invention or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone. Such other drug(s) may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. When a compound of the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present invention is contemplated. However, the combination therapy may also includes therapies in which the compound of the present invention and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to a compound of the present invention. The above combinations include combinations of a compound of the present invention not only with one other active compound, but also with two or more other active compounds.
Likewise, compounds of the present invention may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present invention are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. When a compound of the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of the present invention is contemplated. Accordingly, the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of the present invention.
The weight ratio of the compound of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the present invention is combined with another agent, the weight ratio of the compound of the present invention to the other agent will generally range from about 1000:1 to about 1:1000, such as about 200:1 to about 1:200. Combinations of a compound of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s). The compounds of the present invention may be administered in conbination with other compounds which are known in the art to be useful for enhancing sleep quality and preventing and treating sleep disorders and sleep disturbances, including e.g., sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, antihistamines, benzodiazepines, barbiturates, cyclopyrrolones, GABA agonists, 5HT-2 antagonists including 5HT-2A antagonists and 5HT-2A/2C antagonists, histamine antagonists including histamine H3 antagonists, histamine H3 inverse agonists, imidazopyridines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, other orexin antagonists, orexin agonists, prokineticin agonists and antagonists, pyrazolopyrimidines, T-type calcium channel antagonists, triazolopyridines, and the like, such as: adinazolam, allobarbital, alonimid, alprazolam, amitriptyline, amobarbital, amoxapine, armodafinil, APD- 125, bentazepam, benzoctamine, brotizolam, bupropion, busprione, butabarbital, butalbital, capromorelin, capuride, carbocloral, chloral betaine, chloral hydrate, chlordiazepoxide, clomipramine, clonazepam, cloperidone, clorazepate, clorethate, clozapine, conazepam, cyprazepam, desipramine, dexclamol, diazepam, dichloralphenazone, divalproex, diphenhydramine, doxepin, EMD-281014, eplivanserin, estazolam, eszopiclone, ethchlorynol, etomidate, fenobam, flunitrazepam, flurazepam, fluvoxamine, fluoxetine, fosazepam, gaboxadol, glutethimide, halazepam, hydroxyzine, ibutamoren, imipramine, indiplon, lithium, lorazepam, lormetazepam, LY- 156735, maprotiline, MDL- 100907, mecloqualone, melatonin, mephobarbital, meprobamate, methaqualone, methyprylon, midaflur, midazolam, modafinil, nefazodone, NGD-2-73, nisobamate, nitrazepam, nortriptyline, oxazepam, paraldehyde, paroxetine, pentobarbital, perlapine, perphenazine, phenelzine, phenobarbital, prazepam, promethazine, propofol, protriptyline, quazepam, ramelteon, reclazepam, roletamide, secobarbital, sertraline, suproclone, TAK-375, temazepam, thioridazine, tiagabine, tracazolate, tranylcypromaine, trazodone, triazolam, trepipam, tricetamide, triclofos, trifluoperazine, trimetozine, trimipramine, uldazepam, venlafaxine, zaleplon, zolazepam, zopiclone, Zolpidem, and salts thereof, and combinations thereof, and the like, or the compound of the present invention may be administered in conjunction with the use of physical methods such as with light therapy or electrical stimulation.
In another embodiment, the subject compound may be employed in combination with other compounds which are known in the art, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: insulin sensitizers including (i) PPARγ antagonists such as glitazones (e.g. ciglitazone; darglitazone; englitazone; isaglitazone (MCC-555); pioglitazone; rosiglitazone; troglitazone; tularik; BRL49653; CLX-0921; 5-BTZD), GW-0207, LG-100641, and LY-300512, and the like); (iii) biguanides such as metformin and phenformin; (b) insulin or insulin mimetics, such as biota, LP-100, novarapid, insulin detemir, insulin lispro, insulin glargine, insulin zinc suspension (lente and ultralente); Lys-Pro insulin, GLP-I (73-7) (insulintropin); and GLP-I (7-36)-NH2); (c) sulfonylureas, such as acetohexamide; chlorpropamide; diabinese; glibenclamide; glipizide; glyburide; glimepiride; gliclazide; glipentide; gliquidone; glisolamide; tolazamide; and tolbutamide; (d) α-glucosidase inhibitors, such as acarbose, adiposine; camiglibose; emiglitate; miglitol; voglibose; pradimicin-Q; salbostatin; CKD-711; MDL-25,637; MDL-73,945; and MOR 14, and the like; (e) cholesterol lowering agents such as (i) HMG-CoA reductase inhibitors (atorvastatin, itavastatin, fluvastatin, lovastatin, pravastatin, rivastatin, rosuvastatin, simvastatin, and other statins), (ii) bile acid absorbers/sequestrants, such as cholestyramine, colestipol, dialkylaminoalkyl derivatives of a cross-linked dextran; Colestid®; LoCholest®, and the like, (ii) nicotinyl alcohol, nicotinic acid or a salt thereof, (iii) proliferator-activater receptor α agonists such as fenofibric acid derivatives (gemfibrozil, clofibrate, fenofibrate and benzafibrate), (iv) inhibitors of cholesterol absorption such as stanol esters, beta-sitosterol, sterol glycosides such as tiqueside; and azetidinones such as ezetimibe, and the like, and (acyl CoA:cholesterol acyltransferase (ACAT)) inhibitors such as avasimibe, and melinamide, (v) anti-oxidants, such as probucol, (vi) vitamin E, and (vii) thyromimetics; (f) PP ARa agonists such as beclofibrate, benzafibrate, ciprofibrate, clofibrate, etofibrate, fenofibrate, and gemfibrozil; and other fibric acid derivatives, such as Atromid®, Lopid® and Tricor®, and the like, and PP ARa agonists as described in WO 97/36579 by Glaxo; (g) PPARδ agonists; (h) PPAR α/δ agonists, such as muraglitazar, and the compounds disclosed in US 6,414,002; and (i) anti-obesity agents, such as (1) growth hormone secretogogues, growth hormone secretagogue receptor agonists/antagonists, such as NN703, hexarelin, MK-0677, SM- 130686, CP-424,391, L-692,429, and L-163,255; (2) protein tyrosine phosphatase- IB (PTP-IB) inhibitors; (3) cannabinoid receptor ligands, such as cannabinoid CBi receptor antagonists or inverse agonists, such as rimonabant (Sanofi Synthelabo), AMT-251, and SR- 14778 and SR 141716A (Sanofi Synthelabo), SLV-319 (Solvay), BAY 65-2520 (Bayer); (4) anti-obesity serotonergic agents, such as fenfluramine, dexfenfluramine, phentermine, and sibutramine; (5) β3-adrenoreceptor agonists, such as AD9677/TAK677 (Dainippon/Takeda), CL-316,243, SB 418790, BRL-37344, L-796568, BMS-196085, BRL-35135A, CGP12177A, BTA-243, Trecadrine, Zeneca D7114, SR 59119A; (6) pancreatic lipase inhibitors, such as orlistat (Xenical®), Triton WRl 339, RHC80267, lipstatin, tetrahydrolipstatin, teasaponin, diethylumbelliferyl phosphate; (7) neuropeptide Yl antagonists, such as BIBP3226, J-115814, BIBO 3304, LY-357897, CP-671906, GI-264879A; (8) neuropeptide Y5 antagonists, such as GW-569180A, GW-594884A, GW-587081X, GW-548118X, FR226928, FR 240662, FR252384, 1229U91, GI-264879A, CGP71683A, LY-377897, PD-160170, SR-120562A, SR- 120819A and JCF-104; (9) melanin-concentrating hormone (MCH) receptor antagonists; (10) melanin-concentrating hormone 1 receptor (MCHlR) antagonists, such as T-226296 (Takeda); (11) melanin-concentrating hormone 2 receptor (MCH2R) agonist/antagonists; (12) orexin receptor antagonists, such as SB-334867-A, and those disclosed in patent publications herein; (13) serotonin reuptake inhibitors such as fluoxetine, paroxetine, and sertraline; (14) melanocortin agonists, such as Melanotan II; (15) other Mc4r (melanocortin 4 receptor) agonists, such as CHIR86036 (Chiron), ME-10142, and ME-10145 (Melacure), CHIR86036 (Chiron); PT- 141, and PT-14 (Palatin); (16) 5HT-2 agonists; (17) 5HT2C (serotonin receptor 2C) agonists, such as BVT933, DPCA37215, WAY161503, R-1065; (18) galanin antagonists; (19) CCK agonists; (20) CCK-A (cholecystokinin-A) agonists, such as AR-R 15849, GI 181771, JMV- 180, A-71378, A-71623 and SRl 4613; (22) corticotropin-releasing hormone agonists; (23) histamine receptor-3 (H3) modulators; (24) histamine receptor-3 (H3) antagonists/inverse agonists, such as hioperamide, 3-(lH-imidazol-4-yl)propyl N-(4-pentenyl)carbamate, clobenpropit, iodophenpropit, imoproxifan, GT2394 (Gliatech), and O-[3-(lH-imidazol-4-yl)propanol]- carbamates; (25) β-hydroxy steroid dehydrogenase- 1 inhibitors (β-HSD-1); 26) PDE (phosphodiesterase) inhibitors, such as theophylline, pentoxifylline, zaprinast, sildenafil, amrinone, milrinone, cilostamide, rolipram, and cilomilast; (27) phosphodiesterase-3B (PDE3B) inhibitors; (28) NE (norepinephrine) transport inhibitors, such as GW 320659, despiramine, talsupram, and nomifensine; (29) ghrelin receptor antagonists; (30) leptin, including recombinant human leptin (PEG-OB, Hoffman La Roche) and recombinant methionyl human leptin (Amgen); (31) leptin derivatives; (32) BRS3 (bombesin receptor subtype 3) agonists such as [D-Phe6,beta- Alal l,Phel3,Nlel4]Bn(6-14) and [D-Phe6,Phel3]Bn(6-13)propylamide, and those compounds disclosed in Pept. Sci. 2002 Aug; 8(8): 461-75); (33) CNTF (Ciliary neurotrophic factors), such as GI-181771 (Glaxo-SmithKline), SR146131 (Sanofi Synthelabo), butabindide, PD170,292, and PD 149164 (Pfizer); (34) CNTF derivatives, such as axokine (Regeneron); (35) monoamine reuptake inhibitors, such as sibutramine; (36) UCP-I (uncoupling protein- 1), 2, or 3 activators, such as phytanic acid, 4-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-napthalenyl)-l- propenyljbenzoic acid (TTNPB), retinoic acid; (37) thyroid hormone β agonists, such as KB- 2611 (KaroBioBMS); (38) FAS (fatty acid synthase) inhibitors, such as Cerulenin and C75; (39) DGATl (diacylglycerol acyltransferase 1) inhibitors; (40) DGAT2 (diacylglycerol acyltransferase 2) inhibitors; (41) ACC2 (acetyl-CoA carboxylase-2) inhibitors; (42) glucocorticoid antagonists; (43) acyl-estrogens, such as oleoyl-estrone, disclosed in del Mar- Grasa, M. et al., Obesity Research, 9:202-9 (2001); (44) dipeptidyl peptidase IV (DP-IV) inhibitors, such as isoleucine thiazolidide, valine pyrrolidide, NVP-DPP728, LAF237, MK-431, P93/01., TSL 225, TMC-2A/2B/2C, FE 999011, P9310/K364, VIP 0177, SDZ 274-444; (46) dicarboxylate transporter inhibitors; (47) glucose transporter inhibitors; (48) phosphate transporter inhibitors; (49) Metformin (Glucophage®); and (50) Topiramate (Topimax®); and (50) peptide YY, PYY 3-36, peptide YY analogs, derivatives, and fragments such as BIM- 43073D, BIM-43004C (Olitvak, D.A. et al., Dig. Dis. Sci. 44(3):643-48 (1999)); (51) Neuropeptide Y2 (NP Y2) receptor agonists such NPY3-36, N acetyl [Leu(28,31)] NPY 24-36, TASP-V, and cyclo-(28/32)-Ac-[Lys28-Glu32]-(25-36)-pNPY; (52) Neuropeptide Y4 (NPY4) agonists such as pancreatic peptide (PP), and other Y4 agonists such as 1229U91 ; (54) cyclooxygenase-2 inhibitors such as etoricoxib, celecoxib, valdecoxib, parecoxib, lumiracoxib, BMS347070, tiracoxib or JTE522, ABT963, CS502 and GW406381, and pharmaceutically acceptable salts thereof; (55) Neuropeptide Yl (NPYl) antagonists such as BEBP3226, J-115814, BIBO 3304, LY-357897, CP-671906, GI-264879A; (56) Opioid antagonists such as nalmefene (Revex ®), 3-methoxynaltrexone, naloxone, naltrexone; (57) 1 lβ HSD-I (11-beta hydroxy steroid dehydrogenase type 1) inhibitor such as BVT 3498, BVT 2733; (58) aminorex; (59) amphechloral; (60) amphetamine; (61) benzphetamine; (62) chlorphentermine; (63) clobenzorex; (64) cloforex; (65) clominorex; (66) clorteπnine; (67) cyclexedrine; (68) dextroamphetamine; (69) diphemethoxidine, (70) N-ethylamphetamine; (71) fenbutrazate; (72) fenisorex; (73) fenproporex; (74) fludorex; (75) fluminorex; (76) furfurylmethylamphetamine; (77) levamfetamine; (78) levophacetoperane; (79) mefenorex; (80) metamfepramone; (81) methamphetamine; (82) norpseudoephedrine; (83) pentorex; (84) phendimetrazine; (85) phenmetrazine; (86) picilorex; (87) phytopharm 57; and (88) zonisamide. hi another embodiment, the subject compound may be employed in combination with an anti-depressant or anti-anxiety agent, including norepinephrine reuptake inhibitors (including tertiary amine tricyclics and secondary amine tricyclics), selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, α-adrenoreceptor antagonists, neurokinin- 1 receptor antagonists, atypical anti-depressants, benzodiazepines, 5-HT1A agonists or antagonists, especially 5-HTIA partial agonists, and corticotropin releasing factor (CRF) antagonists. Specific agents include: amitriptyline, clomipramine, doxepin, imipramine and trimipramine; amoxapine, desipramine, maprotiline, nortriptyline and protriptyline; fluoxetine, fiuvoxamine, paroxetine and sertraline; isocarboxazid, phenelzine, tranylcypromine and selegiline; moclobemide: venlafaxine; aprepitant; bupropion, lithium, nefazodone, trazodone and viloxazine; alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazepam, oxazepam and prazepam; buspirone, flesinoxan, gepirone and ipsapirone, and pharmaceutically acceptable salts thereof. In another embodiment, the subject compound may be employed in combination with anti-Alzheimer's agents; beta-secretase inhibitors; gamma-secretase inhibitors; growth hormone secretagogues; recombinant growth hormone; HMG-CoA reductase inhibitors; NSAID's including ibuprofen; vitamin E; anti -amyloid antibodies; CB-I receptor antagonists or CB-I receptor inverse agonists; antibiotics such as doxycycline and rifampin; N-methyl-D- aspartate (NMDA) receptor antagonists, such as memantine; cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, and tacrine; growth hormone secretagogues such as ibutamoren, ibutamoren mesylate, and capromorelin; histamine H3 antagonists; AMPA agonists; PDE IV inhibitors; GABAA inverse agonists; or neuronal nicotinic agonists.
In another embodiment, the subject compound may be employed in combination with sedatives, hypnotics, anxiolytics, antipsychotics, antianxiety agents, cyclopyrrolones, imidazopyri dines, pyrazolopyrimidines, minor tranquilizers, melatonin agonists and antagonists, melatonergic agents, benzodiazepines, barbiturates, 5HT-2 antagonists, and the like, such as: adinazolam, allobarbital, alonimid, alprazolam, amitriptyline, amobarbital, amoxapine, bentazepam, benzoctamine, brotizolam, bupropion, busprione, butabarbital, butalbital, capuride, carbocloral, chloral betaine, chloral hydrate, chlordiazepoxide, clomipramine, clonazepam, cloperidone, clorazepate. clorethate, clozapine, cyprazepam, desipramine, dexclamol, diazepam, dichloralphenazone, divalproex, diphenhydramine, doxepin, estazolam, ethchlorvynol, etomidate, fenobam, flunitrazepam, flurazepam, fluvoxamine, fluoxetine, fosazepam, glutethimide, halazepam, hydroxyzine, imipramine, lithium, lorazepam, lormetazepam, maprotiline, mecloqualone, melatonin, mephobarbital, meprobamate, methaqualone, midaflur, midazolam, nefazodone, nisobamate, nitrazepam, nortriptyline, oxazepam, paraldehyde, paroxetine, pentobarbital, perlapine, peφhenazine, phenelzine, phenobarbital, prazepam, promethazine, propofol, protriptyline, quazepam, reclazepam, roletamide, secobarbital, sertraline, suproclone, temazepam, thioridazine, tracazolate, tranylcypromaine, trazodone, triazolam, trepipam, tricetamide, triclofos, trifluoperazine, trimetozine, trimipramine, uldazepam, venlafaxine, zaleplon, zolazepam, Zolpidem, and salts thereof, and combinations thereof, and the like, or the subject compound may be administered in conjunction with the use of physical methods such as with light therapy or electrical stimulation.
In another embodiment, the subject compound may be employed in combination with levodopa (with or without a selective extracerebral decarboxylase inhibitor such as carbidopa or benserazide), anticholinergics such as biperiden (optionally as its hydrochloride or lactate salt) and trihexyphenidyl (benzhexol) hydrochloride, COMT inhibitors such as entacapone, MOA-B inhibitors, antioxidants, A2a adenosine receptor antagonists, cholinergic agonists, NMDA receptor antagonists, serotonin receptor antagonists and dopamine receptor agonists such as alentemol, bromocriptine, fenoldopam, lisuride, naxagolide, pergolide and pramipexole. It will be appreciated that the dopamine agonist may be in the form of a pharmaceutically acceptable salt, for example, alentemol hydrobromide, bromocriptine mesylate, fenoldopam mesylate, naxagolide hydrochloride and pergolide mesylate. Lisuride and pramipexol are commonly used in a non-salt form.
In another embodiment, the subject compound may be employed in combination with acetophenazine, alentemol, benzhexol, bromocriptine, biperiden, chlorpromazine, chlorprothixene, clozapine, diazepam, fenoldopam, fluphenazine, haloperidol, levodopa, levodopa with benserazide, levodopa with carbidopa, lisuride, loxapine, mesoridazine, molindolone, naxagolide, olanzapine, pergolide, perphenazine, pimozide, pramipexole, risperidone, sulpiride, tetrabenazine, trihexyphenidyl, thioridazine, thiothixene or trifluoperazine.
In another embodiment, the subject compound may be employed in combination with a compound from the phenothiazine, thioxanthene, heterocyclic dibenzazepine, butyrophenone, diphenylbutylpiperidine and indolone classes of neuroleptic agent. Suitable examples of phenothiazines include chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine. Suitable examples of thioxanthenes include chlorprothixene and thiothixene. An example of a dibenzazepine is clozapine. An example of a butyrophenone is haloperidol. An example of a diphenylbutylpiperidine is pimozide. An example of an indolone is molindolone. Other neuroleptic agents include loxapine, sulpiride and risperidone. It will be appreciated that the neuroleptic agents when used in combination with thesubject compound may be in the form of a pharmaceutically acceptable salt, for example, chlorpromazine hydrochloride, mesoridazine besylate, thioridazine hydrochloride, acetophenazine maleate, fluphenazine hydrochloride, fiurphenazine enathate, fluphenazine decanoate, trifluoperazine hydrochloride, thiothixene hydrochloride, haloperidol decanoate, loxapine succinate and molindone hydrochloride. Perphenazine, chlorprothixene, clozapine, haloperidol, pimozide and risperidone are commonly used in a non-salt form.
In another embodiment, the subject compound may be employed in combination with an anoretic agent such as aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine. clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N-ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; selective serotonin reuptake inhibitor (SSRI); halogenated amphetamine derivatives, including chlorphentermine, cloforex, clortermine, dexfenfluramine, fenfluramine, picilorex and sibutramine; and pharmaceutically acceptble salts thereof
In another embodiment, the subject compound may be employed in combination with an opiate agonist, a lipoxygenase inhibitor, such as an inhibitor of 5-lipoxygenase, a cyclooxygenase inhibitor, such as a cyclooxygenase-2 inhibitor, an interleukin inhibitor, such as an interleukin- 1 inhibitor, an NMDA antagonist, an inhibitor of nitric oxide or an inhibitor of the synthesis of nitric oxide, a non-steroidal antiinflammatory agent, or a cytokine-suppressing antiinflammatory agent, for example with a compound such as acetaminophen, asprin, codiene, fentanyl, ibuprofen, indomethacin, ketorolac, morphine, naproxen, phenacetin, piroxicam, a steroidal analgesic, sufentanyl, sunlindac, tenidap, and the like. Similarly, the subject compound may be administered with a pain reliever; a potentiator such as caffeine, an H2-antagonist, simethicone, aluminum or magnesium hydroxide; a decongestant such as phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxy-ephedrine; an antiitussive such as codeine, hydrocodone, caramiphen, carbetapentane, or dextramethorphan; a diuretic; and a sedating or non-sedating antihistamine.
The compounds of the present invention may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. In addition to the treatment of warmblooded animals such as mice, rats, horses, cattle, sheep, dogs, cats, monkeys, etc., the compounds of the invention are effective for use in humans.
The pharmaceutical compositions for the administration of the compounds of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening age.nts, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. Compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil. Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Oily suspensions may be formulated by suspending the active ingredient in a suitable oil. Oil-in-water emulsions may also be employed. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Pharmaceutical compositions of the present compounds may be in the form of a sterile injectable aqueous or oleagenous suspension. The compounds of the present invention may also be administered in the form of suppositories for rectal administration. For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention may be employed. The compounds of the present invention may also be formulated for administered by inhalation. The compounds of the present invention may also be administered by a transdermal patch by methods known in the art.
Several methods for preparing the compounds of this invention are illustrated in the following Schemes and Examples. Starting materials are made according to procedures known in the art or as illustrated herein. The following abbreviations are used herein: Me: methyl; Et: ethyl; t-Bu: tert-butyl; Ar: aryl; Ph: phenyl; Bn: benzyl; Ac: acetyl; THF: tetrahydrofuran; DEAD: diethylazodicarboxylate; DIPEA: N,N-dϊisopropylethylamine; DMSO: dimethylsulfoxide; EDC: N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide; HOBT: hydroxybenzotriazole hydrate; Boc: tert-butyloxy carbonyl; Et3N: triethylamine; DCM: dichloromethane; DCE: dichloroethane; BSA: bovine serum albumin; TFA: trifluoracetic acid; DMF: N,N-dimethylfoπnamide; MTBE: methyl tert-butyl ether;SOCl2: thionyl chloride; CDI: carbonyl diimidazole; rt: room temperature; HPLC: high performance liquid chromatography. The compounds of the present invention can be prepared in a variety of fashions.
In some cases the final product may be further modified, for example, by manipulation of substituents. These manipulations may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art. In some cases the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products. The following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way.
SCHEME A
Figure imgf000030_0001
ferf-butyl 4-(6-chloro-l 3-benzothiazol-2-ylV1.4-diazepane-l-carboxylate (A-2)
A solution of A-I (2.4 g, 12 mmol) in DMF (6 ml) was treated with 2.45 g (12 mmol) 2,6-dichlorobenzothiazole and 1.7 mL (12 mmol) triethylamine and stirred at 1000C overnight. After cooling to room temperature, the reaction was diluted with EtOAc, washed with water and then brine. The organic phase was dried over MgSO4, filtered and concentrated to provide A-2. Data for A-2: 1HNMR (500 MHz, CDCl3) δ 7.55 (s, IH), 7.4 (m, IH)5 7.25 (m, IH), 3.8 - 3.6 (m, 6H), 3.5 - 3.3 (m, 2H), 2.05 (m, 2H), 1.4 (m, 9H) ppm.
6-chloro-2-π-4-diazepan-l-ylVL3-benzothiazole hydrochloride (A-3)
A solution of A-2 in EtOAc was treated with HCl (g) and stirred overnight at room temperature in a sealed flask. The reaction was concentrated by rotary evaporation to provide the hydrochloride salt A-3 as a white solid. Data for A-3: LC/MS: rt = 1.32 min; m/z (M + H) = 268.0 found; 268.1 required for free-base.
6-chloro-2-r4-(2,6-dimethoxybenzoylVl,4-diazepan-l-yl1-l,3-benzothiazole CA-4)
To a solution of A-3 (250 mg, 0.82 mmol) in 3 mL CH2CI2 was added 720 μL
(4.1 mmol) triethylamine and 206 mg (1.0 mml) 2,6-dimethoxybenzoyl chloride. After stirring for 3 h, the reaction was dumped into water and partitioned with the aid of additional DCM. After separation of the layers, the organic was dried with Na2SO4 and concentrated by rotary evaporation. The residue was purified by flash column chromatography (hexanes/EtOAc) to provide A-4 as a white solid. Data for A-4: HRMS (APCI) m/z (M+H): 432.1149 found, 432.1143 required.
SCHEME B
Figure imgf000031_0001
2-(2H-1.2.3-tria2ol-2-ylV5-methylbenzoic acid (B-2)
A solution of 2-iodo-5-methylbenzoic acid (4.0 g, 15.3 mmol) in DMF (10 mL) was treated with 1,2,3-triazole (2.1 g, 30.5 mmol), CsCO3 (9.95 g, 30.5 mmol), CuI (0.145 g, 0.76 mmol) and trans-N,N'-dimethylcyclohexane-l,2-diamine (0.43 g, 3.05 mmol). The mixture was heated at 120 0C for 10 min in a microwave reactor. The reaction was cooled to room temperature, diluted with water, and washed with EtOAc. The aqueous phase was acidified with IN HCl and extracted with EtOAc. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by gradient elution on Siθ2 (0 to 10% MeOH in water with 0.1% AcOH) to give the faster eluting 2-(2H-l,2,3-triazol-2-yl)-5-methylbenzoic acid B-2, followed by the undesired regioisomer isomer, l-(2H-l,2,3-triazol~2-yl)-5-methylbenzoic acid. Data for B-2: 1HNMR (500 MHz, DMSO-d6) d 12.98 (br s , IH), 8.04 (s, 2H), 7.72-7 '.45 (m, 3H), 2.41 (s, 3H) ppm.
Figure imgf000031_0002
To a solution of 150 mg (0.45 mmol) B-I, 101 mg (0.50 mmol) B-2, 77 mg (0.57 mmol) 1-hydroxybenzotriazole hydrate, and 250 μL (1.81 mmol) triethylamine in 3 mL of DMF was added 109 mg (0.57 mmol) EDC and the reaction was stirred overnight at room temperature. The reaction was partitioned between EtOAc and saturated aqueous NaΗCθ3. The layers were separated and the organic was washed twice with brine, dried over Na2SO4 and concentrated by rotary evaporation. The residue was purified by column chromatography on silica gel (EtOAc/hexanes) to provide B-3 as a white solid. Data for B-3: HRMS (APCI) m/z (M+H) 403.1888 found; 403.1877 required. SCHEME C
Figure imgf000032_0001
6-chloro-2-["4-(phenylsulfonylVl ,4-diazepan-l -yl]-l ,3-benzothiazole (C-I)
To a solution of A-3 (30 mg, 0.1 mmol) in 1 mL of CH2CI2 was added 19 μL
(0.15 mmol) benzenesulfonyl chloride and 50 μL (0.3 mmol) DIPEA and the reaction was stirred overnight at room temperature. The reaction was partitioned between EtOAc and saturated aqueous !NILj-Cl. The layers were separated and the organic was washed twice with brine, dried over MgSC*4 and concentrated by rotary evaporation. The residue was purified by reverse phase HPLC (CH3CN/H2O containing 0.1% TFA as a modifier) to provide C-I . Data for C-I : LRMS m/z (M+H) 407.9 found; 408.0 required.
SCHEME D
Figure imgf000032_0002
D-2
/e/-/-buty1 4-f 2,6-dimethoxybenzoylV 1 ,4-diazepane- 1 -carboxylate CD- 1 >
To a solution of A-I (7.2 g, 36.0 mmol) in 50 mL CH2CI2 was added 10.0 mL
(72 mmol) triethylamine and 7.93 g (39.6 mml) 2,6-dimethoxybenzoyl chloride. After stirring for 72 h, the reaction was dumped into 10% aqueous citric acid and partitioned with the aid of additional CH2C12- After separation of the layers, the aqueous was extracted again with CH2C12, the combined organic layers were washed with saturated aqueous NaHCθ3, then water, and dried over Na2SO4. Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide D-I as a white solid. Data for D- 1 : LC/MS: rt = 2.02 min; m/z (M + H) = 365.1, found; 365.2 required.
l-(2,6-dimethoxybenzoylV1.4-diazepane hydrochloride (D-2)
A solution of 5.0 g (13.7 mmol) D-2 in 150 mL EtOAc was treated with HCl (g) until the solution was warm to the touch. The reaction flask was sealed with a stopper and allowed to stir at room temperature overnight. The solvents were removed by rotary evaporation, the residue was triturated with Et2θ, and again concentrated to provide D-2 as an off-white solid.
Data for D-2: LC/MS (see two conformers under these conditions): rt = 0.62 & 0.87 min; m/z (M + H) = 265.1, found; 265.1 required for the free-base.
2-chloroquinazoline (D-3)
To a solution of 1.0 g (8.3 mmol) 2-aminobenzaldehyde in 30 mL THF at 00C was added 1.73 mL (12.4 mmol) triethylamine followed by 980 mg (3.30 mmol) triphosgene. After stirring 4 h at 00C, 9.4 mL (66 mmol) of a 7M solution of NH3 in MeOH was added and the resulting mixture was stirred overnight while slowly warming to room temperature. The solvents were removed by rotary evaporation and the residue was triturated with Et2θ and water.
The resulting white solid was removed by filtration to provide the intermediate carbonyl compound [quinazolin-2(lH)-one]. A solution of 55mg (0.37 mmol) of this material in 1 mL POCI3 was heated to 100°C for 1 h, at which time most of the excess solvent was removed with a stream of N2. The residue was partitioned between EtOAc and a 5% aqueous solution of Na2CO3. After separation of the layers, the organic was washed with brine and dried over Na2SO4. Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide D-3 as a white solid. Data for D-3: LC/MS: rt = 1.51 min; m/z (M + H) = 165.0, found; 165.0 required; 1HNMR (500 MHz, CDCl3) δ 9.35 (s, IH), 8.0 (m, 3H), 7.7 (s, IH) ppm.
∑-^-^ό-dimethoxybenzoylM^-diazepan-l-ylfauinazoline rt)^)
A solution of 100 mg (0.33 mmol) D-2 in DMF (2 ml) was treated with 60 mg (0.37 mmol) D-3 and 140 mg (1 mmol) K2CO3 and stirred at 1000C in a microwave reactor for
20 min. After cooling to room temperature, the reaction was diluted with EtOAc, washed with saturated aqueous NaHCθ3, then 3 times with brine and dried over Na2SO4- Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide D-4 as a beige solid. Data for D-4: HRMS (APCI) m/z (M+H) 393.1930 found; 393.1921 required.
SCHEME E
Figure imgf000034_0001
2-("6-fluoro- 1 ,4-diazepan- 1 -ylV 1 ,3-ben2oxazole (E-2)
A solution of 3.2g (11.4 mmol) E-I (C. B. Ziegler et. al. J. Med. Chem. 1990, 33, 142-146) in DMF (30 ml) was treated with 1.44 mL (12.6 mmol) 2-chlorobenzoxazole and 6.4 g (45.7 mmol) K2CO3 and stirred at 8O0C in an oil bath for Ih. After cooling to room temperature, the reaction was diluted with EtOAc, washed with saturated aqueous NaHCC»3, then 3 times with brine and dried over Na2SO4. LC/MS indicates that the desired material was in the aqueous phases, so they were combined, saturated with NaCl and extracted 3 x with 2:1 CHCl3/EtOH. These extracts were concentrated to provide E-2 as an off-white solid. Data for
E-2: LOMS: rt = 0.94 min; m/z (M + H) = 236.1, found; 236.1 required.
2-f4-[5-methyl-2-('2H"-1.23-triazol-2-vnbenzovn-L4-diazepan-l-yl>-13-benzoxazole fE-3')
To a solution of 410 mg (1.74 mmol) E-2, 425 mg (2.1 mmol) B-2, 294 mg (2.2 mmol) 1-hydroxybenzotriazole hydrate, and 730 μL (5.2 mmol) triethylamine in 25 mL of DMF was added 418 mg (2.2 mmol) EDC and the reaction was stirred at 50°C for 3 h. The reaction was partitioned between EtOAc and saturated aqueous NaHCOs. The layers were separated and the organic was washed twice with brine, dried over Na2SO4 and concentrated by rotary evaporation. The residue was purified by column chromatography on silica gel (EtOAc/hexanes) to provide E-3 as a white solid. Data for E-3: HRMS (APCI) m/z (M+H) 421.1785 found; 421.1783 required. SCHEME F
Figure imgf000035_0001
EtOAc
Figure imgf000035_0002
Figure imgf000035_0003
fert-butyl 6-hydroxy-l,4-diazepane-l-carboxylate (¥-2)
To a solution of F-I (W. S. Sam et. al. J. Org. Chem. 1971, 36, 1711-1714) in a solution of MeOH/CH2Cl2 cooled to O0C was added dropwise άi-tert-bvtyl dicarbonate (in a 0.126 mol ratio) in CH2C-2- The reaction was slowly wanned to room temperature while stirring overnight. The CH2CI2 was removed by rotary evaporation, the residue was dissolved in water and filtered through a Buchner funnel. The filtrate was extracted with CHCI3, dried over Na2SO4, and concentrated by rotary evaporation. The residue was purified by column chromatography on silica gel (1 :9 MeOH/CHCl3) to provide F-2 as a pale yellow oil. Data for F- 2: LC/MS: rt = 0.89 min; m/z (M + H) = 217.1, found; 217.1 required.
fert-butyl 4-(2,6-dimethoxybenzoyl)-6-hydroxy-l ,4-diazepane- 1 -carboxylate (F-3)
To a solution of F-2 (1.02 g, 4.72 mmol) in 100 mL CH2CI2 at -78°C was added
1.31 mL (9.43 mmol) triethylamine and 993 mg (4.95 mml) 2,6-dimethoxybenzoyl chloride. After stirring while slowly warming to room temperature overnight, the reaction was dumped into 10% aqueous citric acid and partitioned with the aid of additional CH2CI2. After separation of the layers, the aqueous was extracted again with CH2CI2, the combined organic layers were washed with saturated aqueous NaHCO3, then water, and dried over Na2SO4- Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide F-3 as a white solid. Data for F-3: LC/MS (see two conformers under these conditions): rt = 1.57 & 1.79 min; m/z (M + H) = 381.0, found; 381.2 required.
_l-(2,6-dimethoxybenzoylM,4-diazepan-6-ol hydrochloride (F-4)
A solution of 182 mg (0.48 mmol) F-3 in 10 mL EtOAc was treated with HCl (g) until the solution was warm to the touch. The reaction flask was sealed with a stopper and allowed to stir at room temperature overnight. The solvents were removed by rotary evaporation, the residue was triturated with Et2θ, and again concentrated to provide F-4 as a white solid.
Data for F-4: LC/MS: rt == 0.41 min; m/z (M + H) = 281.2, found; 281.1 required for the free- base.
1 -(1 ,3-benzoxazol-2-ylV4-(2.6-dimethoxybenzoviyi ,4-diazepan-6-ol (F-5")
A solution of 105 mg (0.33 mmol) F-4 in DMF (3 ml) was treated with 56 mg (0.35 mmol) 2-chlorobenzoxazole and 185 mg (1.32 mmol) K2CO3 and stirred at 8O0C in a microwave reactor for 20 min. After cooling to room temperature, the reaction was diluted with EtOAc, washed with saturated aqueous NaHCO3, then 3 times with brine and dried over Na2SOφ Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide F-5 as a white solid. Data for F-5: HRMS (APCI) m/z (M+H) 398.1722 found; 398.1711 required.
SCHEME G
Figure imgf000037_0001
tert-butyl 4-(2,6-dimethoxybenzoyl)-6-oxo- 1.4-diazepane- 1 -carboxylate CG- 1 )
A solution of 550 rag (1.45 mmol) F-3 in CH2CI2 (40 ml) was treated with 736 mg (1.74 mmol) Dess-Martin Periodinane. After stirring for 1 h at room temperature, a 5% aqueous solution of Na2SO3 and a saturated aqueous solution of NaHCθ3 were added successively, and the resultant biphasic mixture was stirred vigorously for 30 min. The reaction was added to a separatory funnel, the layers were separated, and the aqueous was extracted with CH2CI2. The combined organic phases were washed with saturated aqueous NaHCO3, then water, dried over Na2SO4, and concentrated by rotary evaporation to provide G-I as a white taffy. Data for G-I : LC/MS (see two conformers under these conditions): rt = 1.97 & 2.06 min; m/z (M + H) = 379.0, found; 379.2 required.
1 -(13-benzoxazol-2-yl>4-(2,6-dimethoxybenzovD-l .4-diazepan-6-one (G-3*)
The G-I material made in the previous procedure dissolved in 25 mL EtOAc was treated with HCl (g) until the solution was warm to the touch. The reaction flask was sealed with a stopper and allowed to stir at room temperature 1.5 h. The solvents were removed by rotary evaporation, the residue was triturated with Et2θ, and again concentrated to provide G-2. This material was dissolved in 10 mL DMF and treated with 244 mg (1.6 mmol) 2-chlorobenzoxazole and 607 mg (4.34 mmol) K2CO3 and stirred at 6O0C in an oil bath for 2 h After cooling to room temperature, the reaction was diluted with EtOAc, washed with saturated aqueous NaHCθ3, then 3 times with brine and dried over Na2SO4. Following concentration by rotary evaporation, the residue was purified by flash column chromatography (hexanes/EtOAc) to provide G-3 as a pale yellow solid. Data for G-3: HRMS (APCI) m/z (M+H) 396.1551 found; 396.1554 required. SCHEME H
Figure imgf000038_0001
1 -( 1 ,3-benzoxa2θl-2-yl)-4-(2.6-dimethoxybenzoyπ-.?V-inethyl-l .4-diazepan-6-amine (H- 1 )
To a solution of 61 mg (0.154 mmol) G-3 in 5 inL 1 ,2-dichloroethane was added 770 μL (1.54 mmol) of 2M methylamine in THF and 40 μl (0.62 mmol) HOAc. After stirring for 2 h at room temperature, 65 mg (0.31 mmol) sodium triacetoxyborohydride was added and the reaction was stirred overnight at room temperature. It was then diluted with EtOAc, washed with saturated aqueous NaHCθ3, then brine, and dried over Na2SOφ Following concentration by rotary evaporation, the residue was purified by flash column chromatography (EtO Ac/20: 1 : 1 EtOH:NH4θH:H2θ) to provide H-I as a white solid- Data for H-I : HRMS (APCI) m/z (M+H)
411.2028 found; 411.2027 required.
SCHEME I
2-f 4- ( r5-methyl-2-C2H- 1 ,23-triazol-2-vnphenvncarbonothioyl } - 1.4-diazepan- 1 -ylV 1.3- benzoxazole (1-1)
To a solution of 100 mg (0.25 mmol) B-3 in 1 mL THF was added 60 mg (0.149 mmol) Lawesson's reagent and the reaction was stirred 72 h at room temperature. An additional 60 mg of Lawesson's reagent was added and the mixture was heated at 500C for several hours. The reaction was then heated at 1000C for 5 minutes in a microwave reactor. An additional 100 mg of Lawesson's was added and the reaction was heated at 1000C in the microwave for an additional 10 minutes. To the reaction was added silica gel, the mixtur4e was concentrated, then purified by flash column chromatography (EtOAc/hexanes) to provide I- 1 as an off-white solid. Data for 1-1: HRMS (APCI) m/z (M+H) 419.1652 found; 419.1649 required.
TABLE l The following compounds were prepared using the foregoing methodology, but substituting the appropriately substituted reagent, as described in the foregoing Reaction Schemes and Examples. The requisite starting materials were commercially available, described in the literature or readily synthesized by one skilled in the art of organic synthesis without undue experimentation. Some final products were purified by flash chromatography (Siθ2;
EtOAc/hexanes) and were isolated as the free-base; alternately, some products were purified by reverse phase HPLC (CH3CN/H2O containing 0.1% TFA as a modifier) and isolated as the TFA salt, in which case the masses reported and found are for the free-base.
Figure imgf000039_0001
Figure imgf000040_0001
While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention.

Claims

WHAT IS CLAIMED IS:
1. A compound of the fomrala I:
Figure imgf000042_0001
I wherein:
X is selected from -SO2-, -(C=O)-, and -(C=S)-;
Rl is selected from the group consisting of:
(1 ) phenyl, where the phenyl is substituted with Rl a, Rl b and Rl C5 and
(2) napthyl, where the napthyl is substituted with Rla, Rib and Rlc;
(3) heteroaryl, where the heteroaryl is substituted with RIa5 Rib and Rl c;
R2 is heteroaryl, where the heteroaryl is substituted with R2a5 R2b and R2c;
Rl a, Rib, RIc5 R2a5 R2b and R2c are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -(C=O)m-On-Ci-6alkyl, where m is 0 or 1, n is 0 or 1 (wherein if m is 0 or n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) ~(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(6) -(C=O)m-C2-4alkenyl. where the alkenyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(7) -(C=O)m-C2-4alkynyl, where the alkynyl is unsubstituted or substituted with one or more substituents selected from Rl 3.
(8) -(C=O)m-Oirphenyl or -(C=O)m-On-napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(9) -(C=O)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl3, (10) -(C=O)1n-NRl ORl 1 , wherein Rl 0 and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) C i_6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
(c) C3_6alkenyl, which is unsubstituted or substituted with one or more substituents selected from Rl 39
(d) cycloalkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
(e) phenyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3, and
(f) heterocycle, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
Figure imgf000043_0001
(12) -S(O)q-Rl2, where q is 0, 1 or 2 and where Rl 2 is selected from the definitions of
RlO and Rl I5
(13) -CO2H,
(14) -CN, and
(15) -NO2;
R4 are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -Ci-galkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) -O-Ci-6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3.
(6) -phenyl or -napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(7) -NRl ORI 1, wherein RlO and Rl 1 are independently selected from the group consisting of:
(a) hydrogen, (b) Ci-6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3, or R3 and R4 taken together form C=O;
Rl 3 is selected from the group consisting of:
(1) halogen,
(2) hydroxyl,
(3) -(C=O)1n-On-C i-6alkyl, where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 4,
(4) -On-(C i -3)perfluoroalkyl,
(5) -(C=O)m-On-C3-6cycloalkyl, where the cycloalkyl is unsubstituted or substituted with one or more substituents selected from Rl 4,
(6) -(C=O)m-C2-4alkenyl, where the alkenyl is unsubstituted or substituted with one or more substituents selected from Rl 4,
(7) -(C=O)m-On-ρhenyl or -(C=O)m-On-napthyl, where the phenyl or napthyl is unsubstituted or substituted with one or more substituents selected from Rl4.
(8) -(C=0)m-On-heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from R 14,
(9) -(C=O)1n-NRl ORl I,
Figure imgf000044_0001
(11) -S(O)q-Rl2
(12) -CO2H5
(13) -CN, and
(14) -NO2; .
Rl 4 is selected from the group consisting of:
(1) hydroxyl,
(2) halogen,
(3) Ci_6alkyl,
(4) -C3-6cycloalkyl,
(5) -O-Ci-6alkyl,
(6) -0(C=O)-C i-βalkyl,
(7) -NH-Ci-6alkyl,
(8) phenyl, (9) heterocycle,
(10) -CO2H, and
(11) -CN; or a pharmaceutically acceptable salt thereof.
2. The compound of Claim 1 of the formula Ia:
Figure imgf000045_0001
Ia or a pharmaceutically acceptable salt thereof.
3. The compound of Claim 1 of the formula Ib:
Figure imgf000045_0002
Ib or a pharmaceutically acceptable salt thereof.
4. The compound of Claim 1 of the formula Ic:
Figure imgf000045_0003
Ic or a pharmaceutically acceptable salt thereof.
5. The compound of Claim 1 wherein X is -SO2-.
6. The compound of Claim 1 wherein X is -(C=O)-.
7. The compound of Claim 1 wherein X is -(C=S)-.
8. The compound of Claim 1 wherein Rl is phenyl, which is unsubstituted or substituted with one or more of:
(1) halogen,
(2) hydroxyl,
(3) -On-Cj _6alkyl, where n is 0 or 1 (wherein if n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(4) -Ojtrphenyl. where the phenyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(5) -heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl 3,
(6) -NRIORI 1, wherein RlO and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Cl _6alkyl, which is unsubstituted or substituted with one or more substituents selected from R 13,
Figure imgf000046_0001
(8) -CO2H,
(9) -CN, and
(10) -NO2.
9. The compound of Claim 8 wherein Rl is phenyl, which is unsubstituted or substituted with one or more methyl, -CF3, halo, -OCF3, -OCH3, -OCH2CH3, -CO2CH3, -CN, - N(CH3), -NH(CH2CH3), -NO2, triazolyl or phenyl:
10. The compound of Claim 9 wherein Rl is phenyl, which is unsubstituted or substituted with one or more methyl, -CF3, fluro, -OCF3, -OCH3, -CO2CH3 or phenyl.
11. The compound of Claim 10 wherein Rl is selected from the group consisting of: (1) phenyl,
(2) biphenyl,
(3) 2,6-dimethoxyphenyl,
(4) 2,4-dichlorophenyl,
(5) 2,6-dichlorophenyl,
(6) 2,3-difluophenyl,
(7) 2,4-difluophenyl,
(8) 2,6-difluophenyl,
(9) 2-methoxy-4-methyl-phenyl,
(10) 3-methoxy-biphenyl,
(11) 3-methyl-biphenyl, and
(12) 5-methyl-2-triazolyl-phenyl.
12. The compound of Claim 11 wherein Rl is phenyl or 2,6-dimethoxyphenyl.
13. The compound of Claim 1 wherein R2 is heteroaryl, which is unsubstituted or substituted with one or more of:
(1) halogen,
(2) hydroxyl,
(3) -On-Ci_6alkyl, where n is 0 or 1 (wherein if n is 0, a bond is present) and where the alkyl is unsubstituted or substituted with one or more substituents selected from Rl 3,
(4) -On-phenyl, where the phenyl is unsubstituted or substituted with one or more substituents selected from Rl 3.
(5) -heterocycle, where the heterocycle is unsubstituted or substituted with one or more substituents selected from Rl 35
(6) -NRl ORI 1 , wherein RlO and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Cl -6alkyl, which is unsubstituted or substituted with one or more substituents selected from Rl 3,
Figure imgf000047_0001
(8) -CO2H,
(9) -CN, and
(10) -NO2.
14. The compound of Claim 13 wherein R2 is selected from the group consisting of:
( 1 ) benzimidazolyl,
(2) benzothiazolyl,
(3) benzoxazolyl,
(4) quinazolinyl,
(5) quinolinyl,
(6) thϊadiazolyl, which is unsubstituted or substituted with halogen, hydroxyl, Ci_6alkyl, -O-Ci-6alkyl or phenyl.
15. The compound of Claim 14 wherein R? is selected from the group consisting of: benzothiazolyl, which is unsubstituted or substituted with chloro, 6-chloro-benzothiazolyl, benzoxazolyl, quinazolinyl and quinolinyl.
16. The compound of Claim 1 wherein R3 and R4 are independently selected from the group consisting of:
(1) hydrogen,
(2) halogen,
(3) hydroxyl,
(4) -Cl-6alkyl,
(5) -O-Ci-6alkyl,
(6) -phenyl,
(7) -NRl ORI 1, wherein RlO and Rl 1 are independently selected from the group consisting of:
(a) hydrogen,
(b) Ci-6alkyl, or R3 and R4 taken together form C=O;
17. The compound of Claim 1 wherein R3 and R4 are selected from the following options:
R3 and R4 are hydrogen,
R3 is fluoro and R4 is hydrogen,
R3 is hydroxyl and R4 is hydrogen, R3 is -NH-Ci_6alkyl and R4 is hydrogen, R3 is -O-Ci_6alkyl and R4 is hydrogen, or R3 and R4 taken together form C=O.
18. A compound which is selected from the group consisting of: 6-chloro-2- [4-(2,6-dimethoxybenzoyI)- 1 ,4-diazepan- 1 -yl] - 1 ,3 -benzothiazole; 2-{4-[5-metiiyl-2-(2H-l523-triazol-2-yl)benzoyl3-l,4-diazepan-l-yl}-l,3-benzoxazole; 6-chloro-2-[4-(phenylsulfonyl)-l ,4-diazepan- 1 -yl]-l ,3 -benzothiazole; 2-[4-(2,6-dimethoxybenzoyl)-l,4-diazepan-l-yl]quinazoline;
2- {4-[5-methyl-2-(2H- 1 ,2,3-triazol-2-yl)benzoyl] -1 ,4-diazepan- 1 -yl }- 1 ,3 -benzoxazole; l-(l,3-benzoxazol-2-yl)-4-(2,6-dimethoxybenzoyl)-l,4-diazepan-6-ol;
1 -(1 ,3-benzoxazol-2-yl)-4-(2,6-dimethoxybenzoyl)-l ,4-diazepan-6-one;
1 -(1 ,3-benzoxazol-2-yl)-4-(2,6-dimethoxybenzoyl)-N-methyl- 1 ,4-diazepan-6-amine;
2-(4- { [5-methyl-2-(2H-l ,2,3-triazol-2-yl)phenyl]carbonothioyl}-l ,4-diazepan- 1 -yl)- 1 ,3- benzoxazole;
4-(biphenyl-2-ylcarbonyl)-l-(6-chloro-l,3-benzothiazol-2-yl)-l,4-diazepane; l-(6-chloro-l,3-benzothiazol-2-yl)-4-(2-methoxy-4-methylbenzoyl)-l,4-diazepane;
2-{4-[(2-methyl-5-phenyl-l,3-thiazol-4-yl)carbonyl]-l,4-diazepan-l-yl}-l,3-benzoxazole;
1 -(2,6-dimethoxybenzoyl)-4-(3 -phenyl- 1 ,2,4-thiadiazol-5-yl)- 1 ,4-diazepane;
2-[4-(236-dunethoxybenzoyl)-6-methoxy-l,4-diazepan-l-yl3-l,3-benzoxazole;
2- {4- [(3 -methoxybiphenyl-2-yl)carbonyl]- 1 ,4-diazepan- 1 -yl } - 1 ,3 -benzoxazole;
2-{4-[(3-methylbiphenyl-2-yl)carbonyl]-l,4-diazepan-l-yl}-l,3-benzoxazole;
2-{4-[5-methyl-2-(2H-l,2,3-triazol-2-yl)benzoyl]-l,4-diazepan-l-yl}quinazoline;
1 -(1 ,3-benzoxazol-2-yl)-4-[2-(ethylamino)benzoyl]- 1 ,4-diazepane;
4-(2,6-dimethoxybenzoyl)- 1 -quinolin-2-yl- 1 ,4-diazepane; or a pharmaceutically acceptable salt thereof.
19. A pharmaceutical composition which comprises an inert carrier and a compound of Claim 1 or a pharmaceutically acceptable salt thereof.
20. A compound of Claim 1 or a pharmaceutically acceptable salt thereof for use in medicine.
21. Use of a compound of Claim 1, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a sleep disorder.
22. A method for enhancing the quality of sleep in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
23. A method for treating insomnia in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
24. A method for treating or controlling obesity in a mammalian patient in need thereof which comprises administering to the patient a therapeutically effective amount of the compound of Claim 1 or a pharmaceutically acceptable salt thereof.
PCT/US2007/007738 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists WO2007126935A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA002647678A CA2647678A1 (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists
AU2007245037A AU2007245037A1 (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists
US12/225,506 US8685961B2 (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists
EP07754286.8A EP2001485B1 (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists
JP2009502989A JP2009531447A (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US78707006P 2006-03-29 2006-03-29
US60/787,070 2006-03-29

Publications (2)

Publication Number Publication Date
WO2007126935A2 true WO2007126935A2 (en) 2007-11-08
WO2007126935A3 WO2007126935A3 (en) 2008-08-07

Family

ID=38656057

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/007738 WO2007126935A2 (en) 2006-03-29 2007-03-27 Diazepan orexin receptor antagonists

Country Status (6)

Country Link
US (1) US8685961B2 (en)
EP (1) EP2001485B1 (en)
JP (1) JP2009531447A (en)
AU (1) AU2007245037A1 (en)
CA (1) CA2647678A1 (en)
WO (1) WO2007126935A2 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008069997A1 (en) * 2006-12-01 2008-06-12 Merck & Co., Inc. Substituted diazepan compounds as orexin receptor antagonists
WO2008147518A1 (en) 2007-05-23 2008-12-04 Merck & Co., Inc. Pyridyl piperidine orexin receptor antagonists
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
EP2214676A1 (en) * 2007-10-29 2010-08-11 Merck Sharp & Dohme Corp. Substituted diazepan orexin receptor antagonists
JP2010527924A (en) * 2007-05-18 2010-08-19 メルク・シャープ・エンド・ドーム・コーポレイション Oxo-bridged diazepan orexin receptor antagonist
JP2011500820A (en) * 2007-10-25 2011-01-06 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Diazepane compounds that regulate CB2 receptors
WO2011050198A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Disubstituted octahy - dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2011050200A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Fused heterocyclic compounds as orexin receptor modulators
WO2011050202A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Fused heterocyclic compounds as orexin receptor modulators
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
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
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
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
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
US8288373B2 (en) 2008-11-04 2012-10-16 Chemocentryx, Inc. Modulators of CXCR7
WO2012145581A1 (en) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Disubstituted octahy-dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2013169610A1 (en) * 2012-05-09 2013-11-14 Merck Sharp & Dohme Corp. Process for the preparation of an intermediate for an orexin receptor antagonist
US8618102B2 (en) 2006-07-14 2013-12-31 Merck Sharp & Dohme Corp. Bridged diazepan orexin receptor antagonists
US8853202B2 (en) 2008-11-04 2014-10-07 Chemocentryx, Inc. Modulators of CXCR7
US9156819B2 (en) 2011-10-19 2015-10-13 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
CN105461699A (en) * 2014-09-25 2016-04-06 广东东阳光药业有限公司 Substituted heterocyclic compound, and use method and use thereof
CN107021964A (en) * 2016-02-01 2017-08-08 广东东阳光药业有限公司 7-naphthyridine derivatives and application thereof
US9732077B2 (en) 2013-12-18 2017-08-15 Merck Sharp & Dohme Corp. Diazepane 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
WO2018146466A1 (en) 2017-02-09 2018-08-16 Benevolentai Bio Limited Orexin receptor antagonists
WO2018206959A1 (en) 2017-05-10 2018-11-15 Benevolentai Bio Limited Orexin receptor antagonists
WO2018206956A1 (en) 2017-05-10 2018-11-15 Benevolentai Bio Limited Orexin receptor antagonists
US10131653B2 (en) 2015-04-24 2018-11-20 Takeda Pharmaceutical Company Limited Heterocyclic compound
US10828302B2 (en) 2016-03-10 2020-11-10 Janssen Pharmaceutica Nv Methods of treating depression using orexin-2 receptor antagonists
US11464786B2 (en) 2018-12-12 2022-10-11 Chemocentryx, Inc. CXCR7 inhibitors for the treatment of cancer
US11834452B2 (en) 2012-11-29 2023-12-05 Chemocentryx, Inc. CXCR7 antagonists

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012506379A (en) * 2008-10-21 2012-03-15 メルク・シャープ・エンド・ドーム・コーポレイション Disubstituted azepanorexin receptor antagonist
EP2621492A1 (en) * 2010-10-01 2013-08-07 Indiana University Research and Technology Corporation Treatment of symptoms associated with menopause
US9108959B2 (en) 2011-03-03 2015-08-18 Merck Sharp & Dohme Corp. Process for the preparation of an orexin receptor antagonist
KR102123130B1 (en) 2012-05-31 2020-06-15 머크 샤프 앤드 돔 코포레이션 Solid dosage formulations of an orexin receptor antagonist
EP3134092B1 (en) 2014-04-21 2020-06-24 Merck Sharp & Dohme Corp. Pharmaceutical salts of an orexin receptor antagonist
WO2016065585A1 (en) 2014-10-30 2016-05-06 Merck Sharp & Dohme Corp. Piperidine isoxazole and isothiazole orexin receptor antagonists
WO2016065583A1 (en) 2014-10-30 2016-05-06 Merck Sharp & Dohme Corp. Oxazole orexin receptor antagonists
WO2016065587A1 (en) 2014-10-30 2016-05-06 Merck Sharp & Dohme Corp. Pyrazole orexin receptor antagonists
WO2016065584A1 (en) 2014-10-30 2016-05-06 Merck Sharp & Dohme Corp. Piperidine oxadiazole and thiadiazole orexin receptor antagonists
WO2016065586A1 (en) 2014-10-30 2016-05-06 Merck Sharp & Dohme Corp. Pyrazole, triazole and tetrazole orexin receptor antagonists
WO2016085784A1 (en) 2014-11-26 2016-06-02 Merck Sharp & Dohme Corp. Methyl diazepane orexin receptor antagonists
US9994570B2 (en) 2014-11-26 2018-06-12 Merck Sharp & Dohme Corp. Bridged diazepane orexin receptor antagonists
WO2016086358A1 (en) 2014-12-02 2016-06-09 Merck Sharp & Dohme Corp. Hydroxymethyl piperidine orexin receptor antagonists
WO2016086357A1 (en) 2014-12-02 2016-06-09 Merck Sharp & Dohme Corp. Methyl oxazole orexin receptor antagonists
WO2016100161A1 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. Ethyldiamine orexin receptor antagonists
US9987255B2 (en) 2014-12-19 2018-06-05 Merck Sharp & Dohme Corp. 5,5-bicyclic oxazole orexin receptor antagonists
WO2016095204A1 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. Pyrrolidine orexin receptor antagonists
WO2016100157A2 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. 6,5-bicyclic octahydropyrrolopyridine orexin receptor antagonists
WO2016095205A1 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. Heteroaryl orexin receptor antagonists
WO2016101119A1 (en) 2014-12-23 2016-06-30 Merck Sharp & Dohme Corp. Fused heteroaryl derivatives as orexin receptor antagonists
WO2016101118A1 (en) 2014-12-23 2016-06-30 Merck Sharp & Dohme Corp. Amidoethyl azole orexin receptor antagonists
WO2017012502A1 (en) 2015-07-17 2017-01-26 Sunshine Lake Pharma Co., Ltd. Substituted quinazoline compounds and preparation and uses thereof
SG11201804223TA (en) 2015-11-23 2018-06-28 Sunshine Lake Pharma Co Ltd OCTAHYDROPYRROLO [3, 4-c] PYRROLE DERIVATIVES AND USES THEREOF

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS4966691A (en) * 1972-10-30 1974-06-27
US4093726A (en) * 1976-12-02 1978-06-06 Abbott Laboratories N-(2-benzimidazolyl)-piperazines
US4171361A (en) * 1977-03-31 1979-10-16 Eli Lilly And Company 1-Substituted-3-amino-6,7-dialkoxy-1H-1,2,4-benzothiadiazine-1-oxides
US4426382A (en) 1980-02-13 1984-01-17 Sankyo Company Limited 4-Amino-6,7-dimethoxy-2-piperazinylquinazoline derivatives, their preparation and use
DE3121137A1 (en) * 1981-05-27 1982-12-16 Hoechst Ag, 6000 Frankfurt Novel pyridazino[4,5-b]indoles, processes for their preparation, their use and also pharmaceutical preparations based on these compounds, intermediates and their preparation
US5001125A (en) * 1984-03-26 1991-03-19 Janssen Pharmaceutica N.V. Anti-virally active pyridazinamines
JPH0647540B2 (en) * 1985-07-16 1994-06-22 エーザイ株式会社 Ischemic heart disease / arrhythmia treatment / prevention agent
HU195314B (en) 1985-07-23 1988-04-28 Villamos Ipari Kutato Intezet Cooling insert for equipments carrying out heat and material exchange being between gaseous medium and fluid particularly for cooling towers and degasing units
US6706705B1 (en) 1994-11-25 2004-03-16 Nippon Chemiphar Co., Ltd. Quinazoline derivatives
AU2319100A (en) * 1999-01-28 2000-08-18 Nippon Shinyaku Co. Ltd. Amide derivatives and drug compositions
AU1887401A (en) * 1999-12-14 2001-06-25 Nippon Shinyaku Co. Ltd. Medicinal composition
JP2001261657A (en) * 2000-03-17 2001-09-26 Yamanouchi Pharmaceut Co Ltd Cyanophenyl derivative
AU2002341123A1 (en) 2001-05-05 2002-11-18 Smithkline Beecham P.L.C. N-aroyl cyclic amine derivatives as orexin receptor antagonists
GB0130393D0 (en) 2001-12-19 2002-02-06 Smithkline Beecham Plc Compounds
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
JP4637089B2 (en) 2003-03-26 2011-02-23 アクテリオン ファーマシューティカルズ リミテッド Acetamide derivatives
WO2004092117A1 (en) * 2003-04-07 2004-10-28 Kalypsys, Inc. Para-sulfonyl substituted phenyl compounds as modulators of ppars
CA2520839A1 (en) 2003-04-28 2004-11-11 Actelion Pharmaceuticals Ltd Quinoxalinone-3- one derivatives as orexin receptor antagonists
AR048523A1 (en) 2004-04-07 2006-05-03 Kalypsys Inc COMPOUNDS WITH ARIL SULFONAMIDE AND SULFONYL STRUCTURE AS PPAR MODULATORS AND METHODS TO TREAT METABOLIC DISORDERS
US20060258672A1 (en) 2005-05-13 2006-11-16 Joseph Barbosa Multicyclic compounds and methods of their use
CA2632027A1 (en) 2005-12-14 2007-06-21 Amgen Inc. Diaza heterocyclic sulfonamide derivatives and their uses

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2001485A4 *

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8618102B2 (en) 2006-07-14 2013-12-31 Merck Sharp & Dohme Corp. Bridged diazepan orexin receptor antagonists
EP2392572A1 (en) * 2006-12-01 2011-12-07 Merck Sharp & Dohme Corp. Substituted diazepan compounds as orexin receptor antagonists
US7951797B2 (en) 2006-12-01 2011-05-31 Merck Sharp & Dohme Corp. Substituted diazepan orexin receptor antagonists
WO2008069997A1 (en) * 2006-12-01 2008-06-12 Merck & Co., Inc. Substituted diazepan compounds as orexin receptor antagonists
AU2007328267B2 (en) * 2006-12-01 2011-03-24 Merck Sharp & Dohme Llc Substituted diazepan compounds as orexin receptor antagonists
JP2010527924A (en) * 2007-05-18 2010-08-19 メルク・シャープ・エンド・ドーム・コーポレイション Oxo-bridged diazepan orexin receptor antagonist
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
JP2011500820A (en) * 2007-10-25 2011-01-06 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Diazepane compounds that regulate CB2 receptors
EP2214676A4 (en) * 2007-10-29 2011-08-03 Merck Sharp & Dohme Substituted diazepan orexin receptor antagonists
EP2214676A1 (en) * 2007-10-29 2010-08-11 Merck Sharp & Dohme Corp. Substituted diazepan orexin receptor antagonists
US8362009B2 (en) 2007-10-29 2013-01-29 Merck Sharp & Dohme Corp. Substituted diazepan orexin receptor antagonists
US8288373B2 (en) 2008-11-04 2012-10-16 Chemocentryx, Inc. Modulators of CXCR7
US8853202B2 (en) 2008-11-04 2014-10-07 Chemocentryx, Inc. Modulators of CXCR7
US8653263B2 (en) 2009-10-23 2014-02-18 Janssen Pharmaceutica Disubstituted octahydropyrrolo[3,4-c]pyrroles as orexin receptor modulators
EP3093291A1 (en) 2009-10-23 2016-11-16 Janssen Pharmaceutica N.V. Disubstituted octahy - dropyrrolo [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
USRE48841E1 (en) 2009-10-23 2021-12-07 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-c]pyrroles 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
EP3581575A1 (en) 2009-10-23 2019-12-18 Janssen Pharmaceutica NV Disubstituted octahydropyrrolo[3,4-c]pyrroles as orexin receptor modulators
US9079911B2 (en) 2009-10-23 2015-07-14 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-c]pyrroles as orexin receptor modulators
US9062044B2 (en) 2009-10-23 2015-06-23 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo[3,4-c]pyrroles as orexin receptor modulators
WO2011050202A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Fused heterocyclic compounds as orexin receptor modulators
WO2011050200A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Fused heterocyclic compounds as orexin receptor modulators
US8680275B2 (en) 2009-10-23 2014-03-25 Janssen Pharmaceutica Nv Fused heterocyclic compounds as orexin receptor modulators
WO2011050198A1 (en) 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Disubstituted octahy - dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
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
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
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
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
US9586962B2 (en) 2011-04-20 2017-03-07 Janssen Pharmaceutica Nv Disubstituted octahydropyrrolo [3,4-C] pyrroles as orexin receptor modulators
WO2012145581A1 (en) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Disubstituted octahy-dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
US9156819B2 (en) 2011-10-19 2015-10-13 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
US9441254B2 (en) 2012-05-09 2016-09-13 Merck Sharp & Dohme Corp. Process for the preparation of an intermediate for an orexin receptor antagonist
WO2013169610A1 (en) * 2012-05-09 2013-11-14 Merck Sharp & Dohme Corp. Process for the preparation of an intermediate for an orexin receptor antagonist
US11834452B2 (en) 2012-11-29 2023-12-05 Chemocentryx, Inc. CXCR7 antagonists
US9732077B2 (en) 2013-12-18 2017-08-15 Merck Sharp & Dohme Corp. Diazepane orexin receptor antagonists
CN105461699B (en) * 2014-09-25 2019-07-09 广东东阳光药业有限公司 Substituted heterocyclic compound and its application method and purposes
CN105461699A (en) * 2014-09-25 2016-04-06 广东东阳光药业有限公司 Substituted heterocyclic compound, and use method and use thereof
US10131653B2 (en) 2015-04-24 2018-11-20 Takeda Pharmaceutical Company Limited Heterocyclic compound
CN107021964A (en) * 2016-02-01 2017-08-08 广东东阳光药业有限公司 7-naphthyridine derivatives and application thereof
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
WO2018146466A1 (en) 2017-02-09 2018-08-16 Benevolentai Bio Limited Orexin receptor antagonists
WO2018206956A1 (en) 2017-05-10 2018-11-15 Benevolentai Bio Limited Orexin receptor antagonists
WO2018206959A1 (en) 2017-05-10 2018-11-15 Benevolentai Bio Limited Orexin receptor antagonists
US11464786B2 (en) 2018-12-12 2022-10-11 Chemocentryx, Inc. CXCR7 inhibitors for the treatment of cancer

Also Published As

Publication number Publication date
EP2001485A4 (en) 2010-11-03
WO2007126935A3 (en) 2008-08-07
EP2001485A2 (en) 2008-12-17
JP2009531447A (en) 2009-09-03
US8685961B2 (en) 2014-04-01
EP2001485B1 (en) 2015-09-09
AU2007245037A1 (en) 2007-11-08
CA2647678A1 (en) 2007-11-08
US20090124603A1 (en) 2009-05-14

Similar Documents

Publication Publication Date Title
EP2001485B1 (en) Diazepan orexin receptor antagonists
AU2007272855B2 (en) Substituted diazepan orexin receptor antagonists
EP2089382B1 (en) Substituted diazepan compounds as orexin receptor antagonists
AU2007272854B2 (en) Bridged diazepan orexin receptor antagonists
US8263586B2 (en) OXO bridged diazepan orexin receptor antagonists
EP2150115B1 (en) Cyclopropyl pyrrolidine orexin receptor antagonists
EP2214676B1 (en) Substituted diazepan orexin receptor antagonists
WO2008008551A2 (en) 2-substituted proline bis-amide orexin receptor antagonists
EP2349270A1 (en) 2,5-disubstituted morpholine orexin receptor antagonists
EP1922071A2 (en) Diazaspirodecane orexin receptor antagonists
WO2007126934A2 (en) Amidoethylthioether orexin receptor antagonists
WO2010048014A1 (en) 2,4-disubstituted pyrrolidine orexin receptor antagonists
WO2010048017A1 (en) Disubstituted azepan orexin receptor antagonists
EP2350066A1 (en) 2,5-disubstituted piperidine orexin receptor antagonists
WO2010048016A1 (en) 2,3-disubstituted piperidine orexin receptor antagonists
WO2006110626A1 (en) Amidopropoxyphenyl orexin receptor antagonists
WO2009011775A1 (en) Amidoethyl alkylamino orexin receptor antagonists

Legal Events

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

Ref document number: 07754286

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2007245037

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 12225506

Country of ref document: US

Ref document number: 2007754286

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009502989

Country of ref document: JP

Ref document number: 2647678

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007245037

Country of ref document: AU

Date of ref document: 20070327

Kind code of ref document: A