WO2012044613A1 - Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors - Google Patents

Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors Download PDF

Info

Publication number
WO2012044613A1
WO2012044613A1 PCT/US2011/053442 US2011053442W WO2012044613A1 WO 2012044613 A1 WO2012044613 A1 WO 2012044613A1 US 2011053442 W US2011053442 W US 2011053442W WO 2012044613 A1 WO2012044613 A1 WO 2012044613A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
compound
trifluoromethyl
group
formula
Prior art date
Application number
PCT/US2011/053442
Other languages
French (fr)
Inventor
Peter J. Connolly
Mark J. Macielag
Mark E. Mcdonnell
Bin Zhu
Original Assignee
Janssen Pharmaceutica Nv
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 Janssen Pharmaceutica Nv filed Critical Janssen Pharmaceutica Nv
Priority to BR112013007287A priority Critical patent/BR112013007287A2/en
Priority to CA2810077A priority patent/CA2810077A1/en
Priority to KR1020137010278A priority patent/KR20140001206A/en
Priority to EP11771300.8A priority patent/EP2621918A1/en
Priority to RU2013114852/04A priority patent/RU2013114852A/en
Priority to JP2013530418A priority patent/JP2013537920A/en
Priority to AU2011307252A priority patent/AU2011307252B2/en
Priority to CN2011800423053A priority patent/CN103080103A/en
Publication of WO2012044613A1 publication Critical patent/WO2012044613A1/en

Links

Classifications

    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings

Definitions

  • Cannabis sativa has been used for the treatment of pain for many years.
  • a 9 - tetrahydrocannabinol is a major active ingredient from Cannabis sativa and an agonist of cannabinoid receptors (Pertwee, Brit J Pharmacol, 2008, 153, 199-215).
  • Two cannabinoid G protein-coupled receptors have been cloned, cannabinoid receptor type 1 (CBi Matsuda et al, Nature, 1990, 346, 561-4) and cannabinoid receptor type 2 (CB 2 Munro et al, Nature, 1993, 365, 61-5).
  • CBi is expressed centrally in brain areas such as, the hypothalamus and nucleus accumbens, as well as peripherally in the liver, gastrointestinal tract, pancreas, adipose tissue, and skeletal muscle (Di Marzo et al, Curr Opin Lipidoi, 2007, 18, 129-140).
  • CB 2 is predominantly expressed in immune cells such as, monocytes (Pacher et al, Amer J Physiol, 2008, 294, HI 133-Hl 134), and under certain conditions, also in the brain (Benito et al, Brit J Pharmacol, 2008, 153, 277-285) and in skeletal (Cavuoto et al, Biochem Biophys Res Commun, 2007, 364, 105-1 10) and cardiac (Hajrasouliha et al, Eur J Pharmacol, 2008, 579, 246-252) muscle.
  • An abundance of pharmacological, anatomical and electrophysiological data, using synthetic agonists, indicate that increased cannabinoid signaling through
  • CB ! /CB 2 promotes analgesia in tests of acute nociception and suppresses hyperalgesia in models of chronic neuropathic and inflammatory pain (Cravatt et al, J Neurobiol, 2004, 61, 149-60; Guindon et al , Brit J Pharmacol, 2008, 153 , 319-334).
  • AEA is hydrolyzed by fatty acid amide hydrolase (FAAH) and 2- AG is hydrolyzed by monoacylglycerol lipase (MGL) (Piomelli, Nat Rev Neurosci, 2003, 4, 873-884).
  • FAAH fatty acid amide hydrolase
  • MDL monoacylglycerol lipase
  • endocannabinoid-mediated antinociceptive tone is demonstrated by the formation of AEA in the periaqueductal grey following noxious stimulation in the periphery (Walker et al, Proc Natl Acad Sci USA, 1999, 96, 12198-203) and, conversely, by the induction of hyperalgesia following antisense RNA-mediated inhibition of CBi in the spinal cord (Dogrul et al., Pain, 2002, 100, 203-9).
  • MGL inhibitors are potentially useful for the treatment of pain, inflammation, and CNS disorders (Di Marzo et al, Curr Pharm Des, 2000, 6, 1361-80; Jhaveri et al, Brit J Pharmacol, 2007, 152, 624-632; McCarberg Bill et al, Amer J Ther, 2007, 14, 475- 83), as well as glaucoma and disease states arising from elevated intraocular pressure
  • the present invention is directed to a compound of Formula (la)
  • Y is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of fluoro, chloro, Ci_ 4 alkyl, cyano, and trifluoromethyl;
  • benzothiazolyl benzothienyl, indazolyl, and indolyl, or iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is
  • no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro; n is 1 or 2; and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
  • the present invention is further directed to a compound of Formula (lb)
  • a heteroaryl selected from the group consisting of thiazolyl, oxazolyl, pyridinyl, and pyrimidinyl,
  • Y b is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of fluoro, chloro, Ci- 4 alkyl, Ci_ 4 alkoxy, cyano, and trifluoromethyl;
  • a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl, or iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, C 1-4 alkyl, Ci_ 4 alkoxy, and trifluoromethyl, wherein the Ce-w aryl and the heteroaryl of Z b are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci- 4 alkyl, Ci
  • no more than one substituent of Z b is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro; and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
  • the present invention also provides, inter alia, a pharmaceutical composition
  • a pharmaceutical composition comprising, consisting of and/or consisting essentially of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and/or a pharmaceutically acceptable diluent, and a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof.
  • a pharmaceutical composition comprising, consisting of, and/or consisting essentially of admixing a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof, and a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and/or a pharmaceutically acceptable diluent.
  • the present invention further provides, inter alia, methods for treating or ameliorating a MGL-modulated disorder in a subject, including a human or other mammal in which the disease, syndrome, or condition is affected by the modulation of the MGL enzyme such as pain, and the diseases that lead to such pain, inflammation, and CNS disorders, using a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof.
  • the present invention also provides, inter alia, methods for producing the instant compounds and pharmaceutical compositions and medicaments thereof.
  • alkyl refers to straight and branched carbon chains having 1 to 8 carbon atoms. Therefore, designated numbers of carbon atoms (e.g., C 1-8 ) refer independently to the number of carbon atoms in an alkyl moiety or to the alkyl portion of a larger alkyl-containing substituent. In substituent groups with multiple alkyl groups such as, (Ci_ 6 alkyl) 2 amino-, the groups of the dialkylamino may be the same or different.
  • alkoxy refers to an -O-alkyl group, wherein the term “alkyl” is as defined above.
  • alkenyl and alkynyl refer to straight and branched carbon chains having 2 or more carbon atoms, wherein an alkenyl chain contains at least one double bond and an alkynyl chain contains at least one triple bond.
  • cycloalkyl refers to saturated or partially saturated, monocyclic or polycyclic hydrocarbon rings of 3 to 14 carbon atoms. Examples of such rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and adamantyl.
  • benzo-fused cycloalkyl refers to a 5- to 8- membered monocyclic cycloalkyl ring fused to a benzene ring.
  • the carbon atom ring members that form the cycloalkyl ring may be fully saturated or partially saturated.
  • heterocyclyl refers to a nonaromatic monocyclic or bicyclic ring system having 3 to 10 ring members that include at least 1 carbon atom and from 1 to 4 heteroatoms independently selected from N, O, and S. Included within the term heterocyclyl is a nonaromatic cyclic ring of 5 to 7 members in which 1 to 2 members are N, or a nonaromatic cyclic ring of 5 to 7 members in which 0, 1 or 2 members are N and up to 2 members are O or S and at least one member must be either N, O, or S; wherein, optionally, the ring contains 0 to 1 unsaturated bonds, and, optionally, when the ring is of 6 or 7 members, it contains up to 2 unsaturated bonds.
  • the carbon atom ring members that form a heterocycle ring may be fully saturated or partially saturated.
  • heterocyclyl also includes two 5 membered monocyclic heterocycloalkyl groups bridged to form a bicyclic ring. Such groups are not considered to be fully aromatic and are not referred to as heteroaryl groups.
  • heterocycle is bicyclic, both rings of the heterocycle are non-aromatic and at least one of the rings contains a heteroatom ring member.
  • heterocycle groups include, and are not limited to, pyrrolinyl (including 2H-pyrrole, 2-pyrrolinyl or 3-pyrrolinyl), pyrrolidinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl. Unless otherwise noted, the heterocycle is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • benzo-fused heterocyclyl refers to a 5 to 7 membered monocyclic heterocycle ring fused to a benzene ring.
  • the heterocycle ring contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S.
  • the carbon atom ring members that form the heterocycle ring may be fully saturated or partially saturated.
  • benzo-fused heterocycle ring is attached to its pendant group at a carbon atom of the benzene ring.
  • aryl refers to an unsaturated, aromatic monocyclic or bicyclic ring of 6 to 10 carbon members. Examples of aryl rings include phenyl and naphthalenyl.
  • heteroaryl refers to an aromatic monocyclic or bicyclic aromatic ring system having 5 to 10 ring members and which contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S. Included within the term heteroaryl are aromatic rings of 5 or 6 members wherein the ring consists of carbon atoms and has at least one heteroatom member. Suitable heteroatoms include N, O, and S. In the case of 5 membered rings, the heteroaryl ring preferably contains one member of N, O or S and, in addition, up to 3 additional N atoms. In the case of 6 membered rings, the heteroaryl ring preferably contains from 1 to 3 nitrogen atoms.
  • heteroaryl is bicyclic, at least one heteroatom is present in each ring.
  • heteroaryl groups include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl and pyrazinyl.
  • the heteroaryl is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • benzo fused heteroaryl refers to a 5 to 6 membered monocyclic heteroaryl ring fused to a benzene ring.
  • the heteroaryl ring contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S.
  • heteroaryl groups with the optionally fused benzene rings include indolyl, isoindolyl, benzofuryl, benzothienyl, indazolyl, benzimidazolyl,
  • benzothiazolyl benzoxazolyl, benzisoxazolyl, benzothiadiazolyl, benzotriazolyl, quinolinyl, isoquinolinyl and quinazolinyl.
  • the benzo-fused heteroaryl is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • halogen refers to fluorine, chlorine, bromine and iodine atoms.
  • alkyl or aryl or either of their prefix roots appear in a name of a substituent (e.g., arylalkyl, alkylamino) the name is to be interpreted as including those limitations given above for "alkyl” and "aryl.”
  • Designated numbers of carbon atoms e.g., Ci-Ce refer independently to the number of carbon atoms in an alkyl moiety, an aryl moiety, or in the alkyl portion of a larger substituent in which alkyl appears as its prefix root.
  • the designated number of carbon atoms includes all of the independent members included within a given range specified.
  • C e alkyl would include methyl, ethyl, propyl, butyl, pentyl and hexyl individually as well as sub-combinations thereof (e.g., C 1-2 , Ci_ 3, Ci_4, Ci_5 ; C 2- 6, C3_6, C 4 _6, C5.6, C 2- 5 ; etc.).
  • a "Ci-Ce alkylcarbonyl" substituent refers to a group of the formula:
  • R at a stereocenter designates that the stereocenter is purely of the R-configuration as defined in the art; likewise, the term “S” means that the stereocenter is purely of the ⁇ -configuration.
  • S means that the stereocenter is purely of the ⁇ -configuration.
  • the terms “*R” or “*S” at a stereocenter are used to designate that the stereocenter is of pure but unknown configuration.
  • RS refers to a stereocenter that exists as a mixture of the R- and ⁇ -configurations.
  • *RS or “*SR” refer to a stereocenter that exists as a mixture of the R- and ⁇ -configurations and is of unknown configuration relative to another stereocenter within the molecule.
  • Unlabeled stereocenters drawn without stereo bond designations are a mixture of the R- and ⁇ -configurations.
  • the absolute stereochemistry is as depicted.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount refers to an amount of an active compound or pharmaceutical agent, including a compound of the present invention, which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation or partial alleviation of the symptoms of the disease, syndrome, condition, or disorder being treated.
  • composition refers to a product that includes the specified ingredients in therapeutically effective amounts, as well as any product that results, directly, or indirectly, from combinations of the specified ingredients in the specified amounts.
  • MGL inhibitor is intended to encompass a compound that interacts with MGL to substantially reduce or eliminate its catalytic activity, thereby increasing the concentrations of its substrate(s).
  • MGL-modulated is used to refer to the condition of being affected by the modulation of the MGL enzyme including the condition of being affected by the inhibition of the MGL enzyme such as pain, and the diseases that lead to such pain, inflammation and CNS disorders.
  • the term "affect" or “affected” when referring to a disease, syndrome, condition or disorder that is affected by inhibition of MGL shall include a reduction in the frequency and / or severity of one or more symptoms or manifestations of said disease, syndrome, condition or disorder; and / or include the prevention of the development of one or more symptoms or manifestations of said disease, syndrome, condition or disorder or the development of the disease, condition, syndrome or disorder.
  • the compounds of Formula (la) and (lb) are useful in methods for treating, ameliorating and / or preventing a disease, a syndrome, a condition or a disorder that is affected by the inhibition of MGL.
  • Such methods comprise, consist of and/or consist essentially of administering to a subject, including an animal, a mammal, and a human in need of such treatment, amelioration and / or prevention, a therapeutically effective amount of a compound of Formula (la) or (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
  • the compounds of Formula (la) and (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof are useful for treating, ameliorating and / or preventing pain; diseases, syndromes, conditions, or disorders causing such pain; inflammation and / or CNS disorders.
  • the compounds of Formula (la) and (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof are useful for treating, ameliorating and / or preventing inflammatory pain, inflammatory hypersensitivity conditions and / or neuropathic pain, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (la) or (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof as herein defined.
  • inflammatory pain examples include pain due to a disease, condition, syndrome, disorder, or a pain state including inflammatory bowel disease, visceral pain, migraine, post operative pain, osteoarthritis, rheumatoid arthritis, back pain, lower back pain, joint pain, abdominal pain, chest pain, labor, musculoskeletal diseases, skin diseases, toothache, pyresis, burn, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection, rhinitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome, cholecystitis, pancreatitis, postmastectomy pain syndrome, menstrual pain, endometriosis, pain due to physical trauma, headache, sinus headache, tension headache, or arachnoiditis.
  • One type of inflammatory pain is inflammatory hyperalgesia /
  • inflammatory hyperalgesia examples include a disease, syndrome, condition, disorder, or pain state including inflammation, osteoarthritis, rheumatoid arthritis, back pain, joint pain, abdominal pain, musculoskeletal diseases, skin diseases, post operative pain, headaches, toothache, burn, sunburn, insect sting, neurogenic bladder, urinary incontinence, interstitial cystitis, urinary tract infection, cough, asthma, chronic obstructive pulmonary disease, rhinitis, contact
  • dermatitis/hypersensitivity itch, eczema, pharyngitis, enteritis, irritable bowel syndrome, inflammatory bowel diseases including Crohn's Disease, ulcerative colitis, urinary incontinence, benign prostatic hypertrophy, cough, asthma, rhinitis, nasal hypersensitivity, itch, contact dermatitis and / or dermal allegy and chronic obstructive pulmonary disease.
  • the present invention is directed to a method for treating, ameliorating and / or preventing inflammatory visceral hyperalgesia in which a enhanced visceral irritability exists, comprising, consisting of, and/or consisting essentially of the step of administering to a subject in need of such treatment a therapeutically effective amount of a compound, salt or solvate of Formula (la) or (lb).
  • the present invention is directed to a method for treating inflammatory somatic hyperalgesia in which a hypersensitivity to thermal, mechanical and/or chemical stimuli exists, comprising administering to a mammal in need of such treatment a therapeutically effective amount of a compound of Formula (la) or (lb) or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
  • a further embodiment of the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic pain. Examples of a
  • neuropathic pain include pain due to a disease, syndrome, condition, disorder, or pain state including cancer, neurological disorders, spine and peripheral nerve surgery, brain tumor, traumatic brain injury (TBI), spinal cord trauma, chronic pain syndrome, fibromyalgia, chronic fatigue syndrome, lupus, sarcoidosis, peripheral neuropathy, bilateral peripheral neuropathy, diabetic neuropathy, central pain, neuropathies associated with spinal cord injury, stroke, amyotrophic lateral sclerosis (ALS), Parkinson's disease, multiple sclerosis, sciatic neuritis, mandibular joint neuralgia, peripheral neuritis, polyneuritis, stump pain, phantom limb pain, bony fractures, oral neuropathic pain, Charcot's pain, complex regional pain syndrome I and II (CRPS I/II), radiculopathy, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis
  • neuropathic cold allodynia which can be characterized by the presence of a neuropathy-associated allodynic state in which a hypersensitivity to cooling stimuli exists.
  • neuropathic cold allodynia include allodynia due to a disease, condition, syndrome, disorder or pain state including neuropathic pain (neuralgia), pain arising from spine and peripheral nerve surgery or trauma, traumatic brain injury (TBI), trigeminal neuralgia, postherpetic neuralgia, causalgia, peripheral neuropathy, diabetic neuropathy, central pain, stroke, peripheral neuritis, polyneuritis, complex regional pain syndrome I and II (CRPS I/II) and radiculopathy.
  • neuropathic pain neuralgia
  • TBI traumatic brain injury
  • trigeminal neuralgia postherpetic neuralgia
  • causalgia peripheral neuropathy
  • diabetic neuropathy central pain
  • stroke peripheral neuritis
  • polyneuritis complex regional pain syndrome I and II
  • the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic cold allodynia in which a hypersensitivity to a cooling stimuli exists, comprising, consisting of, and/or consisting essentially of the step of administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula (la) or (lb) or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
  • the present invention is directed to a method for treating, ameliorating and / or preventing CNS disorders.
  • CNS disorders include anxieties such as, social anxiety, post-traumatic stress disorder, phobias, social phobia, special phobias, panic disorder, obsessive-compulsive disorder, acute stress disorder, separation anxiety disorder, and generalized anxiety disorder, as well as depression such as, major depression, bipolar disorder, seasonal affective disorder, post natal depression, manic depression, and bipolar depression.
  • Embodiments of the present invention include a compound of Formula (la)
  • Y is thiazolyl or pyrimidinyl
  • Ce-w aryl and heteroaryl of Z are optionally independently substituted with one to two substitutents selected from the group consisting of chloro, fluoro, Ci- 4 alkyl, trifluoromethyl, and phenyl,
  • Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
  • heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
  • An embodiment of the present invention includes a compound of Formula (la)
  • Y is ii) a heteroaryl that is thiazolyl or pyrimidinyl
  • Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is selected from the group consisting of chloro, fluoro, Ci- 4 alkyl, trifluoromethyl, and phenyl;
  • a further embodiment of the present invention includes a compound of Formula (la)
  • Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, C 1-4 alkyl, trifluoromethyl, and phenyl,
  • n 1 ; and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
  • An embodiment of the present invention includes a compound of Formula (la)
  • Y is thiazolyl or pyrimidinyl
  • Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl, wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
  • n 1 or 2;
  • An embodiment of the present invention includes a compound of Formula (la)
  • Y is thiazolyl or pyrimidinyl
  • Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
  • heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
  • n 1;
  • a further embodiment of the present invention includes a compound of Formula (la)
  • Ce-io aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, Ci- 4 alkyl, trifluoromethyl, and phenyl,
  • n 2; and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
  • the present invention is ound of Formula (la)
  • Embodiments of the present invention are directed to a compound of Formula
  • Yb is thiazolyl or pyrimidinyl
  • Zb is a heteroaryl selected from the group consisting of benzoxazolyl
  • heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl,
  • Zb is benzothienyl or indolyl
  • Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl
  • Yb is thiazolyl or pyrimidinyl
  • Zb is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
  • heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl,
  • Yb is thiazolyl or pyrimidinyl
  • Zb is a heteroaryl selected from the group consisting of benzothienyl and indolyl;
  • heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl
  • An embodiment of the present invention is directed to a compound of Formula
  • salts of compounds of Formula (la) and (lb) refer to nontoxic "pharmaceutically acceptable salts.” Other salts may, however, be useful in the preparation of compounds of Formula (la) and (lb) or of their pharmaceutically acceptable salts thereof.
  • Suitable pharmaceutically acceptable salts of compounds of Formula (la) and (lb) include acid addition salts that can, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable acid such as, hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts such as, sodium or potassium salts; alkaline earth metal salts such as, calcium or magnesium salts; and salts formed with suitable organic ligands such as, quaternary ammonium salts.
  • representative pharmaceutically acceptable salts include acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate,
  • methanesulfonic acid naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, 1- hydroxy -2 -naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, phosphoric acid, L-pyroglutamic acid, salicylic acid, 4- amino-salicylic acid, sebaic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid; and bases including ammonia, L-arginine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)-ethanol, ethanolamine, ethylened
  • prodrugs will be functional derivatives of the compounds that are readily convertible in vivo into the required compound.
  • administering encompasses the treatment or prevention of the various diseases, conditions, syndromes and disorders described with the compound specifically disclosed or with a compound that may not be specifically disclosed, but which converts to the specified compound in vivo after administration to a patient.
  • the compounds according to embodiments of this invention may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. The skilled artisan will understand that the term compound as used herein, is meant to include solvated compounds of Formula (la) and (lb).
  • the processes for the preparation of the compounds according to certain embodiments of the invention give rise to mixture of stereoisomers
  • these isomers may be separated by conventional techniques such as, preparative chromatography.
  • the compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution.
  • the compounds may, for example, be resolved into their component enantiomers by standard techniques such as, the formation of diastereomeric pairs by salt formation with an optically active acid such as, (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base.
  • the compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.
  • compositions including a pharmaceutical composition, comprising, consisting of, and/or consisting essentially of the (+)-enantiomer of a compound of Formula (la) or (lb) wherein said composition is substantially free from the (-)-isomer of said compound.
  • substantially free means less than about 25 %, preferably less than about 10 %, more preferably less than about 5 %, even more preferably less than about 2 % and even more preferably less than about 1 % of the (-)-isomer calculated as. (mass (+) - enantiomer)
  • compositions including a pharmaceutical composition, comprising, consisting of, and consisting essentially of the (-)-enantiomer of a compound of Formula (la) or (lb) wherein said composition is substantially free from the (+)-isomer of said compound.
  • substantially free from means less than about 25 %, preferably less than about 10 %, more preferably less than about 5 %, even more preferably less than about 2 % and even more preferably less than about 1 % of the (+)-isomer calculated as
  • any of the processes for preparation of the compounds of the various embodiments of the present invention it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups such as those described in Protective Groups in Organic Chemistry, Second Edition, J.F.W. McOmie, Plenum Press, 1973; T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 ; and T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, 1999.
  • the protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • compositions comprising compounds of Formula (la) or (lb) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and/or pharmaceutically acceptable diluent.
  • the compounds of Formula (la) and (lb) may be admixed with any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilizing agent(s), and combinations thereof.
  • Solid oral dosage forms such as, tablets or capsules, containing the compounds of the present invention may be administered in at least one dosage form at a time, as appropriate. It is also possible to administer the compounds in sustained release formulations.
  • Additional oral forms in which the present inventive compounds may be administered include elixirs, solutions, syrups, and suspensions; each optionally containing flavoring agents and coloring agents.
  • compounds of Formula (la) and (lb) can be administered by inhalation (intratracheal or intranasal) or in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • inhalation intratracheal or intranasal
  • a suppository or pessary or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • they can be incorporated into a cream comprising, consisting of, and/or consisting essentially of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • An alternative means of administration includes transdermal administration by using a skin or transdermal patch.
  • compositions of the present invention can also be injected parenterally, for example, intracavernosally, intravenously, intramuscularly, subcutaneously, intradermally, or intrathecally.
  • the compositions will also include at least one of a suitable carrier, a suitable excipient, and a suitable diluent.
  • compositions of the present invention are best used in the form of a sterile aqueous solution that may contain other substances, for example, enough salts and monosaccharides to make the solution isotonic with blood.
  • compositions of the present invention may be administered in the form of tablets or lozenges, which can be formulated in a conventional manner.
  • compositions containing at least one of the compounds of Formula (la) or (lb) as the active ingredient can be prepared by mixing the compound(s) with a pharmaceutically acceptable carrier, a pharmaceutically acceptable diluent, and/or a pharmaceutically acceptable excipient according to conventional pharmaceutical compounding techniques.
  • a pharmaceutically acceptable carrier e.g., benzyl alcohol, benzyl ether, benzyl ether, benzyl ether, sulfate, a pharmaceutically acceptable excipient.
  • the carrier, excipient, and diluent may take a wide variety of forms depending upon the desired route of administration (e.g., oral, parenteral, etc.).
  • suitable carriers, excipients and diluents include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents and the like;
  • suitable carriers, excipients and diluents include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.
  • Solid oral preparations also may be optionally coated with substances such as, sugars, or be enterically coated so as to modulate the major site of absorption and disintegration.
  • the carrier, excipient and diluent will usually include sterile water, and other ingredients may be added to increase solubility and preservation of the composition.
  • injectable suspensions or solutions may also be prepared utilizing aqueous carriers along with appropriate additives such as, solubilizers and
  • a therapeutically effective amount of a compound of Formula (la) or (lb) or a pharmaceutical composition thereof includes a dose range from about 0.1 mg to about 3000 mg, or any particular amount or range therein, in particular from about 1 mg to about 1000 mg, or any particular amount or range therein, or, more particularly, from about 10 mg to about 500 mg , or any particular amount or range therein, of active ingredient in a regimen of about 1 to about 4 times per day for an average (70 kg) human; although, it is apparent to one skilled in the art that the therapeutically effective amount for a compound of Formula (la) or (lb) will vary as will the diseases, syndromes, conditions, and disorders being treated.
  • a pharmaceutical composition is preferably provided in the form of tablets containing about 1.0, about 10, about 50, about 100, about 150, about 200, about 250, and about 500 milligrams of a compound of Formula (la) or (lb).
  • a compound of Formula (la) or (lb) may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three and four times daily.
  • Optimal dosages of a compound of Formula (la) or (lb) to be administered may be readily determined and will vary with the particular compound used, the mode of administration, the strength of the preparation and the advancement of the disease, syndrome, condition or disorder.
  • factors associated with the particular subject being treated including subject gender, age, weight, diet and time of administration, will result in the need to adjust the dose to achieve an appropriate therapeutic level and desired therapeutic effect.
  • the above dosages are thus exemplary of the average case. There can be, of course, individual instances wherein higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • Compounds of Formula (la) or (lb) may be administered in any of the foregoing compositions and dosage regimens or by means of those compositions and dosage regimens established in the art whenever use of a compound of Formula (la) or (lb) is required for a subject in need thereof.
  • the compounds of Formula (la) and (lb) are useful in methods for treating and preventing a disease, a syndrome, a condition or a disorder in a subject, including an animal, a mammal and a human in which the disease, the syndrome, the condition or the disorder is affected by the modulation, including inhibition, of the MGL enzyme.
  • Such methods comprise, consist of and/or consist essentially of administering to a subject, including an animal, a mammal, and a human in need of such treatment or prevention a therapeutically effective amount of a compound, salt or solvate of Formula (la) or (lb).
  • Scheme A illustrates a route for the synthesis of compounds of Formula (la), wherein Y and Z are as defined herein.
  • a compound of formula Al is either commercially available or may be prepared by known methods described in the scientific literature.
  • a compound of formula Al may undergo a reductive amination with a compound of formula A2 (wherein P is a conventional amino protecting group such as, Boc, Fmoc, Cbz, and the like) in the presence of a hydride source such as, sodium triacetoxyborohydride to afford a compound of formula A3.
  • P is a conventional amino protecting group such as, Boc, Fmoc, Cbz, and the like
  • a hydride source such as, sodium triacetoxyborohydride
  • the Y group of the present invention may be introduced by reaction with a compound of formula A4, Y-X, (wherein X is a suitable leaving group such as, bromo or iodo, and, under certain reaction conditions, may be chloro or triflate) in the presence of copper iodide, 1,10— phenanthroline, and potassium phosphate, to afford a compound of formula A5. Removal of the protecting group (P) by conventional methods affords a compound of formula A6.
  • a compound of formula A6 may be treated with a carboxylic acid of formula A7 wherein Q is hydroxy, in the presence of an appropriate coupling agent such as, HATU, DCC, EDC, HBTU, PyBrOP, and the like; and optionally in the presence of a base such as, DIPEA, to afford a compound of Formula (la).
  • an acid chloride of formula A7 wherein Q is chloro may be used to effect the acylation of a compound of formula A6.
  • a non-nucleophilic base such as, pyridine, may be added to afford a compound of Formula (la).
  • Scheme B illustrates a route for the synthesis of compounds of Formula (lb), wherein Yb and Zb are as defined herein.
  • a compound of formula A2 is commercially available and may be deprotected using conventional chemistry to afford the corresponding amine of formula Bl.
  • Reductive amination with a diamine of formula B4 (wherein P is a conventional amino protecting group) in the presence of a hydride source such as, sodium triacetoxyborohydride affords a compound of formula B5.
  • a hydride source such as, sodium triacetoxyborohydride
  • a compound of formula B7 may be treated with a compound of formula B8 (wherein X b is fluoro, bromo, chloro, iodo, or triflate) under suitable reaction conditions to afford a compound of Formula (lb).
  • X b is fluoro, bromo, chloro, iodo, or triflate
  • Y b is 2-pyrimidinyl, 4-pyrimidinyl, 2-pyridinyl, 2-thiazolyl, or 2- oxazolyl
  • a base such as, potassium carbonate or sodium bicarbonate
  • Y b groups of the present invention may be incorporated by the addition of Y b - b (B8) in the presence of a palladium catalyst, optionally with a phosphine ligand, and in the presence of an inorganic base such as, potassium carbonate.
  • Scheme C illustrates an alternate route for the synthesis of compounds of Formula (lb), wherein Y b and Z b are as defined herein.
  • a compound of formula B8 (either commercially available or prepared by known methods found in the scientific literature) may be treated with a compound of formula B4 to afford a compound of formula CI. More specifically, when Y b of formula B8 is 2-pyrimidinyl, 4-pyrimidinyl, 2-pyridinyl, 2-thiazolyl, or 2-oxazolyl, the addition of a compound of formula B8 in the presence of a base such as, potassium carbonate or sodium bicarbonate, will provide the desired compounds of formula CI.
  • a base such as, potassium carbonate or sodium bicarbonate
  • Y b groups of the present invention may be incorporated by the addition of B8 in the presence of a palladium catalyst, optionally with a phosphine ligand, and in the presence of an inorganic base such as, potassium carbonate.
  • a palladium catalyst optionally with a phosphine ligand
  • an inorganic base such as, potassium carbonate.
  • Conventional removal of the amino protecting group (P) followed by reductive alkylation with a compound of formula A2 affords a compound of formula C2.
  • Acylation with a compound of formula B6 followed by treatment with a strong base such as, sodium hydride affords the cyclized product of formula C3.
  • Amino deprotection gives the corresponding free amine of formula C4, which, upon acylation with a compound of formula B2 as previously described, affords compounds of Formula (lb).
  • Scheme D illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Z b is a heteroaryl substituted with an optionally substituted phenyl group (Ar D ).
  • the heteroaryl ring is represented by an indole.
  • a compound of formula Dl is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound Dl may be treated with an phenyl iodide of formula D2 in the presence of copper iodide, trans-N, '- dimethylcyclohexane-l,2-diamine, and potassium phosphate to afford a compound of formula D3.
  • Subsequent saponification affords useful carboxylic acid intermediates of formula D4.
  • Scheme E illustrates the preparation of certain useful intermediates of formula A7 and B2 (Q is hydroxy) wherein Z/Z b is benzoxazolyl substituted with an optionally substituted phenyl group (Ar E ).
  • a compound of formula El is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound of formula El may be treated with an optionally substituted benzoyl chloride of formula E2 in an organic solvent such as, dioxane, at an elevated temperature to afford the substituted benzoxazole of formula E3. Subsequent saponification affords useful carboxylic acid intermediates of formula E4.
  • Scheme F illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is an optionally substituted benzothienyl group, and RF represents appropriate substituents as defined in Formula (la) and Formula (lb).
  • a compound of formula Fl is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound of formula Fl may be treated with thionyl chloride in an aprotic organic solvent, followed by treatment with methanol to afford a compound of formula F2. Subsequent
  • Scheme G illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is phenyl substituted by an optionally substituted phenylmethyl (Ar T -methyl) group.
  • a compound of formula Gl is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound of formula Gl may be treated with with an appropriately substituted boronic acid or ester of formula G2, in the presence of a palladium catalyst and a suitable base such as, potassium carbonate to afford a compound of formula G3.
  • a palladium catalyst and a suitable base such as, potassium carbonate
  • Subsequent saponification affords useful carboxylic acid intermediates of formula G4.
  • Scheme H illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is a benzothienyl group substituted with a fluoro substituent and an optionally substituted phenyl group (Ar H ).
  • a compound of formula HI is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound of formula HI may be cross-coupled with a boronic acid or ester (H2) in the presence of a palladium catalyst; and in the presence of a suitable base such as, potassium carbonate, to afford a compound of formula H3.
  • a suitable base such as, potassium carbonate
  • Saponification affords the corresponding carboxylic acid H4, which may be treated with N-fluorobenzenesulfonimide in the presence of an organometallic base such as, n-butyllithium, to afford the fluorinated compound of formula H5.
  • Scheme I illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is a benzothiazole group substituted with an optionally substituted phenyl group (Ari).
  • a compound of formula II is either commercially available or may be prepared by known methods described in the scientific literature.
  • the compound of formula II may be cross-coupled with a boronic acid or ester (12) in the presence of a palladium catalyst; and in the presence of a suitable base such as, potassium carbonate, to afford a compound of formula 13. Saponification affords the corresponding carboxylic acid of formula 14.
  • Step A l-(6-Trifluoromethyl-benzo[b]thiophene-2-carbonyl)-azetidin-3-one le.
  • a solution of N-Boc-azetidin-3-one la (171 mg, 1.0 mmol) and TFA (1 mL) in CH 2 CI 2 (4 mL) was stirred at room temperature for 2h.
  • the solution was concentrated to give intermediate lb, which was used in next step without further purification.
  • Step B ⁇ 2-[l-(6-Trifluoromethyl-benzo[b]thiophene-2-carbonyl)-azetidin-3- ylamino]-ethyl ⁇ -carbamic acid tert-butyl ester lg.
  • Step A [2-(Pyrimidin-2-ylamino)-ethyl]-carbamic acid tert-butyl ester 2a.
  • Step B 3-[2-(Pyrimidin-2-ylamino)-ethylamino]-azetidine-l-carboxylic acid tert-butyl ester 2b.
  • Step C 3-(2-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 2 c.
  • Step A 3-Oxo-4-pyrimidin-2-yl-piperazine-l-carboxylic acid tert-butyl ester
  • Step B 3-(3-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 3 c.
  • A. Methyl l-(4-fluorophenyl)-indole-5-carboxylate 3g.
  • a mixture of methyl indole-5-carboxylate 3e (0.5 g, 2.85 mmol), 1 -bromo-4-fluoro-benzene 3f (2 mL, 18.21 mmol), Cul (0.544 g, 2.85 mmol), and K 2 C0 3 (0.591 g, 4.28 mmol) was heated under microwave at 220 °C for 2.5 hours.
  • Step A 3-(3-Oxo-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester
  • Step B 3-(3-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 3 c.
  • Cpd 4 was prepared according to the procedures described in Example 2, Step D, using intermediates 3c and l-(4-fluoro-phenyl)-lH-indole-5-carboxylic acid 3h as starting materials. MS 471 (M+H + ).
  • Cpd 14 was prepared following the procedures described in Example 5 with intermediates 6a and 5a.
  • Intermediate 6a was prepared following the procedures described in Example 3, Step B, using commercially available 1 -phenyl-piperazin-2- one as starting material.
  • MS 494 (M+H + ).
  • Examples 7-26 provide synthetic routes to useful intermediates for the preparation of compounds of Formula (I).
  • A. Methyl 2-(4-fluoro-benzoylamino)-3-hydroxy-benzoate, 8c A solution of 1.0 g (4.9 mmol) of methyl 2-amino-3-hydroxybenzoate 8a, 1.03 g (7.4 mmol) of 4- fluorobenzoic acid 8b, 10 mL DMF and 2.9 mL (20.6 mmol) of TEA were placed into a flask and stirred for 10 min. HATU (7.4 mmol, 2.8 g) was added and the reaction was stirred overnight. The reaction mixture was poured into water and extracted with EtO Ac.
  • Methyl 2-(4-fluorophenyl)benzo[d]oxazole-4-carboxylate, 8d Methyl 2- (4-fluoro-benzoylamino)-3-hydroxy -benzoate 8c (7.4 mmol, 1.2 g crude) and 1.3 g (7.5 mmol) of p-toluenesulfonic acid were refluxed in 10 mL of xylene overnight. After cooling saturated aHC0 3 was added and the resulting mixture was extracted with EtO Ac. The organic solvent was evaporated to give 1.1 g (55%) of methyl 2-(4- fluorophenyl)benzo[d]oxazole-4-carboxylate, 8d.
  • Methyl 4-(4-(trifluoromethyl)benzyl)benzoate 9b.
  • A. 3-Methyl-[l,l'-biphenyl]-4-carboxylic acid, 14b To a suspension of compound 14a (0.025 g, 0.115 mmol), compound 10b (0.0139 g, 0.14 mmol), and Cs 2 C0 3 (0.094 g, 0.288 mmol) in dioxane (3 mL) and EtOH (1 mL) was added Pd(dppf)Ci2 (0.0084 g, 0.0115 mmol). The reaction mixture was stirred at 80 °C for 3 h. After cooling, the solid was removed by filtration and washed with CH 3 OH. The filtrate was concentrated. The crude product 14b was purified by reverse phase chromatography. MS m/z (M+H + ) 213.1.
  • a mixture of 1-tert-butyl 6-methyl 3-iodo-lH-indole-l,6-dicarboxylate 11a (5.02 mmol, 2.016 g), phenylboronic acid 10b (7.53 mmol, 0.92 g), Pd(OAc) 2 (0.402 mmol, 90 mg), Sphos 0.904 mmol, (0.37 g), and K 3 P0 4 (10.1 mmol, 2.13g) in toluene (10 mL) in sealed reaction vial was stirred at room temperature for 2 min and then heated at 90 °C under 2 for 4 h.
  • reaction mixture was quenched with EtOAc and water.
  • organic layer was concentrated and purified by flash column chromatography (silica gel, 8% EtOAc/hexanes).
  • the desired product was collected as a light yellow solid that was washed with small amount of hexanes to obtain 16a as a white solid.
  • LDA 2.0 M in THF/heptane/ethylbenzene, 25.3 mmol, 12.6 mL
  • 1 -fluoro-4-chloro-bezene 18a 23.0 mmol, 2.45 mL
  • the mixture was stirred for 1 h at -78 °C and ethyl trifluoroacetate 18b (25.3 mmol, 3.02 mL) was added.
  • reaction mixture was allowed to warm to room temperature overnight and was quenched with saturated aqueous NH 4 CI solution.
  • the mixture was extracted with EtOAc.
  • the organic extracts were concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give a mixture of the compound 18c along with a regio-isomeric by-product, l-(5-fluoro-2-chloro-phenyl)- 2,2,2-trifluoro-ethanone, in a ratio of 5: 1 (18c is the major product).
  • the reaction mixture was concentrated and purified by flash column chromatography (silica gel, 15-20% EtOAc/hexanes) to give compound 20b as an off-white solid.
  • n-BuLi 1.6 M solution in hexanes, 5.2 mmol, 3.2 mL.
  • the solution was kept at -60 °C for 4 h and was then poured onto an excess of freshly crushed dry ice. Water was added and the mixture was acidified to pH 4. The organic phase was concentrated and the residue was purified by flash column chromatography (silica gel, 35% EtOAc/hexanes) to give compound 21b.
  • A. Methyl 7-methyl-lH-indole-5-carboxylate, 22c A mixture of compound 22a (0.613 mmol, 156 mg), methylboronic acid 22b (0.92 mmol, 79 mg), Pd(OAc)2 (0.09 mmol, 20 mg), SPhos (0.215 mmol, 88mg), and K 3 P0 4 (1.23 mmol, 0.26 g) in toluene (2 mL) was heated to 100 °C for 3 h in a sealed reaction vessel. The reaction was diluted with EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 10% EtOAc/hexanes) to give compound 22c.
  • Ethyl 2-phenyl-benzothiazole-6-carboxylate, 26b A mixture of ethyl 2- bromo-benzothiazole-6-carboxylate 26a (300 mg, 1.05 mmol), phenylboronic acid 10b (192 mg, 1.57 mmol), K 2 C0 3 (188 mg, 1.36 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (43 mg, 0.05 mmol) in dioxane (2 mL) and H 2 0 (0.4 ml) was heated at 120 °C for 25 min under microwave. The reaction mixture was diluted with CH 2 C1 2 , washed with H 2 0, dried over Na 2 S0 4 , and concentrated. Purification by flash column chromatography (silica gel, 15% EtO Ac/heptane) gave 26b (220 mg).
  • IC5 0 values for compounds of Formula (la) or (lb) were determined using Excel from a fit of the equation to the concentration-response plot of the fractional activity as a function of inhibitor concentration.
  • ThermoFluor (TF) assay is a 384-well plate-based binding assay that measures thermal stability of proteins 1 ' 2 .
  • the experiments were carried out using instruments available from Johnson & Johnson Pharmaceutical Research &
  • TF dye used in all experiments was 1,8-ANS (Invitrogen: A-47).
  • Screening compound plates contained 100% DMSO compound solutions at a single concentration.
  • compounds were arranged in a pre-dispensed plate (Greiner Bio-one: 781280), wherein compounds were serially diluted in 100% DMSO across 11 columns within a series. Columns 12 and 24 were used as DMSO reference and contained no compound.
  • the compound aliquots 46 nL were robotically predispensed directly into 384-well black assay plates (Abgene: TF-0384/k) using the Hummingbird liquid handler.
  • protein and dye solutions were added to achieve the final assay volume of 3 ⁇
  • the assay solutions were overlayed with 1 ⁇ , of silicone oil (Fluka, type DC 200: 8541 1) to prevent evaporation.
  • Bar-coded assay plates were robotically loaded onto a thermostatically controlled PCR-type thermal block and then heated from 40 to 90 °C degrees at a ramp- rate of 1 °C/min for all experiments. Fluorescence was measured by continuous illumination with UV light (Hamamatsu LC6), supplied via fiber optics and filtered through a band-pass filter (380-400 nm; > 6 OD cutoff). Fluorescence emission of the entire 384-well plate was detected by measuring light intensity using a CCD camera (Sensys, Roper Scientific) filtered to detect 500 ⁇ 25 nm, resulting in simultaneous and independent readings of all 384 wells. A single image with 20-sec exposure time was collected at each temperature, and the sum of the pixel intensity in a given area of the assay plate was recorded vs temperature and fit to standard equations to yield the T m l .
  • the Ka values for compounds of Formula (la) and (lb) were determined from a fit of the equation to the concentration-response plot of the fractional activity as a function of T m .
  • quantitative NMR spectroscopy (qNMR) was used to measure concentration of the initial 100% DMSO compound solutions and, using the same fitting method, qKa values were determined.
  • Biological DataTable 3
  • Each rat may be placed in a test chamber on a warm glass surface and allowed to acclimate for approximately 10 min.
  • a radiant thermal stimulus (beam of light) may be focused through the glass onto the plantar surface of each hind paw in turn.
  • the thermal stimulus may be automatically shut off by a photoelectric relay when the paw is moved or when the cut-off time is reached (20 sec for radiant heat at ⁇ 5 amps).
  • An initial (baseline) response latency to the thermal stimulus may be recorded for each animal prior to the injection of complete Freund's adjuvant (CFA). Twenty-four hours following intraplantar CFA injection, the response latency of the animal to the thermal stimulus may be re-evaluated and compared to the animal's baseline response time. Only rats that exhibit at least a 25% reduction in response latency (i.e., are
  • test compound or vehicle may be administered orally.
  • Post-compound treatment withdrawal latencies may be assessed at fixed time intervals, typically 30, 60, 120, 180, and 300 min.
  • the percent reversal (%R) of hypersensitivity may be calculated in one of two different ways: 1) using group mean values or 2) using individual animal values. More specifically:
  • %R of hypersensitivity may be calculated using the mean value for groups of animals at each time point according to the following formula:
  • Results may be given for the maximum % reversal observed for each compound at any time point tested.
  • Results are given as a mean of the maximum % reversal values calculated for each individual animal.
  • rats Prior to testing, rats may be acclimated to the handling procedure twice a day for a period of two days .
  • the test consists of placing the left hindpaw on a
  • the response threshold of the animal to the mechanical stimulus may be re-evaluated and compared to the animal's baseline response threshold.
  • a response may be defined as a withdrawal of the hindpaw, a struggling to remove the hindpaw or vocalization. Only rats that exhibit at least a 25% reduction in response threshold (i.e., hyperalgesia) may be included in further analysis.
  • rats may be administered the indicated test compound or vehicle.
  • Post- treatment withdrawal thresholds may be assessed at 1 h.
  • Paw withdrawal thresholds may be converted to percent reversal of hypersensitivity according to the following formula:
  • CCI Chronic constriction injury
  • Male Sprague-Dawley rats (225-450 g) may be used to evaluate the ability of selected compounds to reverse CCI-induced cold hypersensitivity.
  • Four loose ligatures of 4-0 chromic gut may be surgically placed around the left sciatic nerve under inhalation anesthesia as described by Bennett et al. (Bennett GJ, Xie YK. Pain 1988, 33(1): 87-107).
  • Fourteen to 35 days following CCI surgery subjects may be placed in elevated observation chambers containing wire mesh floors, and five applications of acetone (0.05 mL/application separated by approximately 5 minutes) may be spritzed onto the plantar surface of the paw using a multidose syringe. An abrupt withdrawal or lifting of the paw may be considered a positive response.
  • the number of positive responses may be recorded for each rat over the five trials. Following baseline withdrawal determinations, compounds may be administered in the indicated vehicle, by the indicated route (see Table 6). The number of withdrawals may be re-determined 1 to 4 hr after compound administration. Results may be presented as a percent inhibition of shakes, which may be calculated for each subject as [l-(test compound withdrawals / pre-test withdrawals)] x 100 and then averaged by treatment.
  • SNL Spinal nerve ligation
  • L 5 spinal nerve ligation
  • anesthesia may be induced and maintained on isoflurane inhalation.
  • Fur may be clipped over the dorsal pelvic area, and a 2-cm skin incision may be made just left of midline over the dorsal aspect of the L4-S2 spinal segments, followed by separation of the paraspinal muscles from spinous processes.
  • the transverse process of L 6 may be carefully removed, and the L5 spinal nerve may be identified.
  • the left L5 spinal nerve may be ligated tightly with 6-0 silk thread, the muscle may be sutured with 4-0 vicryl, and the skin may be closed with wound clips.
  • s.c. saline (5 mL) may be administered.
  • Behavioral testing may be performed four weeks post-ligation. Following baseline von Frey determinations to verify the presence of mechanical allodynia, L5 SNL rats may be orally administered the indicated vehicle or drug. Tactile allodynia may be quantified at 30 , 60, 100, 180 and 300 min post-dosing by recording the force at which the paw ipsilateral to the nerve ligation is withdrawn from the application of a series of calibrated von Frey filaments (0.4, 0.6, 1.0, 2.0, 4, 6, 8 and 15 g; Stoelting; Wood Dale, IL).
  • filaments may be applied to the mid-plantar hind paw for approximately 5 seconds to determine the response threshold, a brisk paw withdrawal leads to the presentation of the next lighter stimulus, whereas a lack of a withdrawal response leads to the presentation of the next stronger stimulus.
  • a total of four responses after the first threshold detection may be collected.
  • the 50% withdrawal thresholds may be interpolated by the method of Dixon as modified by Chaplan et.al, and when response thresholds fall above or below the range of detection, respective values of 15.0 or 0.25 g may be assigned.
  • Threshold data from von Frey filament testing may be reported as withdrawal threshold in grams. Data may be normalized and results may be presented as % MPE (maximum possible effect) of the drug calculated according to the following formula:
  • % MPE x g/force - baseline g/force X 100

Abstract

Disclosed are compounds, compositions and methods for treating various diseases, syndromes, conditions and disorders, including pain. Such compounds, and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof, are represented by Formula (la) and Formula (lb) as follows: wherein Y, Z, and n are defined herein; and wherein Yb and Zb are as defined herein.

Description

OXOPIPERAZINE-AZETIDINE AMIDES AND OXODIAZEPINE-AZETIDINE AMIDES AS MONOACYLGLYCEROL LIPASE INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS Not applicable.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT The research and development of the invention described below was not federally sponsored.
BACKGROUND OF THE INVENTION
Cannabis sativa has been used for the treatment of pain for many years. A9- tetrahydrocannabinol is a major active ingredient from Cannabis sativa and an agonist of cannabinoid receptors (Pertwee, Brit J Pharmacol, 2008, 153, 199-215). Two cannabinoid G protein-coupled receptors have been cloned, cannabinoid receptor type 1 (CBi Matsuda et al, Nature, 1990, 346, 561-4) and cannabinoid receptor type 2 (CB2 Munro et al, Nature, 1993, 365, 61-5). CBi is expressed centrally in brain areas such as, the hypothalamus and nucleus accumbens, as well as peripherally in the liver, gastrointestinal tract, pancreas, adipose tissue, and skeletal muscle (Di Marzo et al, Curr Opin Lipidoi, 2007, 18, 129-140). CB2 is predominantly expressed in immune cells such as, monocytes (Pacher et al, Amer J Physiol, 2008, 294, HI 133-Hl 134), and under certain conditions, also in the brain (Benito et al, Brit J Pharmacol, 2008, 153, 277-285) and in skeletal (Cavuoto et al, Biochem Biophys Res Commun, 2007, 364, 105-1 10) and cardiac (Hajrasouliha et al, Eur J Pharmacol, 2008, 579, 246-252) muscle. An abundance of pharmacological, anatomical and electrophysiological data, using synthetic agonists, indicate that increased cannabinoid signaling through
CB!/CB2 promotes analgesia in tests of acute nociception and suppresses hyperalgesia in models of chronic neuropathic and inflammatory pain (Cravatt et al, J Neurobiol, 2004, 61, 149-60; Guindon et al , Brit J Pharmacol, 2008, 153 , 319-334).
Efficacy of synthetic cannabinoid receptor agonists is well documented.
Moreover, studies using cannabinoid receptor antagonists and knockout mice have also implicated the endocannabinoid system as an important modulator of nociception. Anandamide (AEA) (Devane et al, Science, 1992, 258, 1946-9) and 2- arachidinoylglycerol (2-AG) (Mechoulam et al, Biochem Pharmacol, 1995, 50, 83-90; Sugiura et al, Biochem Biophys Res Commun, 1995, 215, 89-97) are two major endocannabinoids. AEA is hydrolyzed by fatty acid amide hydrolase (FAAH) and 2- AG is hydrolyzed by monoacylglycerol lipase (MGL) (Piomelli, Nat Rev Neurosci, 2003, 4, 873-884). Genetic ablation of FAAH elevates endogenous AEA and results in a CBi-dependent analgesia in models of acute and inflammatory pain (Lichtman et al, Pain, 2004, 109, 319-27), suggesting that the endocannabinoid system functions naturally to inhibit pain (Cravatt et al, J Neurobiol, 2004, 61, 149-60). Unlike the constitutive increase in endocannabinoid levels using FAAH knockout mice, use of specific FAAH inhibitors transiently elevates AEA levels and results in antinociception in vivo (Kathuria et al, Nat Med, 2003, 9, 76-81). Further evidence for an
endocannabinoid-mediated antinociceptive tone is demonstrated by the formation of AEA in the periaqueductal grey following noxious stimulation in the periphery (Walker et al, Proc Natl Acad Sci USA, 1999, 96, 12198-203) and, conversely, by the induction of hyperalgesia following antisense RNA-mediated inhibition of CBi in the spinal cord (Dogrul et al., Pain, 2002, 100, 203-9).
With respect to 2-AG, intravenous delivery of 2-AG produces analgesia in the tail flick (Mechoulam et al, Biochem Pharmacol, 1995, 50, 83-90) and hot plate
(Lichtman et al, J Pharmacol Exp Ther, 2002, 302, 73-9) assays. In contrast, it was demonstrated that 2-AG given alone is not analgesic in the hot plate assay, but when combined with other 2-monoacylglycerols (i.e., 2-linoleoyl glycerol and 2-palmitoyl glycerol), significant analgesia is attained, a phenomenon termed the "entourage effect" (Ben-Shabat et al, Eur J Pharmacol, 1998, 353, 23-31). These "entourage" 2- monoacylglycerols are endogenous lipids that are co-released with 2-AG and potentiate endocannabinoid signaling, in part, by inhibiting 2-AG breakdown, most likely by competition for the active site on MGL. This suggests that synthetic MGL Inhibitors will have a similar effect. Indeed, URB602, a relatively weak synthetic MGL Inhibitor, showed an antinociceptive effect in a murine model of acute inflammation (Comelli et al, Brit J Pharmacol, 2007, 152, 787-794).
Although the use of synthetic cannabinoid agonists have conclusively demonstrated that increased cannabinoid signaling produces analgesic and anti- inflammatory effects, it has been difficult to separate these beneficial effects from the unwanted side effects of these compounds. An alternative approach is to enhance the signaling of the endocannabinoid system by elevating the level of 2-AG, the endocannabinoid of highest abundance in the central nervous system (CNS) and gastrointestinal tract, which may be achieved by inhibition of MGL. Therefore, MGL inhibitors are potentially useful for the treatment of pain, inflammation, and CNS disorders (Di Marzo et al, Curr Pharm Des, 2000, 6, 1361-80; Jhaveri et al, Brit J Pharmacol, 2007, 152, 624-632; McCarberg Bill et al, Amer J Ther, 2007, 14, 475- 83), as well as glaucoma and disease states arising from elevated intraocular pressure
(Njie, Ya Fatou; He, Fang; Qiao, Zhuanhong; Song, Zhao-Hui, Exp. Eye Res., 2008, 87(2): 106-14).
SUMMARY OF THE INVENTION
The present invention is directed to a compound of Formula (la)
Figure imgf000005_0001
Formula (la)
wherein
Y is
i) a C6-io aryl or
ϋ) a heteroaryl selected from the group consisting of thiazolyl, oxazolyl, pyridinyl, and pyrimidinyl,
wherein Y is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of fluoro, chloro, Ci_4alkyl,
Figure imgf000005_0002
cyano, and trifluoromethyl;
Z is
i) a C6-io aryl,
ϋ) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, indazolyl, and indolyl, or iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is
unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, C1-4alkyl, Ci_4alkoxy, and trifluoromethyl, wherein the Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, Ci_ 4alkoxy, trifluoromethyl, and phenyl;
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro; n is 1 or 2; and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
The present invention is further directed to a compound of Formula (lb)
Figure imgf000006_0001
Formula (lb)
wherein:
Yb is
Figure imgf000006_0002
ii) a heteroaryl selected from the group consisting of thiazolyl, oxazolyl, pyridinyl, and pyrimidinyl,
wherein Yb is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci_4alkoxy, cyano, and trifluoromethyl;
a C6-io aryl,
a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl, or iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, C1-4alkyl, Ci_4alkoxy, and trifluoromethyl, wherein the Ce-w aryl and the heteroaryl of Zb are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, Ci
4alkoxy, trifluoromethyl, and phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro; and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
The present invention also provides, inter alia, a pharmaceutical composition comprising, consisting of and/or consisting essentially of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and/or a pharmaceutically acceptable diluent, and a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof.
Also provided are processes for making a pharmaceutical composition comprising, consisting of, and/or consisting essentially of admixing a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof, and a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and/or a pharmaceutically acceptable diluent.
The present invention further provides, inter alia, methods for treating or ameliorating a MGL-modulated disorder in a subject, including a human or other mammal in which the disease, syndrome, or condition is affected by the modulation of the MGL enzyme such as pain, and the diseases that lead to such pain, inflammation, and CNS disorders, using a compound of Formula (la) or (lb) or a pharmaceutically acceptable salt form thereof.
The present invention also provides, inter alia, methods for producing the instant compounds and pharmaceutical compositions and medicaments thereof. DETAILED DESCRIPTION OF THE INVENTION
With reference to substituents, the term "independently" refers to the situation where when more than one substituent is possible, the substituents may be the same or different from each other.
The term "alkyl" whether used alone or as part of a substituent group, refers to straight and branched carbon chains having 1 to 8 carbon atoms. Therefore, designated numbers of carbon atoms (e.g., C1-8) refer independently to the number of carbon atoms in an alkyl moiety or to the alkyl portion of a larger alkyl-containing substituent. In substituent groups with multiple alkyl groups such as, (Ci_6alkyl)2amino-, the
Figure imgf000008_0001
groups of the dialkylamino may be the same or different.
The term "alkoxy" refers to an -O-alkyl group, wherein the term "alkyl" is as defined above.
The terms "alkenyl" and "alkynyl" refer to straight and branched carbon chains having 2 or more carbon atoms, wherein an alkenyl chain contains at least one double bond and an alkynyl chain contains at least one triple bond.
The term "cycloalkyl" refers to saturated or partially saturated, monocyclic or polycyclic hydrocarbon rings of 3 to 14 carbon atoms. Examples of such rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and adamantyl.
The term "benzo-fused cycloalkyl" refers to a 5- to 8- membered monocyclic cycloalkyl ring fused to a benzene ring. The carbon atom ring members that form the cycloalkyl ring may be fully saturated or partially saturated.
The term "heterocyclyl" refers to a nonaromatic monocyclic or bicyclic ring system having 3 to 10 ring members that include at least 1 carbon atom and from 1 to 4 heteroatoms independently selected from N, O, and S. Included within the term heterocyclyl is a nonaromatic cyclic ring of 5 to 7 members in which 1 to 2 members are N, or a nonaromatic cyclic ring of 5 to 7 members in which 0, 1 or 2 members are N and up to 2 members are O or S and at least one member must be either N, O, or S; wherein, optionally, the ring contains 0 to 1 unsaturated bonds, and, optionally, when the ring is of 6 or 7 members, it contains up to 2 unsaturated bonds. The carbon atom ring members that form a heterocycle ring may be fully saturated or partially saturated.
The term "heterocyclyl" also includes two 5 membered monocyclic heterocycloalkyl groups bridged to form a bicyclic ring. Such groups are not considered to be fully aromatic and are not referred to as heteroaryl groups. When a heterocycle is bicyclic, both rings of the heterocycle are non-aromatic and at least one of the rings contains a heteroatom ring member. Examples of heterocycle groups include, and are not limited to, pyrrolinyl (including 2H-pyrrole, 2-pyrrolinyl or 3-pyrrolinyl), pyrrolidinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, and piperazinyl. Unless otherwise noted, the heterocycle is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
The term "benzo-fused heterocyclyl" refers to a 5 to 7 membered monocyclic heterocycle ring fused to a benzene ring. The heterocycle ring contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S. The carbon atom ring members that form the heterocycle ring may be fully saturated or partially saturated. Unless otherwise noted, benzo-fused heterocycle ring is attached to its pendant group at a carbon atom of the benzene ring.
The term "aryl" refers to an unsaturated, aromatic monocyclic or bicyclic ring of 6 to 10 carbon members. Examples of aryl rings include phenyl and naphthalenyl.
The term "heteroaryl" refers to an aromatic monocyclic or bicyclic aromatic ring system having 5 to 10 ring members and which contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S. Included within the term heteroaryl are aromatic rings of 5 or 6 members wherein the ring consists of carbon atoms and has at least one heteroatom member. Suitable heteroatoms include N, O, and S. In the case of 5 membered rings, the heteroaryl ring preferably contains one member of N, O or S and, in addition, up to 3 additional N atoms. In the case of 6 membered rings, the heteroaryl ring preferably contains from 1 to 3 nitrogen atoms. For the case wherein the 6 membered ring has 3 N, at most 2 nitrogen atoms are adjacent. When a heteroaryl is bicyclic, at least one heteroatom is present in each ring. Examples of heteroaryl groups include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl and pyrazinyl. Unless otherwise noted, the heteroaryl is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
Unless otherwise noted, the term "benzo fused heteroaryl" refers to a 5 to 6 membered monocyclic heteroaryl ring fused to a benzene ring. The heteroaryl ring contains carbon atoms and from 1 to 4 heteroatoms independently selected from N, O, and S. Examples of heteroaryl groups with the optionally fused benzene rings include indolyl, isoindolyl, benzofuryl, benzothienyl, indazolyl, benzimidazolyl,
benzothiazolyl, benzoxazolyl, benzisoxazolyl, benzothiadiazolyl, benzotriazolyl, quinolinyl, isoquinolinyl and quinazolinyl. Unless otherwise noted, the benzo-fused heteroaryl is attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
The term "halogen" or "halo" refers to fluorine, chlorine, bromine and iodine atoms.
The term "formyl" refers to the group -C(=0)H.
The term "oxo" refers to the group (=0).
Whenever the term "alkyl" or "aryl" or either of their prefix roots appear in a name of a substituent (e.g., arylalkyl, alkylamino) the name is to be interpreted as including those limitations given above for "alkyl" and "aryl." Designated numbers of carbon atoms (e.g., Ci-Ce) refer independently to the number of carbon atoms in an alkyl moiety, an aryl moiety, or in the alkyl portion of a larger substituent in which alkyl appears as its prefix root. For alkyl and alkoxy substituents, the designated number of carbon atoms includes all of the independent members included within a given range specified. For example C e alkyl would include methyl, ethyl, propyl, butyl, pentyl and hexyl individually as well as sub-combinations thereof (e.g., C1-2, Ci_ 3, Ci_4, Ci_5; C2-6, C3_6, C4_6, C5.6, C2-5; etc.).
In general, under standard nomenclature rules used throughout this disclosure, the terminal portion of the designated side chain is described first followed by the adjacent functionality toward the point of attachment. Thus, for example, a "Ci-Ce alkylcarbonyl" substituent refers to a group of the formula:
Figure imgf000010_0001
The term "R" at a stereocenter designates that the stereocenter is purely of the R-configuration as defined in the art; likewise, the term "S" means that the stereocenter is purely of the ^-configuration. As used herein, the terms "*R" or "*S" at a stereocenter are used to designate that the stereocenter is of pure but unknown configuration. As used herein, the term "RS" refers to a stereocenter that exists as a mixture of the R- and ^-configurations. Similarly, the terms "*RS" or "*SR" refer to a stereocenter that exists as a mixture of the R- and ^-configurations and is of unknown configuration relative to another stereocenter within the molecule.
Compounds containing one stereocenter drawn without a stereo bond designation are a mixture of 2 enantiomers. Compounds containing 2 stereocenters both drawn without stereo bond designations are a mixture of 4 diastereomers.
Compounds with 2 stereocenters both labeled "RS" and drawn with stereo bond designations are a 2-component mixture with relative stereochemistry as drawn.
Compounds with 2 stereocenters both labeled "*RS" and drawn with stereo bond designations are a 2-component mixture with relative stereochemistry unknown.
Unlabeled stereocenters drawn without stereo bond designations are a mixture of the R- and ^-configurations. For unlabeled stereocenters drawn with stereo bond designations, the absolute stereochemistry is as depicted.
Unless otherwise noted, it is intended that the definition of any substituent or variable at a particular location in a molecule be independent of its definitions elsewhere in that molecule. It is understood that substituents and substitution patterns on the compounds of Formula (la) and (lb) can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art as well as those methods set forth herein.
The term "subject" refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
The term "therapeutically effective amount" refers to an amount of an active compound or pharmaceutical agent, including a compound of the present invention, which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation or partial alleviation of the symptoms of the disease, syndrome, condition, or disorder being treated.
The term "composition" refers to a product that includes the specified ingredients in therapeutically effective amounts, as well as any product that results, directly, or indirectly, from combinations of the specified ingredients in the specified amounts.
The term " MGL inhibitor" is intended to encompass a compound that interacts with MGL to substantially reduce or eliminate its catalytic activity, thereby increasing the concentrations of its substrate(s). The term "MGL-modulated" is used to refer to the condition of being affected by the modulation of the MGL enzyme including the condition of being affected by the inhibition of the MGL enzyme such as pain, and the diseases that lead to such pain, inflammation and CNS disorders.
As used herein, unless otherwise noted, the term "affect" or "affected" (when referring to a disease, syndrome, condition or disorder that is affected by inhibition of MGL) shall include a reduction in the frequency and / or severity of one or more symptoms or manifestations of said disease, syndrome, condition or disorder; and / or include the prevention of the development of one or more symptoms or manifestations of said disease, syndrome, condition or disorder or the development of the disease, condition, syndrome or disorder.
The compounds of Formula (la) and (lb) are useful in methods for treating, ameliorating and / or preventing a disease, a syndrome, a condition or a disorder that is affected by the inhibition of MGL. Such methods comprise, consist of and/or consist essentially of administering to a subject, including an animal, a mammal, and a human in need of such treatment, amelioration and / or prevention, a therapeutically effective amount of a compound of Formula (la) or (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof. In particular, the compounds of Formula (la) and (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof are useful for treating, ameliorating and / or preventing pain; diseases, syndromes, conditions, or disorders causing such pain; inflammation and / or CNS disorders. More particularly, the compounds of Formula (la) and (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof are useful for treating, ameliorating and / or preventing inflammatory pain, inflammatory hypersensitivity conditions and / or neuropathic pain, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (la) or (lb), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof as herein defined.
Examples of inflammatory pain include pain due to a disease, condition, syndrome, disorder, or a pain state including inflammatory bowel disease, visceral pain, migraine, post operative pain, osteoarthritis, rheumatoid arthritis, back pain, lower back pain, joint pain, abdominal pain, chest pain, labor, musculoskeletal diseases, skin diseases, toothache, pyresis, burn, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection, rhinitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome, cholecystitis, pancreatitis, postmastectomy pain syndrome, menstrual pain, endometriosis, pain due to physical trauma, headache, sinus headache, tension headache, or arachnoiditis.
One type of inflammatory pain is inflammatory hyperalgesia /
hypersensitivity. Examples of inflammatory hyperalgesia include a disease, syndrome, condition, disorder, or pain state including inflammation, osteoarthritis, rheumatoid arthritis, back pain, joint pain, abdominal pain, musculoskeletal diseases, skin diseases, post operative pain, headaches, toothache, burn, sunburn, insect sting, neurogenic bladder, urinary incontinence, interstitial cystitis, urinary tract infection, cough, asthma, chronic obstructive pulmonary disease, rhinitis, contact
dermatitis/hypersensitivity, itch, eczema, pharyngitis, enteritis, irritable bowel syndrome, inflammatory bowel diseases including Crohn's Disease, ulcerative colitis, urinary incontinence, benign prostatic hypertrophy, cough, asthma, rhinitis, nasal hypersensitivity, itch, contact dermatitis and / or dermal allegy and chronic obstructive pulmonary disease.
In an embodiment, the present invention is directed to a method for treating, ameliorating and / or preventing inflammatory visceral hyperalgesia in which a enhanced visceral irritability exists, comprising, consisting of, and/or consisting essentially of the step of administering to a subject in need of such treatment a therapeutically effective amount of a compound, salt or solvate of Formula (la) or (lb). In a further embodiment, the present invention is directed to a method for treating inflammatory somatic hyperalgesia in which a hypersensitivity to thermal, mechanical and/or chemical stimuli exists, comprising administering to a mammal in need of such treatment a therapeutically effective amount of a compound of Formula (la) or (lb) or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
A further embodiment of the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic pain. Examples of a
neuropathic pain include pain due to a disease, syndrome, condition, disorder, or pain state including cancer, neurological disorders, spine and peripheral nerve surgery, brain tumor, traumatic brain injury (TBI), spinal cord trauma, chronic pain syndrome, fibromyalgia, chronic fatigue syndrome, lupus, sarcoidosis, peripheral neuropathy, bilateral peripheral neuropathy, diabetic neuropathy, central pain, neuropathies associated with spinal cord injury, stroke, amyotrophic lateral sclerosis (ALS), Parkinson's disease, multiple sclerosis, sciatic neuritis, mandibular joint neuralgia, peripheral neuritis, polyneuritis, stump pain, phantom limb pain, bony fractures, oral neuropathic pain, Charcot's pain, complex regional pain syndrome I and II (CRPS I/II), radiculopathy, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngeal neuralgia, migrainous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, postherpetic neuralgia, causalgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia, trigeminal neuralgia, vulvodynia, or vidian neuralgia.
One type of neuropathic pain is neuropathic cold allodynia, which can be characterized by the presence of a neuropathy-associated allodynic state in which a hypersensitivity to cooling stimuli exists. Examples of neuropathic cold allodynia include allodynia due to a disease, condition, syndrome, disorder or pain state including neuropathic pain (neuralgia), pain arising from spine and peripheral nerve surgery or trauma, traumatic brain injury (TBI), trigeminal neuralgia, postherpetic neuralgia, causalgia, peripheral neuropathy, diabetic neuropathy, central pain, stroke, peripheral neuritis, polyneuritis, complex regional pain syndrome I and II (CRPS I/II) and radiculopathy.
In a further embodiment, the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic cold allodynia in which a hypersensitivity to a cooling stimuli exists, comprising, consisting of, and/or consisting essentially of the step of administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula (la) or (lb) or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
In a further embodiment, the present invention is directed to a method for treating, ameliorating and / or preventing CNS disorders. Examples of CNS disorders include anxieties such as, social anxiety, post-traumatic stress disorder, phobias, social phobia, special phobias, panic disorder, obsessive-compulsive disorder, acute stress disorder, separation anxiety disorder, and generalized anxiety disorder, as well as depression such as, major depression, bipolar disorder, seasonal affective disorder, post natal depression, manic depression, and bipolar depression.
Embodiments of the present invention include a compound of Formula (la)
Figure imgf000015_0001
Formula (la)
wherein:
Y is
i) phenyl or
ii) a heteroaryl that is thiazolyl or pyrimidinyl;
Y is thiazolyl or pyrimidinyl;
Z is
Figure imgf000015_0002
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein Ce-w aryl and heteroaryl of Z are optionally independently substituted with one to two substitutents selected from the group consisting of chloro, fluoro, Ci-4alkyl, trifluoromethyl, and phenyl,
provided that no more than one substituent of is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
n is 1 ; n is 2; and any combination of embodiments a) through f) above, provided that it is understood that combinations in which different embodiments of the same substituent would be combined are excluded;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
An embodiment of the present invention includes a compound of Formula (la)
Figure imgf000016_0001
Formula (la)
wherein:
Y is ii) a heteroaryl that is thiazolyl or pyrimidinyl;
Z is
i) C6-io aryl, or
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is selected from the group consisting of chloro, fluoro, Ci-4alkyl, trifluoromethyl, and phenyl;
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
or 2; and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
A further embodiment of the present invention includes a compound of Formula (la)
Figure imgf000017_0001
Formula (la)
wherein:
Y is
i) phenyl or
a heteroaryl that is thiazolyl or pyrimidinyl;
Z is
Figure imgf000017_0002
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, C1-4alkyl, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
n is 1 ; and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
An embodiment of the present invention includes a compound of Formula (la)
Figure imgf000017_0003
Formula (la)
wherein:
Y is thiazolyl or pyrimidinyl;
Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl, wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is
trifluoromethyl or fluoro;
n is 1 or 2;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
An embodiment of the present invention includes a compound of Formula (la)
Figure imgf000018_0001
Formula (la)
wherein:
Y is thiazolyl or pyrimidinyl;
Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is
trifluoromethyl or fluoro;
n is 1;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
A further embodiment of the present invention includes a compound of Formula (la)
Figure imgf000019_0001
Formula (la)
wherein:
Y is
i) phenyl or
ii) a heteroaryl that is thiazolyl or pyrimidinyl;
Z is
Figure imgf000019_0002
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the Ce-io aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, Ci-4alkyl, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
n is 2; and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
The present invention is ound of Formula (la)
Figure imgf000019_0003
Formula (la)
selected from the group consisting of
the compound wherein Y is phenyl, Z is 4-(phenylmethyl)-phenyl, and n is 1 ;
the compound wherein Y is phenyl, Z is 4-phenyl-phenyl, and n is 1 ;
the compound wherein Y is pyrimidin-2-yl, Z is 1 -(4-trifluoromethyl-phenyl)- 1H- indol-5-yl, and n is 1 ;
the compound wherein Y is pyrimidin-2-yl, Z is l -(4-fluoro-phenyl)-lH-indol-5-yl, and n is 1 ; the compound wherein Y is thiazol-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 1;
the compound wherein Y is thiazol-4-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 1;
the compound wherein Y is thiazol-4-yl, Z is 3-chloro-6-trifluoromethyl-benzothien-2- yl, and n is 1;
the compound wherein Y is pyrimidin-2-yl, Z is 3-chloro-6-trifluoromethyl-benzothien- 2-yl, and n is 1;
the compound wherein Y is pyrimidin-2-yl, Z is 4-phenyl-phenyl, and n is 1;
the compound wherein Y is pyrimidin-2-yl, Z is 2-phenyl-benzothiazol-6-yl, and n is 1; the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl-benzothien-2- yl, and n is 1;
the compound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzothiazol-6-yl, and n is 1; the compound wherein Y is phenyl, Z is l-(4-trifluoromethyl-phenyl)-lH-indol-5-yl, and n is 1;
the compound wherein Y is phenyl, Z is 3-chloro-6-trifluoromethyl-benzothien-2-yl, and n is 1 ;
the compound wherein Y is phenyl, Z is l-(3,4-difluoro-phenyl)-lH-indol-5-yl, and n is i;
the compound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6-yl, and n is 1 ; the compound wherein Y is pyrimidin-2-yl, Z is 3-chloro-6-trifluoromethyl-benzothien- 2-yl, and n is 2;
the compound wherein Y is thiazol-2-yl, Z is 4-phenyl-phenyl, and n is 1 ;
the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl-benzothien-2- yl, and n is 2;
the compound wherein Y is pyrimidin-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 2;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 2;
the compound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6-yl, and n is 2; and pharmaceutically acceptable salt forms thereof. Embodiments of the present invention are directed to a compound of Formula
Figure imgf000021_0001
Formula (lb)
wherein:
a) Yb is thiazolyl or pyrimidinyl;
b) Zb is a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or fluoro;
c) Zb is benzothienyl or indolyl,
wherein Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl substituent is unsubstituted or substituted with one trifluoromethyl or fluoro substituent; and any combination of embodiments a) through c) above, provided that it is understood that combinations in which different embodiments of the same substituent would be combined are excluded;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
An embodiment of the present invention is directed to a compound of Formula
(lb)
Figure imgf000022_0001
Formula (lb)
wherein:
Yb is thiazolyl or pyrimidinyl;
Zb is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is
trifluoromethyl or fluoro;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof.
An embodiment of the present invention is directed to a compound of Formula
(lb)
Figure imgf000022_0002
Formula (lb)
wherein:
Yb is thiazolyl or pyrimidinyl;
Zb is a heteroaryl selected from the group consisting of benzothienyl and indolyl;
wherein the heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one trifluoromethyl or fluoro substitutent;
and enantiomers, diastereomers, solvates, and pharmaceutically acceptable salts thereof. An embodiment of the present invention is directed to a compound of Formula
Figure imgf000023_0001
Formula (lb)
that is
the compound wherein Yb is pyrimidin-2-yl, and Zb is 6-trifluoromethyl-benzothien-2- yl; or
the compound wherein Yb is pyrimidin-2-yl, and Zb is l-(4-trifluoromethyl-phenyl)-lH- indol-5-yl;
or a pharmaceutically acceptable salt thereof.
For use in medicine, salts of compounds of Formula (la) and (lb) refer to nontoxic "pharmaceutically acceptable salts." Other salts may, however, be useful in the preparation of compounds of Formula (la) and (lb) or of their pharmaceutically acceptable salts thereof. Suitable pharmaceutically acceptable salts of compounds of Formula (la) and (lb) include acid addition salts that can, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable acid such as, hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. Furthermore, where the compounds of Formula (la) and (lb) carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts such as, sodium or potassium salts; alkaline earth metal salts such as, calcium or magnesium salts; and salts formed with suitable organic ligands such as, quaternary ammonium salts. Thus, representative pharmaceutically acceptable salts include acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate,
glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, oleate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate.
Representative acids and bases that may be used in the preparation of pharmaceutically acceptable salts include acids including acetic acid, 2,2-dichloroactic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D- glucoronic acid, L-glutamic acid, a-oxo-glutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, (+)-L-lactic acid, (±)-DL-lactic acid, lactobionic acid, maleic acid, (-)-L-malic acid, malonic acid, (±)-DL-mandelic acid,
methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, 1- hydroxy -2 -naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, phosphoric acid, L-pyroglutamic acid, salicylic acid, 4- amino-salicylic acid, sebaic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid; and bases including ammonia, L-arginine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)-ethanol, ethanolamine, ethylenediamine, N-methyl-glucamine, hydrabamine, lH-imidazole, L-lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholin, piperazine, potassium hydroxide, l-(2-hydroxyethyl)-pyrrolidine, sodium hydroxide, triethanolamine, tromethamine, and zinc hydroxide.
Embodiments of the present invention include prodrugs of compounds of
Formula (la) and (lb). In general, such prodrugs will be functional derivatives of the compounds that are readily convertible in vivo into the required compound. Thus, in the methods of treating or preventing embodiments of the present invention, the term "administering" encompasses the treatment or prevention of the various diseases, conditions, syndromes and disorders described with the compound specifically disclosed or with a compound that may not be specifically disclosed, but which converts to the specified compound in vivo after administration to a patient.
Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
Where the compounds according to embodiments of this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. The skilled artisan will understand that the term compound as used herein, is meant to include solvated compounds of Formula (la) and (lb).
Where the processes for the preparation of the compounds according to certain embodiments of the invention give rise to mixture of stereoisomers, these isomers may be separated by conventional techniques such as, preparative chromatography. The compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. The compounds may, for example, be resolved into their component enantiomers by standard techniques such as, the formation of diastereomeric pairs by salt formation with an optically active acid such as, (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.
One embodiment of the present invention is directed to a composition, including a pharmaceutical composition, comprising, consisting of, and/or consisting essentially of the (+)-enantiomer of a compound of Formula (la) or (lb) wherein said composition is substantially free from the (-)-isomer of said compound. In the present context, substantially free means less than about 25 %, preferably less than about 10 %, more preferably less than about 5 %, even more preferably less than about 2 % and even more preferably less than about 1 % of the (-)-isomer calculated as. (mass (+) - enantiomer)
% (+) - enantiomer x lOO
(mass (+) - enantiomer) + (mass(-) - enantiomer)
Another embodiment of the present invention is a composition, including a pharmaceutical composition, comprising, consisting of, and consisting essentially of the (-)-enantiomer of a compound of Formula (la) or (lb) wherein said composition is substantially free from the (+)-isomer of said compound. In the present context, substantially free from means less than about 25 %, preferably less than about 10 %, more preferably less than about 5 %, even more preferably less than about 2 % and even more preferably less than about 1 % of the (+)-isomer calculated as
(mass (-) - enantiomer)
% (-) - enantiomer x lOO
(mass (+) - enantiomer) + (mass(-) - enantiomer)
During any of the processes for preparation of the compounds of the various embodiments of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups such as those described in Protective Groups in Organic Chemistry, Second Edition, J.F.W. McOmie, Plenum Press, 1973; T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 ; and T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, 1999. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.
Even though the compounds of embodiments of the present invention (including their pharmaceutically acceptable salts and pharmaceutically acceptable solvates) can be administered alone, they will generally be administered in admixture with a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and/or a pharmaceutically acceptable diluent selected with regard to the intended route of administration and standard pharmaceutical or veterinary practice. Thus, particular embodiments of the present invention are directed to pharmaceutical and veterinary compositions comprising compounds of Formula (la) or (lb) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and/or pharmaceutically acceptable diluent. By way of example, in the pharmaceutical compositions of embodiments of the present invention, the compounds of Formula (la) and (lb) may be admixed with any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilizing agent(s), and combinations thereof.
Solid oral dosage forms such as, tablets or capsules, containing the compounds of the present invention may be administered in at least one dosage form at a time, as appropriate. It is also possible to administer the compounds in sustained release formulations.
Additional oral forms in which the present inventive compounds may be administered include elixirs, solutions, syrups, and suspensions; each optionally containing flavoring agents and coloring agents.
Alternatively, compounds of Formula (la) and (lb) can be administered by inhalation (intratracheal or intranasal) or in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder. For example, they can be incorporated into a cream comprising, consisting of, and/or consisting essentially of an aqueous emulsion of polyethylene glycols or liquid paraffin. They can also be incorporated, at a concentration of between about 1 % and about 10 % by weight of the cream, into an ointment comprising, consisting of, and/or consisting essentially of a wax or soft paraffin base together with any stabilizers and preservatives as may be required. An alternative means of administration includes transdermal administration by using a skin or transdermal patch.
The pharmaceutical compositions of the present invention (as well as the compounds of the present invention alone) can also be injected parenterally, for example, intracavernosally, intravenously, intramuscularly, subcutaneously, intradermally, or intrathecally. In this case, the compositions will also include at least one of a suitable carrier, a suitable excipient, and a suitable diluent.
For parenteral administration, the pharmaceutical compositions of the present invention are best used in the form of a sterile aqueous solution that may contain other substances, for example, enough salts and monosaccharides to make the solution isotonic with blood.
For buccal or sublingual administration, the pharmaceutical compositions of the present invention may be administered in the form of tablets or lozenges, which can be formulated in a conventional manner.
By way of further example, pharmaceutical compositions containing at least one of the compounds of Formula (la) or (lb) as the active ingredient can be prepared by mixing the compound(s) with a pharmaceutically acceptable carrier, a pharmaceutically acceptable diluent, and/or a pharmaceutically acceptable excipient according to conventional pharmaceutical compounding techniques. The carrier, excipient, and diluent may take a wide variety of forms depending upon the desired route of administration (e.g., oral, parenteral, etc.). Thus, for liquid oral preparations such as, suspensions, syrups, elixirs and solutions, suitable carriers, excipients and diluents include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents and the like; for solid oral preparations such as, powders, capsules, and tablets, suitable carriers, excipients and diluents include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Solid oral preparations also may be optionally coated with substances such as, sugars, or be enterically coated so as to modulate the major site of absorption and disintegration. For parenteral administration, the carrier, excipient and diluent will usually include sterile water, and other ingredients may be added to increase solubility and preservation of the composition. Injectable suspensions or solutions may also be prepared utilizing aqueous carriers along with appropriate additives such as, solubilizers and
preservatives.
A therapeutically effective amount of a compound of Formula (la) or (lb) or a pharmaceutical composition thereof includes a dose range from about 0.1 mg to about 3000 mg, or any particular amount or range therein, in particular from about 1 mg to about 1000 mg, or any particular amount or range therein, or, more particularly, from about 10 mg to about 500 mg , or any particular amount or range therein, of active ingredient in a regimen of about 1 to about 4 times per day for an average (70 kg) human; although, it is apparent to one skilled in the art that the therapeutically effective amount for a compound of Formula (la) or (lb) will vary as will the diseases, syndromes, conditions, and disorders being treated.
For oral administration, a pharmaceutical composition is preferably provided in the form of tablets containing about 1.0, about 10, about 50, about 100, about 150, about 200, about 250, and about 500 milligrams of a compound of Formula (la) or (lb).
Advantageously, a compound of Formula (la) or (lb) may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three and four times daily.
Optimal dosages of a compound of Formula (la) or (lb) to be administered may be readily determined and will vary with the particular compound used, the mode of administration, the strength of the preparation and the advancement of the disease, syndrome, condition or disorder. In addition, factors associated with the particular subject being treated, including subject gender, age, weight, diet and time of administration, will result in the need to adjust the dose to achieve an appropriate therapeutic level and desired therapeutic effect. The above dosages are thus exemplary of the average case. There can be, of course, individual instances wherein higher or lower dosage ranges are merited, and such are within the scope of this invention.
Compounds of Formula (la) or (lb) may be administered in any of the foregoing compositions and dosage regimens or by means of those compositions and dosage regimens established in the art whenever use of a compound of Formula (la) or (lb) is required for a subject in need thereof.
As MGL Inhibitors, the compounds of Formula (la) and (lb) are useful in methods for treating and preventing a disease, a syndrome, a condition or a disorder in a subject, including an animal, a mammal and a human in which the disease, the syndrome, the condition or the disorder is affected by the modulation, including inhibition, of the MGL enzyme. Such methods comprise, consist of and/or consist essentially of administering to a subject, including an animal, a mammal, and a human in need of such treatment or prevention a therapeutically effective amount of a compound, salt or solvate of Formula (la) or (lb). GENERAL SYNTHETIC METHODS
Representative compounds of the present invention can be synthesized in accordance with the general synthetic methods described below and illustrated in the schemes and examples that follow. Since the schemes are an illustration, the invention should not be construed as being limited by the chemical reactions and conditions described in the schemes. The various starting materials used in the schemes and examples are commercially available or may be prepared by methods well within the skill of persons versed in the art. The variables are as defined herein. Abbreviations used in the instant specification, particularly the schemes and examples, are as follows:
AcCl acetyl chloride
AcOH glacial acetic acid
aq. aqueous
Bn or Bzl benzyl
Boc tert-butyloxycarbonyl
cone. Concentrated
DCC N,N -dicyclohexyl-carbodiimi.de
DCE 1,2-dichloroethane
DCM dichloromethane
DIPEA diisopropyl-ethyl amine
DMAP 4-dimethylaminopyridine
DMF N,N-dimethylformamide
DMSO dimethylsulfoxide
DPPA diphenylphosphoryl azide
EDC N-(3 -dimethylaminopropyl)-N' -ethylcarbodiimide hydrochloride
ESI electrospray ionization
EtOAc ethyl acetate
EtOH ethanol
h hour(s)
HATU 0-( lH-7-azabenzotriazol- 1 -yl)- 1, 1,3,3- tetramethyluronium hexafluorophosphate
HBTU 0-( lH-benzotriazol- 1 -yl)- 1, 1,3,3- tetramethyluronium hexafluorophosphate
ΗΕΚ human embryonic kidney
HEPES (4-(2 -hydroxy ethyl)- 1 -piperazineethane
sulfonic acid
HPLC high performance liquid chromatography mCPBA meto-chloroperoxybenzoic acid
MeCN acetonitrile MeOH methanol
MeOTf methyl triflate
MHz megahertz
min minutes
MS mass spectrometry
NMR nuclear magnetic resonance
PIPES Piperazine-N,N'-bis (2 -ethanesulfonic
PyBrOP bromo-tris-pyrrolidinophosphonium
hexafluorophosphate
RP reverse-phase
Rt retention time
TEA or Et3N triethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography
TMS tetramethylsilane
Scheme A illustrates a route for the synthesis of compounds of Formula (la), wherein Y and Z are as defined herein.
Scheme A
Figure imgf000031_0001
A6 Formula (la)
A compound of formula Al is either commercially available or may be prepared by known methods described in the scientific literature. A compound of formula Al may undergo a reductive amination with a compound of formula A2 (wherein P is a conventional amino protecting group such as, Boc, Fmoc, Cbz, and the like) in the presence of a hydride source such as, sodium triacetoxyborohydride to afford a compound of formula A3. The Y group of the present invention may be introduced by reaction with a compound of formula A4, Y-X, (wherein X is a suitable leaving group such as, bromo or iodo, and, under certain reaction conditions, may be chloro or triflate) in the presence of copper iodide, 1,10— phenanthroline, and potassium phosphate, to afford a compound of formula A5. Removal of the protecting group (P) by conventional methods affords a compound of formula A6. A compound of formula A6 may be treated with a carboxylic acid of formula A7 wherein Q is hydroxy, in the presence of an appropriate coupling agent such as, HATU, DCC, EDC, HBTU, PyBrOP, and the like; and optionally in the presence of a base such as, DIPEA, to afford a compound of Formula (la). Similarly, an acid chloride of formula A7 wherein Q is chloro may be used to effect the acylation of a compound of formula A6. In such case a non-nucleophilic base such as, pyridine, may be added to afford a compound of Formula (la).
Scheme B illustrates a route for the synthesis of compounds of Formula (lb), wherein Yb and Zb are as defined herein.
Scheme B
Figure imgf000032_0001
Bg 2. Amino Deprotection B7
Figure imgf000032_0002
Formula (lb)
A compound of formula A2 is commercially available and may be deprotected using conventional chemistry to afford the corresponding amine of formula Bl. Treatment of a compound of formula Bl with a compound of formula B2, in an analogous manner to the procedure described in scheme A for compound A7, affords a compound of formula B3. Reductive amination with a diamine of formula B4 (wherein P is a conventional amino protecting group) in the presence of a hydride source such as, sodium triacetoxyborohydride, affords a compound of formula B5. Acylation with an acid chloride of formula B6 followed by conventional removal of protecting group (P) provides the cyclized product of formula B7. A compound of formula B7 may be treated with a compound of formula B8 (wherein Xb is fluoro, bromo, chloro, iodo, or triflate) under suitable reaction conditions to afford a compound of Formula (lb). For example, when Yb is 2-pyrimidinyl, 4-pyrimidinyl, 2-pyridinyl, 2-thiazolyl, or 2- oxazolyl, the addition of a compound of formula B8 in the presence of a base such as, potassium carbonate or sodium bicarbonate, will provide the desired compounds of Formula (lb). Alternatively, other Yb groups of the present invention may be incorporated by the addition of Yb- b (B8) in the presence of a palladium catalyst, optionally with a phosphine ligand, and in the presence of an inorganic base such as, potassium carbonate.
Scheme C illustrates an alternate route for the synthesis of compounds of Formula (lb), wherein Yb and Zb are as defined herein.
Scheme C
Figure imgf000033_0001
Formula (lb)
A compound of formula B8 (either commercially available or prepared by known methods found in the scientific literature) may be treated with a compound of formula B4 to afford a compound of formula CI. More specifically, when Yb of formula B8 is 2-pyrimidinyl, 4-pyrimidinyl, 2-pyridinyl, 2-thiazolyl, or 2-oxazolyl, the addition of a compound of formula B8 in the presence of a base such as, potassium carbonate or sodium bicarbonate, will provide the desired compounds of formula CI. Alternatively, other Yb groups of the present invention may be incorporated by the addition of B8 in the presence of a palladium catalyst, optionally with a phosphine ligand, and in the presence of an inorganic base such as, potassium carbonate. Conventional removal of the amino protecting group (P) followed by reductive alkylation with a compound of formula A2 affords a compound of formula C2. Acylation with a compound of formula B6 followed by treatment with a strong base such as, sodium hydride, affords the cyclized product of formula C3. Amino deprotection gives the corresponding free amine of formula C4, which, upon acylation with a compound of formula B2 as previously described, affords compounds of Formula (lb).
Scheme D illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is a heteroaryl substituted with an optionally substituted phenyl group (ArD). For illustrative purposes only, the heteroaryl ring is represented by an indole.
Scheme D
Figure imgf000034_0001
K3PO4, Cul
toluene
110 °C
A compound of formula Dl is either commercially available or may be prepared by known methods described in the scientific literature. The compound Dl may be treated with an phenyl iodide of formula D2 in the presence of copper iodide, trans-N, '- dimethylcyclohexane-l,2-diamine, and potassium phosphate to afford a compound of formula D3. Subsequent saponification affords useful carboxylic acid intermediates of formula D4. Scheme E illustrates the preparation of certain useful intermediates of formula A7 and B2 (Q is hydroxy) wherein Z/Zb is benzoxazolyl substituted with an optionally substituted phenyl group (ArE).
Scheme E
Saponification
Figure imgf000035_0001
E4
A compound of formula El is either commercially available or may be prepared by known methods described in the scientific literature. The compound of formula El may be treated with an optionally substituted benzoyl chloride of formula E2 in an organic solvent such as, dioxane, at an elevated temperature to afford the substituted benzoxazole of formula E3. Subsequent saponification affords useful carboxylic acid intermediates of formula E4.
Scheme F illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is an optionally substituted benzothienyl group, and RF represents appropriate substituents as defined in Formula (la) and Formula (lb).
Scheme F
Figure imgf000035_0002
F1 F2 F3
A compound of formula Fl is either commercially available or may be prepared by known methods described in the scientific literature. The compound of formula Fl may be treated with thionyl chloride in an aprotic organic solvent, followed by treatment with methanol to afford a compound of formula F2. Subsequent
saponification affords useful carboxylic acid intermediates of formula F3. One skilled in the art will recognize that asymmetrically substituted compounds of formula Fl could lead to mixtures of positional isomers upon cyclization with thionyl chloride. The isomers may then be separated and isolated using conventional chromatography known to those skilled in the art.
Scheme G illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is phenyl substituted by an optionally substituted phenylmethyl (ArT-methyl) group.
Scheme G
Figure imgf000036_0001
A compound of formula Gl is either commercially available or may be prepared by known methods described in the scientific literature. The compound of formula Gl may be treated with with an appropriately substituted boronic acid or ester of formula G2, in the presence of a palladium catalyst and a suitable base such as, potassium carbonate to afford a compound of formula G3. Subsequent saponification affords useful carboxylic acid intermediates of formula G4.
Scheme H illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is a benzothienyl group substituted with a fluoro substituent and an optionally substituted phenyl group (ArH).
Scheme H
Figure imgf000036_0002
A compound of formula HI is either commercially available or may be prepared by known methods described in the scientific literature. The compound of formula HI may be cross-coupled with a boronic acid or ester (H2) in the presence of a palladium catalyst; and in the presence of a suitable base such as, potassium carbonate, to afford a compound of formula H3. Saponification affords the corresponding carboxylic acid H4, which may be treated with N-fluorobenzenesulfonimide in the presence of an organometallic base such as, n-butyllithium, to afford the fluorinated compound of formula H5.
Scheme I illustrates the preparation of certain useful intermediates of formulae A7 and B2 (Q is hydroxy) wherein Z/Zb is a benzothiazole group substituted with an optionally substituted phenyl group (Ari).
Scheme I
Figure imgf000037_0001
13 14
A compound of formula II is either commercially available or may be prepared by known methods described in the scientific literature. The compound of formula II may be cross-coupled with a boronic acid or ester (12) in the presence of a palladium catalyst; and in the presence of a suitable base such as, potassium carbonate, to afford a compound of formula 13. Saponification affords the corresponding carboxylic acid of formula 14.
Example 1
4-Pyrimidin-2-yl- 1 -(1 - { [6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} azetidin-3
-2-one, Cpd 23
Figure imgf000037_0002
Step A. l-(6-Trifluoromethyl-benzo[b]thiophene-2-carbonyl)-azetidin-3-one le. A solution of N-Boc-azetidin-3-one la (171 mg, 1.0 mmol) and TFA (1 mL) in CH2CI2 (4 mL) was stirred at room temperature for 2h. The solution was concentrated to give intermediate lb, which was used in next step without further purification. To a suspension of 6-trifluoromethyl-benzo[b]thiophene-2-carboxylic acid lc (246 mg, 1.0 mmol) in CH2CI2 (10 mL) was added oxalyl chloride (0.105 mL, 1.2 mmol), followed by DMF (2 drops). The reaction mixture was stirred at room temperature for 4 h, and concentrated to give the acid chloride Id, which was used in next step without further purification. To a solution of lb (1.0 mmol) and Ets (0.835 mL, 6.0 mmol) in CH2CI2 (7 mL) at 0 °C was added a solution of Id (1.0 mmol) in CH2CI2 (5 mL). The reaction mixture was slowly warmed to room temperature in 2 h, and was quenched with aq. aHC03. The resulting mixture was extracted with CH2CI2. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 40% EtOAc/heptane) gave intermediate le as white solid (100 mg).
Step B. {2-[l-(6-Trifluoromethyl-benzo[b]thiophene-2-carbonyl)-azetidin-3- ylamino]-ethyl}-carbamic acid tert-butyl ester lg.
To a mixture of le (62 mg, 0.207 mmol), N-Boc-ethylenediamine If (100 mg, 0.62 mmol), and acetic acid (0.3 mL) in 1,2-dichloroethane (3 mL) was added Na(OAc)3BH (53 mg, 0.25 mmol). The reaction mixture was stirred at room temperature overnight, and was then quenched with aq. aHC03. The resulting mixture was extracted with CH2CI2. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 4% MeOH/CELC^) gave intermediate lg as colorless oil (30 mg).
Step C. Cpd 23
To a solution of lg (30 mg, 0.068 mmol) and Et3N (0.04 mL, 0.28 mmol) in CH2C12 (3 mL) at 0 °C was added chloroacetyl chloride (1 1.5 mg, 0.10 mmol). The reaction was stirred at 0 °C for 1.5h, and was then quenched with aq. aHC03. The resulting mixture was extracted with CH2CI2. The organic solution was dried over a2S04 and concentrated. The resulting residue was stirred with TFA (0.5 mL) in CH2CI2 (2 mL) for 1.5 h. The solution was concentrated, and the residue was stirred in aq. aHC03 (2 mL) and THF (3 mL) for 24 h. To the resulting intermediate lh in this mixture of aq. aHC03 and THF was added K2CO3 (28 mg, 0.20 mmol) and 2-bromopyrimidine (32 mg, 0.20 mmol). The reaction mixture was heated to reflux for 16 h, and was then extracted with CH2C12. The organic solution was dried over Na2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave Cpd 23 as a white solid (7 mg). MS 462 (M+H+).
Example 2
4-Pyrimidin-2-yl- 1 -[ 1 -( { 1 -[4-(trifluoromethyl)phenyl]- 1 H-indol-5- yl}carbonyl)azetidin-3-yl]piperazin-2-one, Cpd 24
Figure imgf000039_0001
1,2-dichloroethane
Figure imgf000039_0002
Step A. [2-(Pyrimidin-2-ylamino)-ethyl]-carbamic acid tert-butyl ester 2a. A mixture of 2-bromopyrimidine li (600 mg, 3.77 mmol), N-Boc-ethylenediamine If (1000 mg, 6.24 mmol), and K2C03 (781 mg, 5.66 mmol) in dioxane (20 mL) and H20 (10 mL) was heated to reflux for 9 h. The mixture was concentrated to remove most of dioxane, and the resulting mixture was extracted with EtOAc. The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated. Purification by column chromatography (silica gel, 50% EtOAc/heptane) gave intermediate 2a as white solid (770 mg).
Step B. 3-[2-(Pyrimidin-2-ylamino)-ethylamino]-azetidine-l-carboxylic acid tert-butyl ester 2b.
A solution of intermediate 2a (610 mg, 2.56 mmol) in TFA (2 mL) and CH2C12 (8 mL) was stirred at room temperature for 2 h. The solution was concentrated. The resulting residue was stirred with N-Boc-azetidin-3-one la (526 mg, 3.07 mmol) and
Na(OAc)3BH (651, 3.07 mmol) in 1,2-dichloroethane (8 mL) and HOAc (0.8 mL) at room temperature overnight. Additional la (175 mg, 1.02 mmol) and Na(OAc)sBH (217 mg, 1.02 mmol) was added. The reaction was stirred for another 5 h before it was quenched with aq. aHC03. The resulting mixture was extracted with ΟΗ2(¾. The organic solution was dried over Na2S04 and concentrated. Purification by column chromatography (silica gel, 6% MeOH/CH^CL.) gave intermediate 2b as light yellow solid (268 mg).
Step C. 3-(2-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 2 c.
To a solution of intermediate 2b (210 mg, 0.72 mmol) and Ets (0.50 mL, 3.58 mmol) in CH2CI2 (8 mL) at 0 °C was added chloroacetyl chloride (0.086 mL, 1.07 mmol). The reaction was slowly warmed up to room temperature in 3 h. It was quenched with aq. aHC03 and extracted with CH2CI2. The organic solution was dried over Na2S04 and concentrated. To a solution of the above crude product (150 mg, 0.41 mmol) in DMF (3 mL) at 0 °C was added NaH (60% in oil, 24 mg, 0.60 mmol). The reaction was slowly warmed up to room temperature in 3 h. It was quenched with H20 and extracted with EtOAc. The organic solution was washed with aq. NaCl, dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave intermediate 2c as white solid (47 mg).
Step D. Cpd 24
A solution of intermediate 2c (47 mg, 0.14 mmol) in CH2CI2 (2 mL) and TFA (0.5 mL) was stirred at room temperature for 1.5 h. The reaction mixture was concentrated and the resulting residue was dissolved in CH2CI2 (5 mL), and IN HC1 in ether (0.42 mL, 0.42 mmol) was added. The mixture was concentrated to give intermediate 2d, which was used in next step without further purification. A mixture of intermediate 2d (0.14 mmol), l-(4-trifluoromethyl-phenyl)-lH-indole-5-carboxylic acid 2e (56 mg, 0.18 mmol), HATU (70 mg, 0.18 mmol), and Et3N (0.1 1 mL, 0.85 mmol) in CH2C12 (3 mL) was stirred at room temperature overnight. The reaction mixture was diluted with ether and washed with aq. aHC03 and aq. NaCl. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave Cpd 24 (46 mg). MS 521 (M+H+). Example 3
1 -Pyrimidin-2-yl-4-[ 1 -( { 1 -[4-(trifluoromethyl)phenyl]- 1 H-indol-5- yl}carbonyl)azetidin-3-yl]piperazin-2-one, Cpd 3
Figure imgf000041_0001
Step A. 3-Oxo-4-pyrimidin-2-yl-piperazine-l-carboxylic acid tert-butyl ester
3b.
To l-Boc-3-oxopiperazine 3a (390 mg, 1.95 mmol) in DMF (5 mL) at 0 °C was added NaH (60% in oil, 97 mg, 2.44 mmol). The reaction was stirred at 0 °C for 25 min before a solution of 2-bromopyrimidine li (465 mg, 2.92 mmol) in DMF (2 mL) was added. The reaction was slowly warmed up to room temperature overnight. To the reaction mixture was added FLO, and the resulting mixture was extracted with EtOAc. The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave 3b as yellow oil (120 mg).
Step B. 3-(3-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 3 c.
A solution of intermediate 3b (120 mg, 0.43 mmol) in TFA (0.75 mL) and CH2C12 (3 mL) was stirred at room temperature for 1.5 h. The solution was concentrated. The resulting residue was stirred with N-Boc-azetidin-3-one la (81 mg, 0.47 mmol) and Na(OAc)3BH (100, 0.47 mmol) in 1,2-dichloroethane (4 mL) and HOAc (0.2 mL) at room temperature overnight. The reaction was quenched with aq. aHC03. The resulting mixture was extracted with CH2CI2. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Ci2) gave intermediate 3c as light yellow solid (15 mg).
Step C. Cpd 3
Cpd 3 was prepared according to the procedures described in Example 2, Step D, using intermediates 3c and 2e as starting materials. MS 521 (M+H+).
The following compounds were prepared using the procedures described in Example 3, except using commercially available l-phenyl-piperazin-2-one as starting material.
Figure imgf000042_0002
Example 3 a
Figure imgf000042_0001
A. Methyl l-(4-fluorophenyl)-indole-5-carboxylate, 3g. A mixture of methyl indole-5-carboxylate 3e (0.5 g, 2.85 mmol), 1 -bromo-4-fluoro-benzene 3f (2 mL, 18.21 mmol), Cul (0.544 g, 2.85 mmol), and K2C03 (0.591 g, 4.28 mmol) was heated under microwave at 220 °C for 2.5 hours. The reaction mixture was diluted with CH2CI2 and filtered. The solution was concentrated and the residue was purified by flash column chromatography (silica gel, 15% EtOAc/heptane) to give 3g (0.58 g).
I. l-(4-fluorophenyl)-indole-5-carboxylic acid, 3h. A mixture of methyl l-(4- fluorophenyl)-indole-5-carboxylate 3g (0.58 g, 2.15 mmol) and LiOH H20 (0.36 g, 8.6 mmol) in THF (15 niL) and Ι¾0 (10 niL) was stirred at room temperature for 5 days. Aqueous 10% HC1 solution was added to the reaction mixture to adjust pH = 3 ~ 4. The resulting mixture was extracted with EtOAc (2x). The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated to give 3h (0.5 g).
Following the procedure described above for Example 3 a and substituting the appropriate reagents, starting materials, and purification methods known to those skilled in the art, the following intermediate was prepared:
Figure imgf000043_0001
Cpd 2e
Example 4
-Fluorophenyl)- lH-indol-5-yl]carbonyl}azetidin-3-yl)-l-pyrimidin-2- -2-one, Cpd 4
Figure imgf000043_0002
Step A. 3-(3-Oxo-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester
4b. To a mixture of 3-oxopiperazine 4a (490 mg, 4.89 mmol) and N-Boc-azetidin-3-one la (922 mg, 5.38 mmol) in 1,2-dichloroethane (10 mL) and HOAc (0.5 mL) was added Na(OAc)sBH (1141, 5.38 mmol). The reaction was stirred at room temperature overnight. Additional la (335 mg, 1.96 mmol) and Na(OAc)sBH (415 mg, 1.96 mmol) was added. The reaction was stirred for another 24 h before it was quenched with aq. aHC03. The resulting mixture was extracted with (¾(¾. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 4% MeOH/CH2Cl2) gave intermediate 4b as white solid (860 mg).
Step B. 3-(3-Oxo-4-pyrimidin-2-yl-piperazin-l-yl)-azetidine-l-carboxylic acid tert-butyl ester 3 c.
A mixture of intermediate 4b (52 mg, 0.204 mmol), 2-bromopyrimidine li (65 mg, 0.41 mmol), Cul (8 mg, 0.04 mmol), 1,10-phenanthroline (18 mg, 0.10 mmol), and K.3P04 (95 mg, 0.45 mmol) in toluene (1 mL) was heated at 110 °C under 2 for 9 h. The reaction mixture was diluted with CH2C12 and filtered. The solution was concentrated and purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave intermediate 3c as light yellow solid (25 mg).
Step C. Cpd 4
Cpd 4 was prepared according to the procedures described in Example 2, Step D, using intermediates 3c and l-(4-fluoro-phenyl)-lH-indole-5-carboxylic acid 3h as starting materials. MS 471 (M+H+).
The following compounds were prepared following the procedures described in
Example 4.
Figure imgf000044_0001
Figure imgf000045_0001
Example 5
4-( 1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} azetidin-3 -yl)- 1 - -2-ylpiperazin-2-one, Cpd 8
Figure imgf000045_0002
A solution of intermediate 3c (prepared by procedures described in Example 4) in CH2CI2 (2 mL) and TFA (0.5 mL) was stirred at room temperature for lh. The solution was concentrated, and the residue was dissolved in CH2CI2 (2.5 mL). To the resulting solution at 0 °C was added Et3N (0.1 1 niL, 0.79 mmol), followed by a solution of 3- chloro-6-trifluoromethyl-benzo[b]thiophene-2-carbonyl chloride 5a (prepared from 3- chloro-6-trifluoromethyl-benzo[b]thiophene-2-carboxylic acid following the procedures described in Example 1, Step A) (0.14 mmol) in CH2CI2 (1.5 mL). The reaction was slowly warmed up to room temperature in 2 h, and was then quenched with aq.
aHC03. The mixture was extracted with CH2CI2. The organic solution was dried over a2S04 and concentrated. Purification by column chromatography (silica gel, 3% MeOH/CH2Cl2) gave Cpd 8 as yellow solid (28 mg). MS 496 (M+H+).
The following compounds were prepared following the procedures described in Example 5.
Figure imgf000046_0001
Example 6
4-( 1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} azetidin-3 -yl)- 1 - phenylpiperazin-2-one, Cpd 14
Figure imgf000047_0001
Cpd 14 was prepared following the procedures described in Example 5 with intermediates 6a and 5a. Intermediate 6a was prepared following the procedures described in Example 3, Step B, using commercially available 1 -phenyl-piperazin-2- one as starting material. MS 494 (M+H+).
Examples 7-26 provide synthetic routes to useful intermediates for the preparation of compounds of Formula (I).
Example 7
Figure imgf000047_0002
A. Ethyl l-(3-trifluoromethyl-phenyl)-lH-indazole-5-carboxylate, 7c and Ethyl 2-(3-trifluoromethyl-phenyl)-2H-indazole-5-carboxylate, 7d. A mixture of ethyl lH-Indazole-5-carboxylate 7a (150 mg, 0.79 mmol), l-bromo-3- trifluoromethylbenzene 7b (0.13 mL, 0.95 mmol), Cul (22.5 mg, 0.12 mmol), trans-N, N'-dimethylcyclohexane-l,2-diamine (0.056 mL, 0.36 mmol), and K3PO4 (0.37 g, 1.74 mmol) in toluene (1.5 mL) was heated at 1 10 °C for 16 hours. The reaction mixture was diluted with CH2CI2 and filtered. The solution was concentrated and the residue was purified by flash column chromatography (silica gel, 10% EtO Ac/heptane) to give 7c (190 mg), followed by 7d (37 mg).
B. l-(3-Trifluoromethyl-phenyl)-lH-indazole-5-carboxylic acid, 7e and 2- (3-Trifluoromethyl-phenyl)-2H-indazole-5-carboxylic acid, 7f. A mixture of 7c and 7d and LiOH in THF (120 mL) and FLO (60 mL) was stirred at room temperature for 5 days. Aqueous 10% HC1 solution was added to the reaction mixture to adjust pH = 3 ~ 4. The resulting mixture was extracted with EtO Ac (2x). The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated to give 7e and 7f.
Example 8
Figure imgf000048_0001
8d 8e
A. Methyl 2-(4-fluoro-benzoylamino)-3-hydroxy-benzoate, 8c. A solution of 1.0 g (4.9 mmol) of methyl 2-amino-3-hydroxybenzoate 8a, 1.03 g (7.4 mmol) of 4- fluorobenzoic acid 8b, 10 mL DMF and 2.9 mL (20.6 mmol) of TEA were placed into a flask and stirred for 10 min. HATU (7.4 mmol, 2.8 g) was added and the reaction was stirred overnight. The reaction mixture was poured into water and extracted with EtO Ac. The organics were washed with water and brine and the solvent was evaporated to give 1.2 g of crude product, methyl 2-(4-fluoro-benzoylamino)-3- hydroxy -benzoate, 8c, which was used without purification. MS m/z (M+H+) 290.1.
B. Methyl 2-(4-fluorophenyl)benzo[d]oxazole-4-carboxylate, 8d. Methyl 2- (4-fluoro-benzoylamino)-3-hydroxy -benzoate 8c (7.4 mmol, 1.2 g crude) and 1.3 g (7.5 mmol) of p-toluenesulfonic acid were refluxed in 10 mL of xylene overnight. After cooling saturated aHC03 was added and the resulting mixture was extracted with EtO Ac. The organic solvent was evaporated to give 1.1 g (55%) of methyl 2-(4- fluorophenyl)benzo[d]oxazole-4-carboxylate, 8d. MS m/z (M+H+) 272.0. C. 2-(4-Fluorophenyl)-benzo[d]oxazole-4-carboxylic acid, 8e. A mixture of 1.1 g (4.0 mmol) methyl 2-(4-fluorophenyl)benzo[d]oxazole-4-carboxylate 8d and 3.7 mL of 3N aqueous NaOH in 10 mL of THF was refluxed overnight. After cooling the reaction mixture was poured into water and acidified with cone. HC1. The resulting solid was filtered and dried to give 830 mg (79%) of 2-(4-fluorophenyl)- benzo[d]oxazole-4-carboxylic acid, 8e. MS m/z (M+H+) 258.1.
Following the procedure described above for Example 8, and substituting the appropriate reagents, starting materials, and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Figure imgf000049_0001
A. Methyl 4-(4-(trifluoromethyl)benzyl)benzoate, 9b. Argon was bubbled through a mixture of methyl 4-bromobenzoate 9a (9.3 mmol, 2.0 g), 2mL of THF, and 4-trifluoromethylbenzylzinc chloride (0.5 M in THF, 46.5 mmol, 93 mL) for 5 min. Pd(dffp)Cl2 · ΟΗ2(¾(0.5 mol, 409 mg) was added and the reaction tube was capped and heated at 70 °C for 16 h. The mixture was cooled and filtered through
diatomaceous earth. Water was added to the filtrate and the resulting solid was filtered off. The organic solution was dried over MgS04 and concentrated. The crude product was purified by flash chromatography (silica gel, 0-10% EtOAc in heptane) to give 1.5 g (55%) of methyl 4-(4-(trifluoromethyl)benzyl)benzoate, 9b.
B. 4-(4-(Trifluoromethyl)benzyl)benzoic acid, 9c. A mixture of methyl 4-(4- (trifluoromethyl)benzyl)benzoate 9b (1.5 g, 5.1 mmol) and 3N aqueous NaOH was refluxed in 8 mL of THF overnight. After cooling, the mixture was poured into ice water and acidified with cone. HC1. The resulting solid was filtered and dried to give methyl 1.31 g (92%) of 4-(4-(trifluoromethyl)benzyl)benzoic acid, 9c. MS m/z (M+H+) 279.1.
Intermediate compounds were optionally prepared by an alternative procedure:
Figure imgf000050_0001
Methyl 4-(4-(trifluoromethyl)benzyl)benzoate, 9b. A mixture of 4- bromomethyl-benzoic acid methyl ester 9d (1.0 g, 4.37 mmol), 4-trifluorophenyl boronic acid 9e (0.995 g, 5.24 mmol), and Pd(PPh3)4 (50 mg, 0.044 mmol) in dioxane (15 mL) was stirred at room temperature for 1 min. Next, 4 mL of 2 M aqueous a2C03 solution was added. The resulting solution was heated at 90 °C for 5 h and was then cooled to rt. EtOAc and water were added to the reaction mixture. The organics were concentrated and purified by flash chromatography (silica gel, 5%
EtOAc/hexanes) to give methyl 4-(4-(trifluoromethyl)benzyl)benzoate, 9b.
Example 10
Figure imgf000050_0002
A. Methyl 5-phenyl-benzo[b]thiophene-2-carboxylate, 10c. A mixture of compound 10a (542.3 mg, 2 mmol), phenyl boronic acid 10b (268.2 mg, 2.2 mmol), Pd(dppf)Cl2.CH2Ci2 (98 mg, 0.12 mmol), and K2C03 (414.6 mg, 3 mmol), in a dioxane (4 mL) / water (1 mL) mixture, was placed in a capped vial and heated at 80 °C overnight. The reaction mixture was then diluted with EtOAc and water. The organic layer was concentrated under reduced pressure and purified by flash column chromatography (silica gel, 2-10% EtOAc/heptane) to give compound 10c (510 mg). MS m/z (M+H+) 269.1.
B. 5-Phenyl-benzo[b]thiophene-2-carboxylic acid, lOd. A solution of compound 10c (510 mg, 1.9 mmol) and LiOH.H20 (319 mg, 7.6 mmol) in THF/H20 (10/10 mL) was stirred at room temperature overnight. The resulting mixture was concentrated and diluted with water. The water layer was acidified with IN aqueous HC1 to pH~4 and extracted with CH2C12. The organic solution was dried over Na2S04 and concentrated to give lOd (479 mg), which was used in the next reaction without further purification. MS m/z (M+H+) 255.0.
C. 3-Fluoro-5-phenyl-benzo[b]thiophene-2-carboxylic acid, lOe. To a solution of compound lOd (507 mg, 1.99 mmol) in THF (8 mL) at -70 C was added n- BuLi (1.6 M in hexane, 2.62 mL, 4.19 mmol). The mixture was stirred at -70 C for lh; then a solution of N-fluorobenzenesulfonimide (817.3 mg, 2.59 mmol) in THF (2 mL) was slowly added. The reaction mixture was allowed to warm to room
temperature and was stirred overnight. The resulting mixture was partitioned between dilute aqueous HC1 and EtOAc. The organic solution was washed with water and brine, dried over Na2S04, and concentrated. The residue was tritiated with CH2C12, filtered and the solid dried to give compound lOe (391.9 mg). MS m/z (M+H+) 273.0.
D. 3-Fluoro-5-phenyl-benzo[b]thiophene-2-carbonyl chloride, lOf. To a solution of compound lOe (136.2 mg, 0.5 mmol) in CH2C12 (5 mL) at room temperature was added (COCl)2 (0.064 mL, 0.75 mmol), followed by DMF (0.01 mL, 0.125 mmol). The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was then concentrated to give compound lOf (light pink powder). Example 11
Figure imgf000052_0001
A. 1-teri-Butyl 6-methyl 3-(4-fluorophenyl)-lH-indole-l,6-dicarboxylate, 11c. A mixture of compound 11a (1.00 g, 2.49 mmol), 4-fluorophenyl boronic acid lib (523 mg, 3.74 mmol), Pd(OAc)2 (44.8 mg, 0.2 mmol), 2-dicyclohexylphosphino- 2',6'-dimethoxybiphenyl (SPhos, 204.7 mg, 0.5 mmol), and K3P04 (1.06 g, 4.99 mmol), in toluene (5 mL) was placed in a capped vial and heated at 90 °C under 2 for 3 h. The reaction mixture was then diluted with EtOAc and water. The organic layer was washed with brine, concentrated under reduced pressure, and purified by flash column chromatography (silica gel, 2-10% EtOAc/heptane) to give compound 11c as a light yellow solid, which was further recrystallized from heptane to obtain white solid (707 mg). MS m/z (M+H+) 370.2.
B. Methyl 3-(4-fluorophenyl)-lH-indole-6-carboxylate, lid. To a solution of compound 11c (705 mg, 1.91 mmol) in CH2CI2 (4 mL) was added trifluoroacetic acid (1.5 mL) at room temperature. The mixture was stirred at room temperature for 2 h. The resulting mixture was concentrated to give compound lid (603.3 mg) as a white solid. MS m/z (M+H+) 270.1.
C. 3-(4-Fluoro-phenyl)-lH-indole-6-carboxylic acid, lie. A solution of compound lid (303 mg, 0.79 mmol), and LiOH.H20 (132.7 mg, 3.16 mmol) in
THF/H2O (lOmL/10 mL) was stirred at 45 °C for 5 h. The resulting mixture was concentrated and diluted with water. The water layer was acidified with IN aqueous HC1 to pH~4 and extracted with CH2CI2. The organic solution was dried over Na2S04 and concentrated to give lie (249 mg). MS m/z (M+H+) 256.0. Following the procedure described above for Example 11, and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Figure imgf000053_0001
Example 12
Figure imgf000053_0002
A. Methyl 3-(4-fluoro-phenyl)-l-methyl-lH-indole-6-carboxylate, 12a. To a solution of compound lid (300 mg, 0.78 mmol) in DMF (3 mL) was added NaH (60% in mineral oil, 68.9 mg, 1.72 mmol) at 0 °C. The mixture was stirred at 0 °C for 30 min, then CH3I (0.053 mL, 0.86 mmol) was added and stirring continued at 0 °C for another 1 h. The resulting mixture was diluted with EtOAc and water. The organic layer was washed with brine and concentrated. The residue was recrystallized from heptane, filtered and the solid dried to give compound 12a (265 mg) as a light yellow solid. MS m/z (M+H+) 284.1.
B. 3-(4-Fluoro-phenyl)-l-methyl-lH-indole-6-carboxylic acid, 12b. To a solution of compound 12a (264 mg, 0.93 mmol), and LiOH.H20 (156.4 mg, 3.73 mmol) in THF/H2O (lOmL/10 mL) was stirred at 45 °C for 5 h. The resulting mixture was concentrated and diluted with water. The water layer was acidified with IN aqueous HC1 to pH~4 and extracted with CH2CI2. The organic solution was dried over Na2S04 and concentrated to give compound 12b (252 mg). MS m/z (M+H+) 270.1.
Following the procedure described above for Example 12 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compound was prepared:
Figure imgf000054_0001
xam le 13
Figure imgf000054_0002
A. Ethyl l-Methyl-3-phenyl-lH-indazole-5-carboxylate, 13b. A mixture of compound 13a (300 mg, 0.91 mmol), phenyl boronic acid 10b (133 mg, 1.09 mmol), Pd(dppf)Ci2.CH2Ci2 (40 mg, 0.055 mmol), and K2C03 (251.2 mg, 1.82 mmol), in a toluene (2 mL) / water (0.4 mL) mixture, was placed in a capped vial and heated at 90 °C overnight. The reaction mixture was then diluted with EtO Ac and water. The organic layer was concentrated under reduced pressure and purified by flash column chromatography (silica gel, 2-10% EtOAc/Heptanes) to give compound 13b (231 mg). MS m/z (M+H+) 281.1.
B. l-Methyl-3-phenyl-lH-indazole-5-carboxylic acid, 13c. A solution compound 13b (230 mg, 0.58 mmol), and LiOH.H20 (98 mg, 2.33 mmol) in THF/H20 (10/10 mL) was stirred at 45 °C for 8 h. The resulting mixture was concentrated and diluted with water. The water layer was acidified with IN aqueous HC1 to pH~4 and extracted with CH2C12. The organic solution was dried over Na2S04 and concentrated to give 13c (206 mg). MS m/z (M+H+) 253.1.
Following the procedure described above for Example 13 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Figure imgf000055_0001
Example 14
Figure imgf000055_0002
A. 3-Methyl-[l,l'-biphenyl]-4-carboxylic acid, 14b. To a suspension of compound 14a (0.025 g, 0.115 mmol), compound 10b (0.0139 g, 0.14 mmol), and Cs2C03 (0.094 g, 0.288 mmol) in dioxane (3 mL) and EtOH (1 mL) was added Pd(dppf)Ci2 (0.0084 g, 0.0115 mmol). The reaction mixture was stirred at 80 °C for 3 h. After cooling, the solid was removed by filtration and washed with CH3OH. The filtrate was concentrated. The crude product 14b was purified by reverse phase chromatography. MS m/z (M+H+) 213.1.
Following the procedure described above for Example 14 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Figure imgf000055_0003
14-11 14-12 14-13 14-14
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000056_0003
Figure imgf000056_0004
15c
A. 3-Fluoro-6-trifluoromethyl-benzo[b]thiophene-2-carboxylic acid, 15b. A solution of 6-trifluoromethyl-benzo[b]thiophene-2-carboxylic acid 15a (2.031 mmol, 0.50 g) in THF (8 mL) at -70 °C was treated with a 1.6 M solution of n-BuLi in hexanes (4.26 mmol, 2.66 mL). After 1 h at -70 °C, N-fluorobenzenesulfonimide (2.64 mmol, 0.833 g) in THF (2 mL) was slowly added and the reaction was warmed to room temperature. After 1 h the mixture was partitioned between dilute aqueous HCl and EtOAc. The organic layer was washed with water and brine, and then concentrated. The residue was triturated with (¾(¾. The off-white precipitate was filtered and collected to provide 15b.
B. 3-Fluoro-6-trifluoromethyl-benzo[b]thiophene-2-carbonyl chloride, 15c.
To compound 15b (0.14 g, 0.53 mmol) in (¾(¾ (5 mL) at room temperature was added (COCl)2 (0.051 mL, 0.58 mmol), followed by 2 drops of DMF. The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was then concentrated to give compound 15c.
Example 16
Figure imgf000057_0001
A. 1-teri-Butyl 6-methyl 3-phenyl-lH-indole-l,6-dicarboxylate, 16a. A mixture of 1-tert-butyl 6-methyl 3-iodo-lH-indole-l,6-dicarboxylate 11a (5.02 mmol, 2.016 g), phenylboronic acid 10b (7.53 mmol, 0.92 g), Pd(OAc)2 (0.402 mmol, 90 mg), Sphos 0.904 mmol, (0.37 g), and K3P04 (10.1 mmol, 2.13g) in toluene (10 mL) in sealed reaction vial was stirred at room temperature for 2 min and then heated at 90 °C under 2 for 4 h. The reaction mixture was quenched with EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 8% EtOAc/hexanes). The desired product was collected as a light yellow solid that was washed with small amount of hexanes to obtain 16a as a white solid.
B. Methyl 3-phenyl-lH-indole-6-carboxylate TFA salt, 16b. To a solution of 1-tert-butyl 6-methyl 3 -phenyl- 1H- indole- 1,6-dicarboxylate 16a (4.04 mmol, 1.42 g) in CH2CI2 (8 mL) was added 6 mL of TFA. The resulting solution was stirred for 3 h. The mixture was then concentrated and washed with hexanes to afford 16b.
C. Methyl l-methyl-3-phenyl-lH-indole-6-carboxylate, 16c. NaH (60% dispersion in mineral oil, 4.52 mmol, 186 mg) was added portion-wise to a solution of methyl 3 -phenyl- lH-indole-6-carboxy late TFA salt (2.07 mmol, 757 mg) in DMF at 0 °C and the mixture was stirred for 20 min. Methyl iodide (2.28 mmol, 0.14 mL) was added and the reaction mixture was maintained at 0 °C for 1 h. Water was then added and the reaction was extracted with EtOAc. The organics were concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give 16c.
D. l-Methyl-3-phenyl-lH-indole-6-carboxylic acid, 16d. A mixture of compound 16c (517 mg, 1.95 mmol) and LiOH (187 mg, 7.80 mmol) in
THF/MeOH/H20 (4/4/4mL) was stirred for 4 h. A 15% citric acid solution (20 mL) was added, and the mixture was then extracted with EtOAc (3X). The combined extracts were washed with brine, dried over Na2S04, filtered, and concentrated under reduced pressure. The residue, compound 16d, was dried under reduced pressurefor 18 h.
Following the procedure described above for Example 16 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Figure imgf000058_0001
Example 17
Figure imgf000058_0002
A. 1-teri-Butyl 6-methyl 3-(3-(trifluoromethyl)phenyl)-lH-indole-l,6- dicarboxylate, 17b. The title compound 17b was prepared using the method described in Example 16, substituting 17a for 10b in Step A.
B. Methyl 3-(3-(trifluoromethyl)phenyl)-lH-indole-6-carboxylate TFA salt, 17c. The title compound 17c was prepared using the method described in Example 16, substituting 17b for 16a in Step B.
C. 3-(3-(Trifluoromethyl)phenyl)-lH-indole-6-carboxylic acid, 17d. The title compound was prepared using the method described in Example 16, substituting 17c for 16c in Step D.
Following the procedure described above for Example 17 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compound was prepared:
Figure imgf000059_0001
Example 18
Figure imgf000059_0002
Figure imgf000059_0003
18g
A. l-(5-Chloro-2-fluoro-phenyl)-2,2,2-trifluoro-ethanone, 18c. To a solution of LDA (2.0 M in THF/heptane/ethylbenzene, 25.3 mmol, 12.6 mL) in dry THF was slowly added 1 -fluoro-4-chloro-bezene 18a (23.0 mmol, 2.45 mL) at -78 °C. The mixture was stirred for 1 h at -78 °C and ethyl trifluoroacetate 18b (25.3 mmol, 3.02 mL) was added. The reaction mixture was allowed to warm to room temperature overnight and was quenched with saturated aqueous NH4CI solution. The mixture was extracted with EtOAc. The organic extracts were concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give a mixture of the compound 18c along with a regio-isomeric by-product, l-(5-fluoro-2-chloro-phenyl)- 2,2,2-trifluoro-ethanone, in a ratio of 5: 1 (18c is the major product).
B. Methyl 5-chloro-3-(trifluoromethyl)benzo[b]thiophene-2-carboxylate,
18e. A solution of compound 18c (6.62 mmol, 1.5 g), methyl 2-mercaptoacetate 18d (6.62 mmol, 0.6 mL), and Ets (8.6 mmol, 1.2 mL) in acetonitrile (12 mL) was heated at 75 °C for 4 h. The reaction was diluted with EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 10% EtOAc/hexanes) to provide the compound 18e.
C. 5-Chloro-3-trifluoromethyl-benzo[b]thiophene-2-carboxylic acid, 18f. A mixture of compound 18e (574 mg, 1.95 mmol) and LiOH (187 mg, 7.80 mmol) in THF/MeOH/H20 (4/4/4mL) was stirred for 4 h. A 15% citric acid solution (20 mL) was added, and the mixture was then extracted with EtOAc (3X). The combined extracts were washed with brine, dried over Na2S04, filtered, and concentrated under reduced pressure. The residue, compound 18f, was dried under reduced pressure for 18 h and was used without purification.
D. 5-Chloro-3-trifluoromethyl-benzo[b]thiophene-2-carbonyl chloride, 18g. To compound 18f in CH2CI2 (5 mL) at room temperature was added (COCl)2, followed by 2 drops of DMF. The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was then concentrated to give compound 18g.
Exam le 19
Figure imgf000060_0001
18b
A. l-(4-Chloro-2-fluoro-phenyl)-2,2,2-trifluoro-ethanone, 19b. To a solution of n-BuLi (1.6 M in hexanes, 4.68 mmol, 2.93 mL) in dry THF was slowly added 4- chloro-2-fluoro-l-iodo-benzene 19a (3.9 mmol, 1.0 g) at -78 °C under N2. The mixture was stirred for 1 h at -78 °C and ethyl trifluoroacetate 18b (0.51 mL, 4.29 mmol) was added. The reaction was allowed to warm to room temperature overnight and was quenched with saturated aqueous NH4C1 solution. The mixture was extracted with EtOAc. The organic extracts were concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give compound 19b.
B. Methyl 6-chloro-3-(trifluoromethyl)benzo[b]thiophene-2-carboxylate, 19c. The title compound 19c was prepared using a similar method described in Example 18, substituting 19b for 18c in Step B.
Example 20
Figure imgf000061_0001
A. Methyl 3-fluoro-lH-indole-6-carboxylate, 20b. A solution of methyl 1H- indole-6-carboxylate 20a (1 1.4 mmol, 2.0 g) and N-fluoro-2,4,6-trimethylpyridinium triflate (14.8 mmol, 4.3 g) in MeOH (100 mL) was heated at reflux for 18 h. The reaction mixture was concentrated and purified by flash column chromatography (silica gel, 15-20% EtOAc/hexanes) to give compound 20b as an off-white solid.
B. Methyl 3-fluoro-l-(4-fluorophenyl)-lH-indole-6-carboxylate, 20d.
Compound 20b (0.264 mmol, 51 mg), Cul (0.0264 mmol, 5 mg) and K3P04 (0.66 mmol, 40 mg) were combined in a sealed reaction tube and the vial was back- flushed with N2. 4-fluoro-iodobezene 20c (0.264 mmol, 0.0394 mL) and Ν,Ν'- dimethylcyclohexane-l,2-diamine (0.0792 mmol, 0.0125 mL) were added via syringe, followed by toluene. The reaction mixture was heated at 95 °C for 6 h. The reaction was diluted with EtOAc and water. The reaction mixture was concentrated and purified by flash column chromatography (silica gel, 20% EtOAc/hexanes) to give compound 20d. C. 3-Fluoro-l-(4-fluorophenyl)-lH-indole-6-carboxylic acid, 20e. The title compound 20e was prepared using the method described in Example 18, Step C.
Example 21
Figure imgf000062_0001
A. 7-Fluoro-lH-indole-5-carboxylic acid, 21b. To a solution of 5-bromo-7- fluoroindole 21a (1.71 mmol, 365 mg) in THF at -60 °C was added n-BuLi (1.6 M solution in hexanes, 5.2 mmol, 3.2 mL). The solution was kept at -60 °C for 4 h and was then poured onto an excess of freshly crushed dry ice. Water was added and the mixture was acidified to pH 4. The organic phase was concentrated and the residue was purified by flash column chromatography (silica gel, 35% EtOAc/hexanes) to give compound 21b.
Figure imgf000062_0002
A. Methyl 7-methyl-lH-indole-5-carboxylate, 22c. A mixture of compound 22a (0.613 mmol, 156 mg), methylboronic acid 22b (0.92 mmol, 79 mg), Pd(OAc)2 (0.09 mmol, 20 mg), SPhos (0.215 mmol, 88mg), and K3P04 (1.23 mmol, 0.26 g) in toluene (2 mL) was heated to 100 °C for 3 h in a sealed reaction vessel. The reaction was diluted with EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 10% EtOAc/hexanes) to give compound 22c.
B. Methyl l-(4-fluorophenyl)-7-methyl-lH-indole-5-carboxylate, 22d. The title compound was prepared using the method described in Example 20, substituting 22c for 20b in Step B.
C. l-(4-Fluorophenyl)-7-methyl-lH-indole-5-carboxylate, 22e. The title compound was prepared using the method described in Example 18, Step C.
Following the procedure described above for Example 22, and substituting the appropriate reagents, starting materials, and purification methods known to those skilled in the art, the following intermediate compound was prepared:
Figure imgf000063_0001
Example 23
Figure imgf000063_0002
Figure imgf000063_0003
A. Methyl 4-amino-2-chloro-benzoate, 23b. Acetyl chloride (35.2 mmol, 2.5 mL) was added dropwise to a stirring solution of 4-amino-2-chloro-benzoic acid 23a (12.9 mmol, 2.22 g) in methanol (50 mL). The mixture was heated at reflux for 18 h, cooled, and concentrated under vacuum. The residue was taken up in EtOAc, washed with saturated aqueous aHC03 and brine, dried, and concentrated under vacuum. The crude product was purified by flash column chromatography (silica gel, 30%
EtOAc/hexanes) to give compound 23b.
B. Methyl 4-amino-2-chloro-5-iodo-benzoate, 23c. To a suspension of compound 23b (1.18 g, 6.38 mmol) and CaC03 (12.8 mmol, 1.28 g) in MeOH (13 mL) was added a solution of iodine monchloride (6.70 mmol, 1.09 g) in (¾(¾ (6 mL) dropwise at room temperature. The resulting reaction mixture was stirred at room temperature for 1.5 h. The reaction mixture was concentrated and then partitioned between EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 20-25% EtOAc/hexanes) to provide methyl 4- amino-2-chloro-5-iodo-benzoate 23c as the major the product and methyl 4-amino-2- chloro-3-iodo-benzoate 23d as the minor product.
C. Methyl 4-amino-2-chloro-5-((trimethylsilyl)ethynyl)benzoate, 23e. To a mixture of compound 23c (0.642 mmol, 200 mg), Cul (0.064 mmol, 12.2 mg) and Pd(PPli3)2Cl2 (0.064 mmol, 45mg) in THF (2 mL) was added ethynyltrimethylsilane (0.963 mmol, 95 mg) followed by Ets (7.19 mmol, 1 mL) under N2. The reaction mixture was stirred at room temperature for 1.5 h and then partitioned between EtOAc and water. The organic layer was concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give compound 23e.
D. Methyl 6-chloro-lH-indole-5-carboxylate, 23f. A mixture of compound 23e (0.532 mmol, 150 mg) and Cul (0.32 mmol, 60 mg) in DMF (1.5 mL) was heated at 1 10 °C for 5 h and them cooled to room temperature. The reaction was quenched with water and extracted with EtOAc. The organic layer was concentrated and purified by flash column chromatography (silica gel, 15% EtOAc/hexanes) to give compound 23f.
Example 24
Figure imgf000064_0001
A. Methyl l-(3,4-difluorophenyl)-indole-5-carboxylate, 24c. A mixture of methyl indole-5-carboxylate 24a (2 g, 1 1.4 mmol), l-iodo-3,4-difluoro-benzene 24b (1.5 mL, 12.5 mmol), Cul (0.22 g, 1.14 mmol), trans-N, N'-dimethylcyclohexane-l,2- diamine (0.54 mL, 3.43 mmol), and K3PO4 (6.06 g, 28.5 mmol) in toluene (12 mL) was heated at 1 10 °C for 7 hours. The reaction mixture was diluted with CH2CI2 and filtered. The solution was concentrated and the residue was purified by flash column chromatography (silica gel, 20% EtOAc/heptane) to give 24c (3.0 g).
B. l-(3,4-difluorophenyl)-indole-5-carboxylic acid, 24d. A mixture of methyl l-(3,4-difluorophenyl)-indole-5-carboxylate 24c (3.0 g, 10.4 mmol) and LiOH (1.0 g, 41.8 mmol) in THF (120 mL) and H2O (60 mL) was stirred at room temperature for 5 days. Aqueous 10% HCl solution was added to the reaction mixture to adjust pH = 3 ~ 4. The resulting mixture was extracted with EtOAc (2x). The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated to give 24d (2.85 g).
Example 25
Figure imgf000065_0001
25d
A. Methyl 2-phenyl-benzooxazole-6-carboxylate, 25c. A mixture of methyl
4-amino-3 -hydroxy -benzoate 25a (0.3 g, 1.8 mmol) and benzoyl chloride 25b (0.23 mL, 2.0 mmol) in dioxane (2.5 mL) was heated at 210 °C under microwave for 15 min. The reaction mixture was diluted with CH2CI2 and washed with aq. aHC03. The organic solution was dried over Na2S04, concentrated and purified by flash column chromatography (silica gel, 20% EtOAc/heptane) to give 25c (0.39 g).
B. 2-Phenyl-benzooxazole-6-carboxylic acid, 25d. A mixture of methyl 2- phenyl-benzooxazole-6-carboxylate 25c (0.37 g, 1.46 mmol) and LiOH (0.10 g, 4.2 mmol) in THF (4 mL), MeOH (4 mL), and H2O (4 mL) was stirred at room temperature for 6 h. Aqueous IN HCl solution was added to the mixture to adjust pH to 3-4. The resulting mixture was extracted with EtOAc (2x). The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated to give 25d (0.34 g). Example 26
Figure imgf000066_0001
26a 10b 26b
Figure imgf000066_0002
26c
A. Ethyl 2-phenyl-benzothiazole-6-carboxylate, 26b. A mixture of ethyl 2- bromo-benzothiazole-6-carboxylate 26a (300 mg, 1.05 mmol), phenylboronic acid 10b (192 mg, 1.57 mmol), K2C03 (188 mg, 1.36 mmol) and Pd(dppf)Cl2.CH2Cl2 (43 mg, 0.05 mmol) in dioxane (2 mL) and H20 (0.4 ml) was heated at 120 °C for 25 min under microwave. The reaction mixture was diluted with CH2C12, washed with H20, dried over Na2S04, and concentrated. Purification by flash column chromatography (silica gel, 15% EtO Ac/heptane) gave 26b (220 mg).
B. 2-Phenyl-benzothiazole-6-carboxylic acid, 26c. Ethyl 2-phenyl- benzothiazole-6-carboxylate 26b (220 mg, 0.78 mmol) was stirred with LiOH (74 mg, 3.1 mmol) in THF (4 mL) and H20 (4 mL) for 16 h. Aqueous IN HC1 solution was added to the mixture to adjust pH to 3~4. The resulting mixture was extracted with EtO Ac (2x). The organic solution was washed with aq. NaCl, dried over Na2S04 and concentrated to give 26c (200 mg).
Biological Examples
In Vitro Methods
Example 1
MGL Enzyme Activity Assay
All rate-based assays were performed in black 384-well polypropylene PCR microplates (Abgene) in a total volume of 30 μί. Substrate 4-methylumbelliferyl butyrate (4MU-B; Sigma) and either purified mutant MGL (mut-MGLL 11-313 LI 79 S L186S) or purified wild type MGL (wt-MGLL 6H-11-313) were diluted separately into 20 niM PIPES buffer (pH = 7.0), containing 150 niM NaCl and 0.001% Tween 20. Compounds of Formula (la) and (lb) were pre-dispensed (50 nL) into the assay plate using a Cartesian Hummingbird prior to adding 4MU-B (25 μϊ^ of 1.2X solution to a final concentration of 10 μΜ) followed by enzyme (5 μϊ^ of a 6X solution to a final concentration of 5 nM) to initiate the reaction. Final compound concentrations ranged from 17 to 0.0003 μΜ. The fluorescence change due to 4MU-B cleavage was monitored with excitation and emission wavelengths of 335 and 440 nm, respectively, and a bandwidth of 10 nm (Safire2, Tecan) at 37°C for 5 mm.
The IC50 values for compounds of Formula (la) or (lb) were determined using Excel from a fit of the equation to the concentration-response plot of the fractional activity as a function of inhibitor concentration.
Biological Data Table 1
Figure imgf000067_0001
Example 2
2-AG Accumulation assay To measure the accumulation of 2-AG due to inhibition of MGL, one g rat brain was homogenized using a Polytron homogenizer (Brinkmann, PT300) in 10 mL of 20 mM HEPES buffer (pH = 7.4), containing 125 mM NaCl, 1 mM EDTA, 5 mM KC1 and 20 mM glucose. Compounds of Formula (la) or (lb) (10 μΜ) were pre-incubated with rat brain homogenate (50 mg). After a 15-min incubation time at 37°C, CaCl2 (final concentration = 10 mM) was added and then incubated for 15 min at 37°C in a total volume of 5 mL. The reactions were stopped with 6 mL organic solvent extraction solution of 2: 1 chloroform/methanol. Accumulated 2-AG in the organic phase was measured by a HPLC/MS method, according to the following equation: percent vehicle = (2-AG accumulation in the presence of compound/2-AG accumulation in vehicle) x 100.
Biological Data Table 2
Figure imgf000068_0001
Example 3
MGL ThermoFluor® Assay - mutant
The ThermoFluor (TF) assay is a 384-well plate-based binding assay that measures thermal stability of proteins1'2. The experiments were carried out using instruments available from Johnson & Johnson Pharmaceutical Research &
Development, LLC. TF dye used in all experiments was 1,8-ANS (Invitrogen: A-47). Final TF assay conditions used for MGL studies were 0.07 mg/ml of mutant MGL, 100 μΜ ANS, 200 mM NaCl, 0.001% Tween-20 in 50 mM PIPES (pH = 7.0).
Screening compound plates contained 100% DMSO compound solutions at a single concentration. For follow-up concentration-response studies, compounds were arranged in a pre-dispensed plate (Greiner Bio-one: 781280), wherein compounds were serially diluted in 100% DMSO across 11 columns within a series. Columns 12 and 24 were used as DMSO reference and contained no compound. For both single and multiple compound concentration-repsonse experiments, the compound aliquots (46 nL) were robotically predispensed directly into 384-well black assay plates (Abgene: TF-0384/k) using the Hummingbird liquid handler. Following compound dispension, protein and dye solutions were added to achieve the final assay volume of 3 μ The assay solutions were overlayed with 1 μΐ, of silicone oil (Fluka, type DC 200: 8541 1) to prevent evaporation.
Bar-coded assay plates were robotically loaded onto a thermostatically controlled PCR-type thermal block and then heated from 40 to 90 °C degrees at a ramp- rate of 1 °C/min for all experiments. Fluorescence was measured by continuous illumination with UV light (Hamamatsu LC6), supplied via fiber optics and filtered through a band-pass filter (380-400 nm; > 6 OD cutoff). Fluorescence emission of the entire 384-well plate was detected by measuring light intensity using a CCD camera (Sensys, Roper Scientific) filtered to detect 500 ± 25 nm, resulting in simultaneous and independent readings of all 384 wells. A single image with 20-sec exposure time was collected at each temperature, and the sum of the pixel intensity in a given area of the assay plate was recorded vs temperature and fit to standard equations to yield the Tm l.
Pantoliano, M. W., Petrella, E. C, Kwasnoski, J. D., Lobanov, V. S., Myslik, J., Graf, E., Carver, T., Asel, E., Springer, B. A., Lane, P., and Salemme, F. R. (2001) J Biomol Screen 6, 429-40.
Matulis, D., Kranz, J. K., Salemme, F. R., and Todd, M. J. (2005) Biochemistry 44, 5258-66.
The Ka values for compounds of Formula (la) and (lb) were determined from a fit of the equation to the concentration-response plot of the fractional activity as a function of Tm. For some experiments, quantitative NMR spectroscopy (qNMR) was used to measure concentration of the initial 100% DMSO compound solutions and, using the same fitting method, qKa values were determined. Biological DataTable 3
Figure imgf000070_0001
In Vivo Methods
Example 4
CFA-Induced Paw Radiant Heat Hypersensitivity
Each rat may be placed in a test chamber on a warm glass surface and allowed to acclimate for approximately 10 min. A radiant thermal stimulus (beam of light) may be focused through the glass onto the plantar surface of each hind paw in turn. The thermal stimulus may be automatically shut off by a photoelectric relay when the paw is moved or when the cut-off time is reached (20 sec for radiant heat at ~5 amps). An initial (baseline) response latency to the thermal stimulus may be recorded for each animal prior to the injection of complete Freund's adjuvant (CFA). Twenty-four hours following intraplantar CFA injection, the response latency of the animal to the thermal stimulus may be re-evaluated and compared to the animal's baseline response time. Only rats that exhibit at least a 25% reduction in response latency (i.e., are
hyperalgesic) are included in further analysis. Immediately following the post-CFA latency assessment, the indicated test compound or vehicle may be administered orally. Post-compound treatment withdrawal latencies may be assessed at fixed time intervals, typically 30, 60, 120, 180, and 300 min.
The percent reversal (%R) of hypersensitivity may be calculated in one of two different ways: 1) using group mean values or 2) using individual animal values. More specifically:
Method 1. For all compounds, the %R of hypersensitivity may be calculated using the mean value for groups of animals at each time point according to the following formula:
% reversal = [(group treatment response - group CFA response)/(group baseline response - group CFA response)] x 100
Results may be given for the maximum % reversal observed for each compound at any time point tested.
Method 2. For some compounds, the %R of hypersensitivity was calculated separately for each animal according to the following formula:
% reversal = [(individual treatment response - individual CFA
response)/(individual baseline response - individual CFA response)] x 100. Results are given as a mean of the maximum % reversal values calculated for each individual animal.
Example 5
CFA-Induced Paw Pressure Hypersensitivity
Prior to testing, rats may be acclimated to the handling procedure twice a day for a period of two days . The test consists of placing the left hindpaw on a
polytetrafluoroethylene platform and applying a linearly increasing mechanical force (constant rate of 12.5 mmHg/s) in between the third and fourth metatarsal of the dorsum of the rat's hindpaw, with a dome-tipped plinth (0.7 mm in radius), using an analgesy-meter (Stoelting, Chicago, IL), also known as a Randall-Selitto apparatus. The endpoint may be automatically reached upon hindpaw withdrawal, and the terminal force may be noted (in grams). An initial (baseline) response threshold to the mechanical stimulus may be recorded for each animal prior to the injection of complete Freund's adjuvant (CFA). Forty hours following intraplantar CFA injection, the response threshold of the animal to the mechanical stimulus may be re-evaluated and compared to the animal's baseline response threshold. A response may be defined as a withdrawal of the hindpaw, a struggling to remove the hindpaw or vocalization. Only rats that exhibit at least a 25% reduction in response threshold (i.e., hyperalgesia) may be included in further analysis. Immediately following the post-CFA threshold assessment, rats may be administered the indicated test compound or vehicle. Post- treatment withdrawal thresholds may be assessed at 1 h. Paw withdrawal thresholds may be converted to percent reversal of hypersensitivity according to the following formula:
% reversal = [(post treatment response-predose response)/(baseline response- predose response)] x 100. Example 6
Chronic constriction injury (CCI)-induced model of neuropathic pain - cold acetone-hypersensitivity test
Male Sprague-Dawley rats (225-450 g) may be used to evaluate the ability of selected compounds to reverse CCI-induced cold hypersensitivity. Four loose ligatures of 4-0 chromic gut may be surgically placed around the left sciatic nerve under inhalation anesthesia as described by Bennett et al. (Bennett GJ, Xie YK. Pain 1988, 33(1): 87-107). Fourteen to 35 days following CCI surgery, subjects may be placed in elevated observation chambers containing wire mesh floors, and five applications of acetone (0.05 mL/application separated by approximately 5 minutes) may be spritzed onto the plantar surface of the paw using a multidose syringe. An abrupt withdrawal or lifting of the paw may be considered a positive response. The number of positive responses may be recorded for each rat over the five trials. Following baseline withdrawal determinations, compounds may be administered in the indicated vehicle, by the indicated route (see Table 6). The number of withdrawals may be re-determined 1 to 4 hr after compound administration. Results may be presented as a percent inhibition of shakes, which may be calculated for each subject as [l-(test compound withdrawals / pre-test withdrawals)] x 100 and then averaged by treatment. Example 7
Spinal nerve ligation (SNL) model of neuropathic pain - tactile allodynia test For lumbar 5 (L5) spinal nerve ligation (SNL) studies, anesthesia may be induced and maintained on isoflurane inhalation. Fur may be clipped over the dorsal pelvic area, and a 2-cm skin incision may be made just left of midline over the dorsal aspect of the L4-S2 spinal segments, followed by separation of the paraspinal muscles from spinous processes. The transverse process of L6 may be carefully removed, and the L5 spinal nerve may be identified. The left L5 spinal nerve may be ligated tightly with 6-0 silk thread, the muscle may be sutured with 4-0 vicryl, and the skin may be closed with wound clips. Following surgery, s.c. saline (5 mL) may be administered.
Behavioral testing may be performed four weeks post-ligation. Following baseline von Frey determinations to verify the presence of mechanical allodynia, L5 SNL rats may be orally administered the indicated vehicle or drug. Tactile allodynia may be quantified at 30 , 60, 100, 180 and 300 min post-dosing by recording the force at which the paw ipsilateral to the nerve ligation is withdrawn from the application of a series of calibrated von Frey filaments (0.4, 0.6, 1.0, 2.0, 4, 6, 8 and 15 g; Stoelting; Wood Dale, IL). Beginning at an intermediate stiffness (2.0 g), filaments may be applied to the mid-plantar hind paw for approximately 5 seconds to determine the response threshold, a brisk paw withdrawal leads to the presentation of the next lighter stimulus, whereas a lack of a withdrawal response leads to the presentation of the next stronger stimulus. A total of four responses after the first threshold detection may be collected. The 50% withdrawal thresholds may be interpolated by the method of Dixon as modified by Chaplan et.al, and when response thresholds fall above or below the range of detection, respective values of 15.0 or 0.25 g may be assigned. Threshold data from von Frey filament testing may be reported as withdrawal threshold in grams. Data may be normalized and results may be presented as % MPE (maximum possible effect) of the drug calculated according to the following formula:
% MPE = x g/force - baseline g/force X 100
15 g/force - baseline g/force While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, it will be understood that the practice of the invention encompasses all of the usual variations, adaptations and/or modifications as come within the scope of the following claims and their equivalents.

Claims

compound of Formula la)
Figure imgf000075_0001
Formula (la)
wherein
Y is
i) a C6-io aryl or
ii) a heteroaryl selected from the group consisting of thiazolyl, oxazolyl, pyridinyl, and pyrimidinyl,
wherein Y is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci-4alkoxy, cyano, and trifluoromethyl;
Z is selected from the group consisting of
i) a C6-io aryl,
ii) a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, indazolyl, and indolyl, or iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, Ci-4alkoxy, and trifluoromethyl; wherein the C6-10 aryl and the heteroaryl of Z are unsubstituted or
substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, Ci-4alkoxy, trifluoromethyl, and phenyl, provided that no more than one substituent of Z is phenyl and said phenyl is unsubsituted or substituted with one or two subsituents each of which is independently selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro;
n is 1 or 2; and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
2. The compound of claim 1 wherein Y is phenyl, thiazolyl, or pyrimidinyl.
3. The compound of claim 3 wherein Y is thiazolyl or pyrimidinyl.
4. The compound of claim 1 wherein
Z is
Figure imgf000076_0001
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, C1-4alkyl, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro.
5. The compound of claim 4 wherein Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro.
6. The compound of claim 1 wherein n is 1. The compound of claim 1 wherein n is 2.
A compound of Formula (la)
Figure imgf000077_0001
Formula (la)
wherein:
Y is
i) phenyl or
ϋ) a heteroaryl that is thiazolyl or pyrimidinyl;
Z is
Figure imgf000077_0002
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzothienyl, and indolyl,
wherein the Ce-w aryl and the heteroaryl of Z are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, C1-4alkyl, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro;
n is 1 or 2; and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
The compound of claim 8 wherein n is 1.
The compound of claim 8 wherein n is 2.
A compound of Formula (la)
Figure imgf000077_0003
Formula (la)
wherein:
Y is thiazolyl or pyrimidinyl;
Z is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Z is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, trifluoromethyl, and phenyl,
provided that no more than one substituent of Z is phenyl, and said phenyl is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or fluoro; n is 1 or 2;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
12. The compound of claim 1 1 wherein n is 1.
13. A compound of Formula (la)
Figure imgf000078_0001
Formula (la)
selected from the group consisting of
the compound wherein Y is phenyl, Z is 4-(phenylmethyl)-phenyl, and n is 1 ; the compound wherein Y is phenyl, Z is 4-phenyl-phenyl, and n is 1;
the compound wherein Y is pyrimidin-2-yl, Z is 1 -(4-trifluoromethyl-phenyl)- lH-indol-5-yl, and n is 1;
thecompound wherein Y is pyrimidin-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5- yl, and n is 1 ;
thecompound wherein Y is thiazol-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 1; thecompound wherein Y is thiazol-4-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 1;
thecompound wherein Y is thiazol-4-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and n is 1 ;
thecompound wherein Y is pyrimidin-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and n is 1 ;
thecompound wherein Y is pyrimidin-2-yl, Z is 4-phenyl-phenyl, and n is 1 ; thecompound wherein Y is pyrimidin-2-yl, Z is 2-phenyl-benzothiazol-6-yl, and n is 1;
thecompound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and n is 1 ;
thecompound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzothiazol-6-yl, and n is 1;
thecompound wherein Y is phenyl, Z is l-(4-trifluoromethyl-phenyl)-lH-indol- 5-yl, and n is 1;
thecompound wherein Y is phenyl, Z is 3-chloro-6-trifluoromethyl-benzothien- 2-yl, and n is 1;
thecompound wherein Y is phenyl, Z is l-(3,4-difluoro-phenyl)-lH-indol-5-yl, and n is 1;
thecompound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6-yl, and n is i ;
thecompound wherein Y is pyrimidin-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and n is 2;
thecompound wherein Y is thiazol-2-yl, Z is 4-phenyl-phenyl, and n is 1 ;
thecompound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and n is 2;
thecompound wherein Y is pyrimidin-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5- yl, and n is 2;
thecompound wherein Y is thiazol-2-yl, Z is l-(4-fluoro-phenyl)-lH-indol-5-yl, and n is 2;
thecompound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6-yl, and n is 2; and pharmaceutically acceptable salt forms thereof. A compound of Formula lb)
Figure imgf000080_0001
Formula (lb)
wherein:
Yb is
Figure imgf000080_0002
ii) a heteroaryl selected from the group consisting of thiazolyl, oxazolyl, pyridinyl, and pyrimidinyl,
wherein Yb is unsubstituted or substituted with one or two substituents each of which is selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci-4alkoxy, cyano, and trifluoromethyl;
Zb is
i) a C6-io aryl,
ii) a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl, or
iii) phenylmethyl-phenyl; wherein the phenyl of phenylmethyl is unsubstituted or substituted with one or two substituents each of which is selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, Ci_ 4alkoxy, and trifluoromethyl,
wherein the Ce-w aryl and the heteroaryl of Zb are unsubstituted or substituted with one or two substitutents each of which is selected from the group consisting of bromo, chloro, fluoro, iodo, Ci_4alkyl, Ci_4alkoxy, trifluoromethyl, and phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is selected from the group consisting of trifluoromethyl, chloro, cyano, and fluoro; and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
15. The compound of claim 14 wherein Yb is thiazolyl or pyrimidinyl.
16. The compound of claim 14 wherein Zb is the heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl;
wherein the heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl;
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro.
17. The compound of claim 16 wherein Zb is benzothienyl or indolyl,
wherein Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one trifluoromethyl or fluoro substituent.
18. A compound of Formula lb)
Figure imgf000081_0001
Formula (lb)
wherein:
Yb is thiazolyl or pyrimidinyl;
Zb is a heteroaryl selected from the group consisting of benzoxazolyl, benzothiazolyl, benzothienyl, and indolyl,
wherein the heteroaryl of Zb is unsubstituted or substituted with one or two substitutents each of which is selected from the group consisting of bromo, chloro, fluoro, trifluoromethyl, and phenyl,
provided that no more than one substituent is phenyl and said phenyl is unsubstituted or substituted with one or two substituents each of which is trifluoromethyl or fluoro; and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof. A compound of Formula lb)
Figure imgf000082_0001
Formula (lb)
wherein:
Yb is thiazolyl or pyrimidinyl;
Zb is benzothienyl or indolyl,
wherein Zb is unsubstituted or substituted with one or two substitutents each of which is trifluoromethyl or phenyl,
provided that no more than one substituent of Zb is phenyl and said phenyl is unsubstituted or substituted with one trifluoromethyl or fluoro substituent;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof. A compound of Formula lb)
Figure imgf000082_0002
Formula (lb)
selected from the group consisting of:
the compound wherein Yb is pyrimidin-2-yl, and Zb is 6-trifluoromethyl- benzothien-2-yl;
the compound wherein Yb is pyrimidin-2-yl, and Zb is 1 -(4-trifluoromethyl- phenyl)- lH-indol-5-yl;
and pharmaceutically acceptable salts thereof.
A pharmaceutical composition comprising the compound of claim 1 or 13 and member selected from the group consisting of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and a pharmaceutically acceptable diluent.
22. A pharmaceutical composition of claim 21, wherein the composition is a solid oral dosage form.
23. A pharmaceutical composition of claim 21, wherein the composition is selected from the group consisting of a syrup, an elixir, and a suspension.
24. A method for treating inflammatory pain in a subject in need thereof comprising administering a therapeutically effective amount of the compound of claim 1 or 13 to the subject.
25. The method of claim 24 wherein the inflammatory pain is due to inflammatory bowel disease, visceral pain, migraine, post operative pain, osteoarthritis, rheumatoid arthritis, back pain, lower back pain, joint pain, abdominal pain, chest pain, labor, musculoskeletal diseases, skin diseases, toothache, pyresis, burn, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection, rhinitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome, cholecystitis, pancreatitis, postmastectomy pain syndrome, menstrual pain, endometriosis, pain, pain due to physical trauma, headache, sinus headache, tension headache, or arachnoiditis.
A pharmaceutical composition comprising the compound of claim 14 or 20 and a member selected from the group consisting of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and a pharmaceutically acceptable diluent.
27. A pharmaceutical composition of claim 26, wherein the composition is a solid oral dosage form.
28. A pharmaceutical composition of claim 26, wherein the composition is selected from the group consisting of a syrup, an elixir, and a suspension.
29. A method for treating inflammatory pain in a subject in need thereof comprising administering a therapeutically effective amount of the compound of claim 14 or 20 to the subject.
30. The method of claim 29 wherein the inflammatory pain is due to inflammatory bowel disease, visceral pain, migraine, post operative pain, osteoarthritis, rheumatoid arthritis, back pain, lower back pain, joint pain, abdominal pain, chest pain, labor, musculoskeletal diseases, skin diseases, toothache, pyresis, burn, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection, rhinitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome, cholecystitis, pancreatitis, postmastectomy pain syndrome, menstrual pain, endometriosis, pain, pain due to physical trauma, headache, sinus headache, tension headache, or arachnoiditis.
PCT/US2011/053442 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors WO2012044613A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
BR112013007287A BR112013007287A2 (en) 2010-09-27 2011-09-27 oxopiperazine azetidine amides and oxodiazepine azetine amides as monoacylglycerol lipase inhibitors
CA2810077A CA2810077A1 (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
KR1020137010278A KR20140001206A (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
EP11771300.8A EP2621918A1 (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
RU2013114852/04A RU2013114852A (en) 2010-09-27 2011-09-27 OXOPIPERAZINE-AZETIN AMIDES AND OXODIAZEPIN-AZETIDINE AMIDES AS LIPASE MONACYLGLYCEROL INHIBITORS
JP2013530418A JP2013537920A (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidineamide and oxodiazepine-azetidineamide as monoacylglycerol lipase inhibitors
AU2011307252A AU2011307252B2 (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
CN2011800423053A CN103080103A (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38677710P 2010-09-27 2010-09-27
US61/386,777 2010-09-27

Publications (1)

Publication Number Publication Date
WO2012044613A1 true WO2012044613A1 (en) 2012-04-05

Family

ID=44906361

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/053442 WO2012044613A1 (en) 2010-09-27 2011-09-27 Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors

Country Status (11)

Country Link
US (1) US8637498B2 (en)
EP (1) EP2621918A1 (en)
JP (1) JP2013537920A (en)
KR (1) KR20140001206A (en)
CN (1) CN103080103A (en)
AR (1) AR083180A1 (en)
AU (1) AU2011307252B2 (en)
BR (1) BR112013007287A2 (en)
CA (1) CA2810077A1 (en)
RU (1) RU2013114852A (en)
WO (1) WO2012044613A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015099196A1 (en) 2013-12-26 2015-07-02 Takeda Pharmaceutical Company Limited 4-(piperrazin-1-yl)-pyrrolidin-2-one compounds as monoacylglycerol lipase (magl) inhibitors
WO2017171100A1 (en) 2016-03-31 2017-10-05 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2017170830A1 (en) 2016-03-31 2017-10-05 武田薬品工業株式会社 Heterocyclic compound
US10106556B2 (en) 2015-03-30 2018-10-23 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2019065791A1 (en) 2017-09-29 2019-04-04 武田薬品工業株式会社 Heterocyclic compound

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ726874A (en) * 2014-05-19 2018-03-23 Merial Inc Anthelmintic compounds
EP3197450A1 (en) 2014-09-22 2017-08-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of fibrosis
BR112018000041A2 (en) 2015-07-31 2018-09-04 Pfizer Inc. 1,1,1-TRIFLUORO-3-HYDROXYPROPAN-2-ILA CARBAMATE DERIVATIVES AND 1,1,1-TRIFLUORO-4-HYDROXYBUTAN-2-ILA CARBAMATE DERIVATIVES AS MAGL INHIBITORS
EP3571191A1 (en) 2017-01-20 2019-11-27 Pfizer Inc 1,1,1-trifluoro-3-hydroxypropan-2-yl carbamate derivatives as magl inhibitors
EP3571202B1 (en) 2017-01-23 2021-06-30 Pfizer Inc. Heterocyclic spiro compounds as magl inhibitors
CN110577515B (en) * 2019-09-02 2022-05-17 南通大学 Synthesis method of 4- (azetidin-3-yl) -1-methylpiperazine-2-one dihydrochloride
CN115124447A (en) * 2022-08-29 2022-09-30 山东诚创蓝海医药科技有限公司 Preparation method of 3-azetidinone hydrochloride
CN115417804A (en) * 2022-09-16 2022-12-02 天津药明康德新药开发有限公司 Synthesis method of 7-methyl-1H-indole-5-carboxylic acid

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056800A1 (en) * 2002-12-23 2004-07-08 Janssen Pharmaceutica N.V. Substituted 1-piperidin-4-yl-4-azetidin-3-yl-piperazine derivatives and their use as neurokinin antagonists
US20090269785A1 (en) * 2008-04-25 2009-10-29 Carsten Schubert Crystal structure of monoacylglycerol lipase (mgll)
WO2010124121A1 (en) * 2009-04-22 2010-10-28 Jansen Pharmaceutica Nv Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitor

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6124299A (en) 1997-02-24 2000-09-26 Zymogenetics, Inc. Calcitonin mimetics
AU1089599A (en) 1997-10-15 1999-05-03 Coelacanth Chemical Corporation Synthesis of azetidine derivatives
AU3565999A (en) 1999-04-16 2000-11-02 Coelacanth Chemical Corporation Synthesis of azetidine derivatives
AR033517A1 (en) 2000-04-08 2003-12-26 Astrazeneca Ab PIPERIDINE DERIVATIVES, PROCESS FOR THE PREPARATION AND USE OF THESE DERIVATIVES IN THE MANUFACTURE OF MEDICINES
US6995144B2 (en) 2002-03-14 2006-02-07 Eisai Co., Ltd. Nitrogen containing heterocyclic compounds and medicines containing the same
EP1522314B1 (en) 2002-06-26 2014-03-05 Ono Pharmaceutical Co., Ltd. Remedies for diseases caused by vascular contraction or dilation
EP1702916A1 (en) 2005-03-18 2006-09-20 Santhera Pharmaceuticals (Schweiz) GmbH DPP-IV inhibitors
PE20071136A1 (en) 2005-12-21 2007-12-29 Schering Corp DERIVATIVES OF ANILINE SUBSTITUTED AS ANTAGONISTS OF HISTAMINE H3
FR2915199B1 (en) 2007-04-18 2010-01-22 Sanofi Aventis TRIAZOLOPYRIDINE-CARBOXAMIDE AND TRIAZOLOPYRIMIDINE-CARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE.
FR2915197B1 (en) 2007-04-18 2009-06-12 Sanofi Aventis Sa TRIAZOLOPYRIDINE CARBOXAMIDE DERIVATIVES, PREPARATION THEREOF AND THERAPEUTIC USE THEREOF.
FR2915198B1 (en) 2007-04-18 2009-12-18 Sanofi Aventis TRIAZOLOPYRIDINE CARBOXAMIDE AND TRIAZOLOPYRIDINE -CARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE.
WO2009117444A1 (en) 2008-03-17 2009-09-24 Northeastern University Inhibitors of fatty acid amide hydrolase and monoacylglycerol lipase for modulation of cannabinoid receptors
JP5767579B2 (en) * 2008-04-25 2015-08-19 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプJanssen Pharmaceutica Naamloze Vennootschap Protein modification of monoacylglycerol lipase (MGLL)
US8435977B2 (en) * 2009-04-22 2013-05-07 Janssen Pharmaceutica, Nv Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitors
RU2011147200A (en) * 2009-04-22 2013-05-27 Янссен Фармацевтика Нв ASETIDINYLDIAMIDES AS MONOACYGLYCERINE LIPASE INHIBITORS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056800A1 (en) * 2002-12-23 2004-07-08 Janssen Pharmaceutica N.V. Substituted 1-piperidin-4-yl-4-azetidin-3-yl-piperazine derivatives and their use as neurokinin antagonists
US20090269785A1 (en) * 2008-04-25 2009-10-29 Carsten Schubert Crystal structure of monoacylglycerol lipase (mgll)
WO2010124121A1 (en) * 2009-04-22 2010-10-28 Jansen Pharmaceutica Nv Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitor

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"Design of Prodrugs", 1985, ELSEVIER
BENITO ET AL., BRIT JPHARMACOL, vol. 153, 2008, pages 277 - 285
BENNETT GJ, XIE YK, PAIN, vol. 33, no. 1, 1988, pages 87 - 107
BEN-SHABAT ET AL., EUR J PHARMACOL, vol. 353, 1998, pages 23 - 31
CAVUOTO ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 364, 2007, pages 105 - 110
COMELLI ET AL., BRIT JPHARMACOL, vol. 152, 2007, pages 787 - 794
CRAVATT ET AL., J NEUROBIOL, vol. 61, 2004, pages 149 - 60
DEVANE ET AL., SCIENCE, vol. 258, 1992, pages 1946 - 9
DI MARZO ET AL., CURR OPIN LIPIDOL, vol. 18, 2007, pages 129 - 140
DI MARZO ET AL., CURR PHARM DES, vol. 6, 2000, pages 1361 - 80
DOGRUL ET AL., PAIN, vol. 100, 2002, pages 203 - 9
GUINDON ET AL., BRIT JPHARMACOL, vol. 153, 2008, pages 319 - 334
HAJRASOULIHA ET AL., EUR JPHARMACOL, vol. 579, 2008, pages 246 - 252
J.F.W. MCOMIE: "Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
JHAVERI ET AL., BRIT J PHARMACOL, vol. 152, 2007, pages 624 - 632
KATHURIA ET AL., NAT MED, vol. 9, 2003, pages 76 - 81
LICHTMAN ET AL., JPHARMACOL EXP THER, vol. 302, 2002, pages 73 - 9
LICHTMAN ET AL., PAIN, vol. 109, 2004, pages 319 - 27
MATSUDA ET AL., NATURE, vol. 346, 1990, pages 561 - 4
MATULIS, D., KRANZ, J. K., SALEMME, F. R., TODD, M. J., BIOCHEMISTRY, vol. 44, 2005, pages 5258 - 66
MCCARBERG BILL ET AL., AMER J THER, vol. 14, 2007, pages 475 - 83
MECHOULAM ET AL., BIOCHEM PHARMACOL, vol. 50, 1995, pages 83 - 90
MECHOULAM, BIOCHEM PHARMACOL, vol. 50, 1995, pages 83 - 90
MUNRO ET AL., NATURE, vol. 365, 1993, pages 61 - 5
NJIE, YA FATOU, HE, FANG, QIAO, ZHUANHONG, SONG, ZHAO-HUI, EXP. EYE RES., vol. 87, no. 2, 2008, pages 106 - 14
PACHER ET AL., AMER JPHYSIOL, vol. 294, 2008, pages H1133 - H1134
PANTOLIANO, M. W., PETRELLA, E. C., KWASNOSKI, J. D., LOBANOV, V. S., MYSLIK, J., GRAF, E., CARVER, T., ASEL, E., SPRINGER, B. A.,, JBIOMOL SCREEN, vol. 6, 2001, pages 429 - 40
PERTWEE, BRIT JPHARMACOL, vol. 153, 2008, pages 199 - 215
PIOMELLI, NAT REV NEUROSCI, vol. 4, 2003, pages 873 - 884
SUGIURA, BIOCHEM BIOPHYS RES COMMUN, vol. 215, 1995, pages 89 - 97
T.W. GREENE, P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
T.W. GREENE, P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
WALKER ET AL., PROC NATL ACAD SCI USA, vol. 96, 1999, pages 12198 - 203

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015099196A1 (en) 2013-12-26 2015-07-02 Takeda Pharmaceutical Company Limited 4-(piperrazin-1-yl)-pyrrolidin-2-one compounds as monoacylglycerol lipase (magl) inhibitors
JP2017502957A (en) * 2013-12-26 2017-01-26 武田薬品工業株式会社 4- (Piperazin-1-yl) -pyrrolidin-2-one compounds as monoacylglycerol lipase (MAGL) inhibitors
US9624170B2 (en) 2013-12-26 2017-04-18 Takeda Pharmaceutical Company Limited 4-(piperrazin-1-yl)-pyrrolidin-2-one compounds as monoacylglycerol lipase (MAGL) inhibitors
US10106556B2 (en) 2015-03-30 2018-10-23 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2017171100A1 (en) 2016-03-31 2017-10-05 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2017170830A1 (en) 2016-03-31 2017-10-05 武田薬品工業株式会社 Heterocyclic compound
US10323026B2 (en) 2016-03-31 2019-06-18 Takeda Pharmaceutical Company Limited Heterocyclic compound
US10610520B2 (en) 2016-03-31 2020-04-07 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2019065791A1 (en) 2017-09-29 2019-04-04 武田薬品工業株式会社 Heterocyclic compound
US11274101B2 (en) 2017-09-29 2022-03-15 Takeda Pharmaceutical Company Limited Heterocyclic compound

Also Published As

Publication number Publication date
AR083180A1 (en) 2013-02-06
CA2810077A1 (en) 2012-04-05
US8637498B2 (en) 2014-01-28
CN103080103A (en) 2013-05-01
BR112013007287A2 (en) 2016-06-14
AU2011307252A1 (en) 2013-03-14
RU2013114852A (en) 2014-11-10
AU2011307252B2 (en) 2015-06-25
EP2621918A1 (en) 2013-08-07
US20120077797A1 (en) 2012-03-29
JP2013537920A (en) 2013-10-07
KR20140001206A (en) 2014-01-06

Similar Documents

Publication Publication Date Title
EP2611774B1 (en) Di-azetidinyl diamide as monoacylglycerol lipase inhibitors
AU2011307252B2 (en) Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
US8748417B2 (en) Amino-pyrrolidine-azetidine diamides as monoacylglycerol lipase inhibitors
JP2013540159A (en) Piperidin-4-yl-azetidine diamide as monoacylglycerol lipase inhibitor
AU2010238746B2 (en) Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitors
KR20120034616A (en) Azetidinyl diamides as monoacylglycerol lipase inhibitors
US8513423B2 (en) Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors
WO2010124121A1 (en) Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitor
TW201313702A (en) Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors

Legal Events

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

Ref document number: 201180042305.3

Country of ref document: CN

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

Ref document number: 11771300

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2810077

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013530418

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2011307252

Country of ref document: AU

Date of ref document: 20110927

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011771300

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20137010278

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013114852

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013007287

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013007287

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130327