EP2629851A1 - Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors - Google Patents

Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors

Info

Publication number
EP2629851A1
EP2629851A1 EP11784828.3A EP11784828A EP2629851A1 EP 2629851 A1 EP2629851 A1 EP 2629851A1 EP 11784828 A EP11784828 A EP 11784828A EP 2629851 A1 EP2629851 A1 EP 2629851A1
Authority
EP
European Patent Office
Prior art keywords
compound
phenyl
thiazol
group
trifluoromethyl
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP11784828.3A
Other languages
German (de)
French (fr)
Inventor
Peter J. Connolly
Haiyan Bian
Xun Li
Li Liu
Mark J. Macielag
Mark E. Mcdonnell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceutica NV
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
Publication of EP2629851A1 publication Critical patent/EP2629851A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic 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
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P1/18Drugs 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs 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
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • Cannabis sativa has been used for the treatment of pain for many years.
  • ⁇ 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 Lipidol, 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 1 /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 (I)
  • Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
  • C6-10 aryl is unsubstituted or substituted with a substituent selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci-4alkoxy, cyano, and trifluoromethyl; or
  • an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
  • Group b) other than phenylmethyl-phenyl is 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_ 4alkoxy, and 3 ⁇ 4; provided that no more than one substituent is 3 ⁇ 4; and
  • R b is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of 3 ⁇ 4 are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of trifluoromethyl, methyl, chloro, cyano, and fluoro; R is hydrogen or hydroxy; 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 (I) or a pharmaceutically acceptable salt form thereof.
  • a pharmaceutical composition comprising, consisting of, and/or consisting essentially of admixing a compound of Formula (I) 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 (I) or a pharmaceutically acceptable salt form thereof.
  • 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.
  • 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.
  • substituent groups with multiple alkyl groups such as, (Ci_ 6 alkyl) 2 amino-, the Ci- 6 alkyl 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 to 8 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.
  • 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 the group consisting of N, O, and S.
  • heteroaryl aromatic rings of 5 or 6 members wherein the ring consists of carbon atoms and has at least one heteroatom member. Suitable heteroatoms include nitrogen, oxygen, and sulfur. In the case of 5 membered rings, the heteroaryl ring preferably contains one member of nitrogen, oxygen or sulfur and, in addition, up to 3 additional nitrogens. 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 nitrogens, at most 2 nitrogen atoms are adjacent.
  • heteroaryl groups include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, isoindolyl, benzofuryl, benzothienyl, indazolyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzis oxazolyl,
  • 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
  • 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_
  • 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) includes 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 (I) 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 (I), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
  • the compounds of Formula (I), 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 (I), 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 (I), or an enantiomer, diastereomer, solvate or
  • 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.
  • inflammatory hyperalgesia is inflammatory hyperalges
  • 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 and/or dermal allergy itch, eczema, pharyngitis, enteritis, irritable bowel syndrome, inflammatory bowel diseases including Crohn's Disease, ulcerative colitis, benign prostatic hypertrophy, and nasal hypersensitivity.
  • 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 (I).
  • 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 (I) 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 (I) 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 (I)
  • Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
  • Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
  • Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, 1H- pyrrol-2-yl, and lH-pyrrol-3-yl;
  • Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, methyl, and 3 ⁇ 4,; provided that no more than one substituent is 3 ⁇ 4; and
  • R b is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of 3 ⁇ 4 are unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro; Group b) is
  • benzimidazolyl benzothienyl, and indolyl; iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; or
  • Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and 3 ⁇ 4; provided that no more than one substituent is 3 ⁇ 4; and
  • R b is selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of 3 ⁇ 4 are optionally independently substituted with one to two trifluoromethyl, methyl, chloro, or fluoro substituents; e) R is hydrogen; and any combination of embodiments a) through e) above, provided that it is understood that combinations in which different embodiments of the same substituent would be combined are excluded;
  • An embodiment of the present invention includes a compound of Formula (I)
  • Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
  • Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
  • Group b) is
  • phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; or iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
  • Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and 3 ⁇ 4; provided that no more than one substituent is 3 ⁇ 4; and
  • Rbis selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of 3 ⁇ 4 are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
  • R is hydrogen
  • Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
  • Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
  • Group b) is
  • Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and 3 ⁇ 4; provided that no more than one substituent is and
  • R b is selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of 3 ⁇ 4 are unsubstituted or substituted with one or two substitutents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
  • R is hydrogen or hydroxy
  • Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
  • Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, lH-pyrrol- 2-yl, and lH-pyrrol-3-yl;
  • Group b) is
  • Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and 3 ⁇ 4; provided that no more than one substituent is 3 ⁇ 4; and
  • Rbis selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of R b are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
  • R is hydrogen or hydroxy
  • An embodiment of the present invention includes a compound of Formula (I)
  • salts of compounds of Formula (I) refer to non-toxic
  • Suitable pharmaceutically acceptable salts of compounds of Formula (I) 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.
  • 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.
  • 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.
  • 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
  • Embodiments of the present invention include prodrugs of compounds of Formula (I).
  • 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 (I).
  • 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 (I) 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
  • compositions including a pharmaceutical composition, comprising, consisting of, and consisting essentially of the (-)-enantiomer of a compound of Formula (I) 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
  • compositions comprising compounds of Formula (I) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and/or pharmaceutically acceptable diluent.
  • the compounds of Formula (I) 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 (I) 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.
  • 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 (I) 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, benzyl, sulfonyl, sulfonyl, adiluent, 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.).
  • 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 preservatives.
  • a therapeutically effective amount of a compound of Formula (I) 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 (I) 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 (I).
  • a compound of Formula (I) 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 (I) 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 (I) 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 (I) is required for a subject in need thereof.
  • the compounds of Formula (I) 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 (I).
  • Scheme A illustrates a route for the synthesis of intermediates that are useful for the preparation of compounds of Formula (I) wherein R is hydrogen.
  • a compound of formula Al (wherein P is a conventional amino protecting group) is either commercially available or may be prepared by known methods described in the scientific literature.
  • a compound of formula Al may be treated with zinc metal in the presence of TMS-C1, in an aprotic solvent, followed by addition of a compound of formula A2, in the presence of palladium catalyst to afford a compound of formula A3.
  • Treatment with benzyl bromide affords the pyridinium bromide of formula A4.
  • a compound of formula A4 may be reduced to a compound of formula A5 in the presence of a hydride source such as, sodium borohydride, in an organic alcoholic solvent such as, ethanol. Removal of the benzyl group and reduction of the double bond may be achieved by palladium catalyzed hydrogenation to afford the desired intermediate of formula A6.
  • Scheme B illustrates a route for the preparation of compounds of Formula (I)-B wherein Y and Z are as defined herein and R of Formula (I) is hydrogen.
  • the amino protecting group (P) of a compound of formula A3 may be removed by conventional synthetic methods to afford a secondary amine of formula Bl.
  • the amino group may be coupled with a carboxylic acid of formula B2 (wherein Q is hydroxy) in the presence of an appropriate coupling agent such as HATU, DCC, EDC, HBTU, PyBrOP, and the like, optionally in the presence of a base such as DIPEA, to afford an amide of formula B3.
  • an acid chloride of formula B2 (wherein Q is chloro) may be used to effect the acylation of a compound of formula Bl.
  • a non- nucleophilic base such as pyridine may be added to afford an amide of formula B3.
  • Reduction of the pyridine ring of a compound of formula B3 may be achieved by palladium catalyzed hydrogenation to afford a compound of formula B4.
  • a second acylation with an appropriately Y-substituted carboxylic acid or acid chloride of formula B5 affords a compound of Formula (I)-B wherein R of Formula (I) is hydrogen.
  • Scheme C illustrates an alternate route for the preparation of compounds of Formula (I)-B wherein Y and Z are as defined herein and R of Formula (I) is hydrogen.
  • the compound of formula A6 may be acylated according to the synthetic methods described under Scheme B to afford the acylated compound of formula CI.
  • Scheme D illustrates a route for the preparation of compounds of Formula (I)-D wherein Y and Z are as defined herein and R of Formula (I) is hydroxy.
  • a compound of formula Dl may be treated with samarium iodide in the presence of HMPA, in an aprotic solvent, followed by the addition of a ketone of formula D2 to afford the condensed product of formula D3. Removal of the benzhydryl group may be effected by palladium catalyzed hydrogenation to afford the free amine of formula D4.
  • the amine of formula D4 may be acylated with a Z -substituted compound of formula B2 by the methods previously described herein to afford a compound of Formula (I)-D.
  • Scheme E illustrates an alternate route for the preparation of compounds of Formula (I)-D wherein Y and Z are as defined herein and R of Formula (I) is hydroxy.
  • a compound of formula Dl may be condensed with a compound of formula El in the presence of samarium iodide and HMPA to afford a compound of formula E2.
  • the above prepared zinc iodide reagent of compound la in DMA was added as a suspension.
  • the mixture was degassed with vacuum/N2 twice and then heated to 80 °C. (Note: The reaction was exothermic.)
  • the progress of the reaction was monitored by HPLC and LC-MS and was complete after 2 h.
  • the reaction mixture was cooled to 40 °C; EtOAc (1.6 L) was added and the mixture was stirred for 10 min.
  • the insoluble material (excess Zn and Cu
  • the separated aqueous phase was extracted with EtOAc (1 L), and the combined organic phases were treated with saturated aqueous NH 4 C1 (0.8 L x 2) and stirred for 15 min (repeated again), washed with 5% aqueous NaHS0 3 (500 mL) and brine (1 L), and dried over MgS0 4 .
  • the catalyst was removed by filtration though a diatomaceous earth pad, which was washed with MeOH (50 mL x 3). Concentration of the filtrate at 50 °C under high- vacuum ( ⁇ 10 mmHg) afforded 13.4 g (103% isolated yield, 97% pure at 210 nm, HPLC area%) of pure compound lg as a slight yellowish, thick oil, which contained a trace amount of EtOH residue by ⁇ - MR analysis.
  • Methyl thioglycolate 3b (30.3 mmol, 2.76 mL) was added dropwise to a suspension of NaH (60% oil dispersion, 75.8 mmol, 3.03 g) in 10 mL of THF and 50 mL of DMSO at 20 °C. The mixture was stirred for 15 min and a solution of l-(2-fluoro-4- (trifluoromethyl)phenyl)ethanone 3a (24.3 mmol, 5.0 g) in 10 mL of DMSO was added. The reaction mixture was stirred at 20 °C for 4 h and water was added. The mixture was extracted with EtOAc. The organic layer was dried over MgS0 4 and concentrated to give compound 3c as a white solid.
  • Lead (IV) acetate (29.9 mmol, 13.3 g) was added in one portion and the mixture was refluxed for 20 min. After cooling, the precipitate was removed by filtration and washed with acetonitrile.
  • A. Methyl l-(4-fluorophenyl)-indole-5-carboxylate, 7c A mixture of methyl indole-5-carboxylate 7a (0.5 g, 2.85 mmol), 1 -bromo-4-fluoro-benzene 7b (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 in a microwave reactor at 220 °C for 2.5 h. 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% EtO Ac/heptane) to give 0.58 g of compound 7c. MS m/z 270.1 (M+H + ).
  • Methyl l-(3,4-difluorophenyl)-indole-5-carboxylate, 8b A mixture of methyl indole-5-carboxylate 7a (2 g, 11.4 mmol), l-iodo-3,4-difluoro-benzene 8a (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 K 3 PO 4 (6.06 g, 28.5 mmol) in toluene (12 mL) was heated at 110 °C for 7 h.
  • A. Methyl 4-((4-fluorophenyl)amino)-3-nitrobenzoate, 9c A mixture of methyl 4-fluoro-3-nitrobenzoate 9a (1 g, 5.02 mmol), 4-fluoroaniline 9b (4.34 mL, 5.02 mmol), and DIPEA (1.04 mL, 6.03 mmol) in DMF (10 mL) was stirred at room temperature for 2 h. Water was added to the mixture, the resulting solid was collected by filtration, washed with water, and dried. The crude compound 9c was used in the next reaction without further purification.
  • a solution of l-benzhydryl-3-iodoazetidine 13a (1.4 mmol, 490 mg) in 5 mL of THF was added to a stirring mixture of Sml 2 (0.1 M THF solution, 3 mmol, 30 mL) and 1.7 mL of HMPA. After 5 min, a solution of 1- benzoylpiperidin-4-one 13b (3.1 mmol, 626 mg) in 5 mL of THF was added. The reaction mixture was stirred for 2 h.
  • IC5 0 values for compounds of Formula (I) 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.
  • percent vehicle (2-AG accumulation in the presence of compound/2-AG accumulation in vehicle) x 100.
  • 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 (I) 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.
  • Each rat was 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) was focused through the glass onto the plantar surface of each hind paw in turn.
  • the thermal stimulus was automatically shut off by a photoelectric relay when the paw was moved or when the cut-off time was reached (20 sec for radiant heat at ⁇ 5 amps).
  • An initial (baseline) response latency to the thermal stimulus was 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 was re-evaluated and compared to the animal's baseline response time.
  • CFA complete Freund's adjuvant
  • the percent reversal (%R) of hypersensitivity was calculated in one of two different ways: 1) using group mean values or 2) using individual animal values. More specifically:
  • 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, L 5 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 (I) as follows: Formula (I), wherein Y, Z, and R are defined herein.

Description

PIPERIDIN-4-YL-AZETIDINE DIAMIDES 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. Δ9- 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 Lipidol, 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
CB1/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 has 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 (I)
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is
i) C6-10 aryl is unsubstituted or substituted with a substituent selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci-4alkoxy, cyano, and trifluoromethyl; or
an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
Group b) is
i) Ce-io aryl;
ϋ) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzimidazolyl, benzothienyl, indazolyl, and indolyl; iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; or iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is 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, and ¾; provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of trifluoromethyl, methyl, chloro, cyano, and fluoro; R is hydrogen or hydroxy; 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 (I) 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 (I) 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 (I) 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 Ci-6alkyl 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 to 8 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 the group consisting of 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 nitrogen, oxygen, and sulfur. In the case of 5 membered rings, the heteroaryl ring preferably contains one member of nitrogen, oxygen or sulfur and, in addition, up to 3 additional nitrogens. 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 nitrogens, at most 2 nitrogen atoms are adjacent. Examples of heteroaryl groups include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, isoindolyl, benzofuryl, benzothienyl, indazolyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzis oxazolyl,
benzothiadiazolyl, benzotriazolyl, quinolinyl, isoquinolinyl and quinazolinyl. Unless otherwise noted, the 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, Cl_4, Cl_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:
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 (I) 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) includes 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 (I) 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 (I), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof. In particular, the compounds of Formula (I), 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 (I), 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 (I), 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 and/or dermal allergy, itch, eczema, pharyngitis, enteritis, irritable bowel syndrome, inflammatory bowel diseases including Crohn's Disease, ulcerative colitis, benign prostatic hypertrophy, and nasal hypersensitivity.
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 (I). 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 (I) 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 (I) 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 (I)
Formula (I) wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b); and
Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, 1H- pyrrol-2-yl, and lH-pyrrol-3-yl;
Group b) is
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl
isunsubstituted or substituted with with trifluoromethyl or fluoro; or iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents each of which is independently selected from the group consisting of chloro, fluoro, methyl, and ¾,; provided that no more than one substituent is ¾; and
Rbis selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents each of which is independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro; Group b) is
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl; iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; or
iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rbis selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are optionally independently substituted with one to two trifluoromethyl, methyl, chloro, or fluoro substituents; e) R is hydrogen; and any combination of embodiments a) through e) 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 (I)
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl; Group b) is
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; or iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rbis selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen;
and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof. An embodiment of the p compound of Formula (I)
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl; Group b) is
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; or
iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is and
Rbis selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substitutents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen or hydroxy;
and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
An embodiment of the p compound of Formula (I)
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, lH-pyrrol- 2-yl, and lH-pyrrol-3-yl; Group b) is
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; or
iv) 1 ,3-dihydro-3H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rbis selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of Rb are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen or hydroxy;
and enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof.
An embodiment of the present invention includes a compound of Formula (I)
Formula (I)
selected from the group consisting of
the compound wherein Y is thiazol-4-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is isothiazol-5-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is lH-pyrrol-3-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is thiazol-5-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is phenyl, Z is 5-trifluoromethyl-benzothien-2-yl, and R is OH;
the compound wherein Y is thiazol-4-yl, Z is 3-chloro-6-fluoro-benzothien-2-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-fluoro-benzothien-2-yl, and R is H;
the compound wherein Y s thiazol-4-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H; the compound wherein Y s thiazol-4-yl, Z is 4-(3-trifluoromethylphenyl)-phenyl, and R is H;
the compound wherein Y thiazo -4-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H; the compound wherein Y thiazo -4-yl, Z is 4-(5-trifluoromethylthien-2-yl)-phenyl, and R is H;
the compound wherein Y thiazo -4-yl, Z is 4-(3-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y thiazo ■4-yl, Z is 3-methyl-6-trifluoromethyl-benzothien-2- yl, and R is H;
the compound wherein Y thiazo ■2-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H; the compound wherein Y thiazo -2-yl, Z is 4-(3-trifluoromethylphenyl)-phenyl, and R is H;
the compound wherein Y thiazo -2-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H; the compound wherein Y thiazo -2-yl, Z is 4-(5-trifluoromethylthien-2-yl)-phenyl, and R is H;
the compound wherein Y thiazo -2-yl, Z is 4-(3-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y thiazo -2-yl, Z is 3-methyl-6-trifluoromethyl-benzothien-2- yl, and R is H;
the compound wherein Y lH-pyrrol-2-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H; the compound wherein Y lH-pyrrol-2-yl, Z is 4-(3-trifluoromethylphenyl)-phenyl, and R is H;
the compound wherein Y lH-pyrrol-2-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H; the compound wherein Y lH-pyrrol-2-yl, Z is 4-(5-trifluoromethylthien-2-yl)-phenyl, and R is H;
the compound wherein Y lH-pyrrol-2-yl, Z is 4-(3-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y lH-pyrrol-2-yl, Z is 3-methyl-6-trifluoromethyl- benzothien-2-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is 4-(4-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl-benzothien-2- yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-lH-indol-5- yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 4-(4-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 2-phenyl-benzoxazol-6- yl, and R is H; the compound wherein Y is thiazol-4-yl, Z is 3-chloro-6-trifluoromethyl-benzothien-2- yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(4-fluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(4-trifluoromethylphenyl)-lH-indol-5- yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(3,4-difluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 4-(4-trifluoromethylphenylmethyl)- phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 2-phenyl-benzoxazol-6- yl, and R is H; the compound wherein Y is lH-pyrrol-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is l-(4-fluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is l-(4-trifluoromethylphenyl)-lH-indol- 5-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is l-(3,4-difluorophenyl)-lH-indol-5-yl, and R is H; the compound wherein Y is 2-fluoro-4-phenyl-phenyl, Z is thiazol-2-yl, and R is H; the compound wherein Y is 2- fluoro-4-phenyl-phenyl, Z is thiazol-4-yl, and R is H; the compound wherein Y is lH-pyrrol-2-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H; the compound wherein Y is 3-trifluoromethylphenyl)-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 3-trifluoromethylphenyl)-phenyl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 3-trifluoromethylphenyl)-phenyl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is 3- (3-fluorophenyl)-phenyl, Z is thiazol-2-yl, and R is H; the compound wherein Y is 3-(3-fluorophenyl)-phenyl, Z is thiazol-4-yl, and R is H; the compound wherein Y is 3- (3-fluorophenyl)-phenyl, Z is lH-pyrrol-2-yl, and R is H; the compound wherein Y is 4- (5-trifluoromethyl-thien-2-yl)-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 5-trifluoromethyl-thien-2-yl)-phenyl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 5-trifluoromethyl-thien-2-yl)-phenyl, Z is lH-pyrrol-2- yl, and R is H;
the compound wherein Y is 3-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-2- yl, and R is H;
the compound wherein Y is 3-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenylmethyl)-phenyl, Z is lH-pyrrol- 2-yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-2- yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-2- yl, and R is H; the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is lH-pyrrol- 2-yl, and R is H;
the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is thiazol-2-yl, and R is H; the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is thiazol-4-yl, and R is H; the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is lH-pyrrol-2-yl, and R is H; the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-2- yl, and R is H;
the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is thiazol-2- yl, and R is H;
the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is lH-pyrrol- 2-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-lH-benzimidazol-5-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-lH-benzimidazol- 5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(2,2,2-trifluoroethyl)-lH-benzimidazol- 5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-phenyl-2-methyl-lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 1 -(4-trifluoromethylphenyl)-2-methyl- lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(2,2,2-trifluoroethyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4,4-difluorocyclohexyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(5-chloropyridin-2-yl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 6-trifluoromethyl-benzothien-2-yl, and R is OH;
the compound wherein Y is thiazol-2-yl, Z is l-(2-methylpyridin-4-yl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-phenyl-l,3-dihydro-3H-benzimidazol- 2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-l,3-dihydro-3H- benzimidazol-2-on-5-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-l,3-dihydro-3H- benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-l,3-dihydro- 3H-benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-l,3-dihydro-3H- benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4,4-difluorocyclohexyl)-l,3-dihydro-
3H-benzimidazol-2-on-5-yl, and R is H;
and pharmaceutically acceptable salt forms thereof.
For use in medicine, salts of compounds of Formula (I) refer to non-toxic
"pharmaceutically acceptable salts." Other salts may, however, be useful in the preparation of compounds of Formula (I) or of their pharmaceutically acceptable salt forms thereof. Suitable pharmaceutically acceptable salts of compounds of Formula (I) 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 (I) 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-dichloroacetic 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 (I). 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 (I).
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 (I) 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 (I) 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 :
(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 (I) 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 (I) 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 (I) 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 (I) 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 (I) 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 (I) 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 (I).
Advantageously, a compound of Formula (I) 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 (I) 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 (I) 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 (I) is required for a subject in need thereof.
As MGL Inhibitors, the compounds of Formula (I) 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 (I).
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
DBE 1,2-dibromoethane
DCC N,N -dicyclohexyl-carbodiimi.de
DCE 1,2-dichloroethane
DCM dichloromethane
DIPEA diisopropylethylamine
DMAP 4-(N,N-dimethylamino)pyridine
DMA N,N-dimethylacetamide
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-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate
ΗΕΚ human embryonic kidney
HEPES (4-(2 -hydroxy ethyl)- 1 -piperazineethane
sulfonic acid
HMPA hexamethylphosphoramide
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 intermediates that are useful for the preparation of compounds of Formula (I) wherein R is hydrogen.
Scheme A
Pd- Cross-Coupling
A5 A compound of formula Al (wherein P is a conventional amino protecting group) is either commercially available or may be prepared by known methods described in the scientific literature. A compound of formula Al may be treated with zinc metal in the presence of TMS-C1, in an aprotic solvent, followed by addition of a compound of formula A2, in the presence of palladium catalyst to afford a compound of formula A3. Treatment with benzyl bromide affords the pyridinium bromide of formula A4. A compound of formula A4 may be reduced to a compound of formula A5 in the presence of a hydride source such as, sodium borohydride, in an organic alcoholic solvent such as, ethanol. Removal of the benzyl group and reduction of the double bond may be achieved by palladium catalyzed hydrogenation to afford the desired intermediate of formula A6.
Scheme B illustrates a route for the preparation of compounds of Formula (I)-B wherein Y and Z are as defined herein and R of Formula (I) is hydrogen.
Scheme B
The amino protecting group (P) of a compound of formula A3 may be removed by conventional synthetic methods to afford a secondary amine of formula Bl. The amino group may be coupled with a carboxylic acid of formula B2 (wherein Q is hydroxy) in the presence of an appropriate coupling agent such as HATU, DCC, EDC, HBTU, PyBrOP, and the like, optionally in the presence of a base such as DIPEA, to afford an amide of formula B3. Similarly, an acid chloride of formula B2 (wherein Q is chloro) may be used to effect the acylation of a compound of formula Bl. In such case a non- nucleophilic base such as pyridine may be added to afford an amide of formula B3. Reduction of the pyridine ring of a compound of formula B3 may be achieved by palladium catalyzed hydrogenation to afford a compound of formula B4. A second acylation with an appropriately Y-substituted carboxylic acid or acid chloride of formula B5 affords a compound of Formula (I)-B wherein R of Formula (I) is hydrogen. Scheme C illustrates an alternate route for the preparation of compounds of Formula (I)-B wherein Y and Z are as defined herein and R of Formula (I) is hydrogen.
Scheme C
The compound of formula A6 may be acylated according to the synthetic methods described under Scheme B to afford the acylated compound of formula CI.
Conventional amino deprotection affords the amine of formula C2, which may undergo a second acylation as previously described to afford a compound of Formula (I)-B.
Scheme D illustrates a route for the preparation of compounds of Formula (I)-D wherein Y and Z are as defined herein and R of Formula (I) is hydroxy.
Scheme D
ac c or e acy at on
Compounds of formulae Dl and D2 are either commercially available or may be prepared by known methods described in the scientific literature. A compound of formula Dl may be treated with samarium iodide in the presence of HMPA, in an aprotic solvent, followed by the addition of a ketone of formula D2 to afford the condensed product of formula D3. Removal of the benzhydryl group may be effected by palladium catalyzed hydrogenation to afford the free amine of formula D4. The amine of formula D4 may be acylated with a Z -substituted compound of formula B2 by the methods previously described herein to afford a compound of Formula (I)-D.
Scheme E illustrates an alternate route for the preparation of compounds of Formula (I)-D wherein Y and Z are as defined herein and R of Formula (I) is hydroxy. Scheme E
A compound of formula Dl may be condensed with a compound of formula El in the presence of samarium iodide and HMPA to afford a compound of formula E2.
Removal of the amino protecting group (P) using conventional synthetic methods affords a compound of formula E3. Acylation with a compound of formula B5 affords a compound of formula E4, which, upon benzhydryl removal, affords a free amine of formula E5. A second acylation with an appropriately substituted Z- substituted carboxylic acid or acid chloride of formula B2 affords a compound of Formula (I)-D.
1b
Pd(dppf)CI2 / Cul, DMA,
80 °C, 2 h (89%)
NaBH4, EtOH Pd(OH)2, H2 (50 psi)
N-Boc * HN N-Boc
20 °C, 1 h (91 %) EtOH, 20 °C, 96 h \ / ^
(100%) 1 g
A. teri-Butyl 3-(pyridin-4-yl)azetidine-l-carboxylate, lc. A 1 -liter 3-neck round bottom flask equipped with a thermocouple, magnetic stirrer, condenser, heating mantle, and 2 inlet adapter was charged with anhydrous dimethylacetamide (DMA, 100 mL) and zinc (42.94 g, 650.2 mmol). The mixture was stirred at 20 °C while a mixture of 1,2-dibromoethane (DBE, 5.38 mL, 62.34 mmol) and trimethylsilyl chloride (TMS-C1, 7.54 mL, 59.28 mmol) was added at a rate to maintain the temperature below 65 °C over 30 min. The resulting slurry was aged for 15 min. A solution of tert-butyl 3-iodoazetidine-l-carboxylate la (122.78 g, 420.69 mmol) in DMA (201 mL) was added dropwise over 1 h at a rate to maintain the temperature below 65 °C and the milky suspension was stirred for 30 min while slowly cooling to 20 °C.
Another 3 -liter 4-neck round bottom flask equipped a thermocouple, mechanical stirrer, condenser, heating mantle, and N2 inlet adapter was charged with [Ι, Γ- bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane complex (4.73 g, 5.74 mmol), cuprous iodide (2.19 g, 11.47 mmol), and 4-iodopyridine lb (80.0 g, 382.44 mmol) in DMA (255 mL) under N2. The resulting mixture was degassed with alternate vacuum/N2 purges. The above prepared zinc iodide reagent of compound la in DMA was added as a suspension. The mixture was degassed with vacuum/N2 twice and then heated to 80 °C. (Note: The reaction was exothermic.) The progress of the reaction was monitored by HPLC and LC-MS and was complete after 2 h. The reaction mixture was cooled to 40 °C; EtOAc (1.6 L) was added and the mixture was stirred for 10 min. The insoluble material (excess Zn and Cu
complexes/salts) was removed by passing through a diatomaceous earth pad, which was washed with EtOAc (200 mL x 2). The combined filtrate was stirred with 1 N aqueous NH4C1 (0.8 L) at 20 °C for 30 min and the aqueous layer (pH = 5-6) was adjusted to pH = 9-10 using 3 N aqueous NaOH solution (-480 mL) while a significant amount of brown precipitate was formed. The precipitate was removed by paper filtration and was washed with deionized water (100 mL). The separated aqueous phase was extracted with EtOAc (1 L), and the combined organic phases were treated with saturated aqueous NH4C1 (0.8 L x 2) and stirred for 15 min (repeated again), washed with 5% aqueous NaHS03 (500 mL) and brine (1 L), and dried over MgS04. The organic solvent was concentrated at 66 °C under house vacuum (-120 mmHg) and then high-vacuum (12 mmHg) to afford 80.1 g (89% isolated yield) of crude compound lc as an oil (88% purity at 254 nm and 86% purity at 230 nm; HPLC area%. Retention time = 2.39 min), which was used in the next step without further purification.
B. l-Benzyl-4-(l-(teri-butoxycarbonyl)azetidin-3-yl)pyridin-l-ium bromide, le. A 2-liter 4-neck round bottom flask equipped a thermocouple, mechanical stirrer, condenser, and N2 inlet adapter was charged with crude compound lc (78.22 g, 290.5 mmol) and acetonitrile (503 mL). The mixture was stirred at 20 °C and benzyl bromide Id (36.41 mL, 299.2 mmol) was added. The mixture was warmed to 80 °C and stirred for 1 h. The reaction was cooled to 20 °C and the solvent was concentrated at 60 °C under house-/ high-vacuum. The resulting material was chased with MeOH (100 mL) once to afford 128.9 g (109% isolated yield; 80-84% purity; HPLC area%. HPLC retention time = 3.61 min) of crude le as a syrup, which was used in next step without further purification.
C. tert-Butyl 3-(l-benzyl-l,2,3,6-tetrahydropyridin-4-yl)azetidine-l- carboxylate, If. A 3-liter 4-neck round bottom flask equipped a thermocouple, mechanical stirrer, condenser, and N2 inlet adapter was charged with crude compound le (117.73 g, 232.4 mmol) and EtOH (1.04 L). The solution was cooled to 0 °C with stirring; sodium tetrahydroborate (17.8 g, 464.7 mmol) was added and the mixture was stirred at 0 °C for 10 min, then gradually warmed to 20 °C and stirred for 1 h. The mixture was cooled to 0 °C and quenched with half-saturated NaHCC (100 mL, prepared by adding 50 mL of deionized water to 50 mL of saturated aHCOs). The organic solvent was concentrated at 60 °C under house vacuum to a wet solid, which was dissolved in EtOAc (1.5 L) and stirred for 10 min with half saturated aHC03 (1L). After phase separation, the milky aqueous layer (pH = 6-7) was adjusted to pH = 10-11 using 3 N aqueous NaOH solution and extracted with EtOAc (500 mL). The combined organic phases were washed with brine (500 mL) and then concentrated at 60°C under house-/high-vacuum to afford 98.7 g of crude If as a syrup, which was purified using flash column chromatography (silica gel, EtOAc/heptane/MeOH 20/80/0 - 50/50/3) to afford 67.1 1 g (91% isolated yield, 95% purity at 210 nm; HPLC area%) of compound If as a yellow syrup.
D. teri-Butyl 3-(piperidin-4-yl)azetidine-l-carboxylate, lg. A 500-mL Parr pressure bottle was charged with compound If (18.4 g, 54.3 mmol), EtOH (152 mL), and Pd(OH)2 (1.91 g). The mixture was purged twice with 2 and then shaken under a 50 psi ¾ atmosphere at 20 °C. After 40 h, the ¾ was removed and additional Pd(OH)2 (1.9 g) was added to the mixture of If, dihydro-lf, and lg, which was purged twice with 2 and shaken under a 50 psi ¾ atmosphere at 20 °C for an additional 56 h. The catalyst was removed by filtration though a diatomaceous earth pad, which was washed with MeOH (50 mL x 3). Concentration of the filtrate at 50 °C under high- vacuum (~10 mmHg) afforded 13.4 g (103% isolated yield, 97% pure at 210 nm, HPLC area%) of pure compound lg as a slight yellowish, thick oil, which contained a trace amount of EtOH residue by ^- MR analysis.
E. tert-Butyl 3-(l-(thiazole-2-carbonyl)piperidin-4-yl)azetidine-l- carboxylate, li. To a stirring solution of compound lg (14.3 mmol, 3.44 g) and thiazole-2-carboxylic acid lh (15.7 mmol, 2.03 g) in 50 mL of CH2CI2 was added Ets (42.9 mmol, 5.98 mL). After 20 min at 20 °C, HATU (17.2 mmol, 6.53 g) was added and the mixture was stirred at 20 °C for 5 h. Water was added to the mixture and the organic layer was separated, dried over MgS04, and concentrated. The residue was purified using flash column chromatography (silica gel, 30-70% EtO Ac/heptane) to give 3.8 g (75% yield) of compound li. MS m/z 37 '4.2 (M+Na+), 296.1 (M+H-C4H8), 252.1 (M+H-C5H802). F. (4-(Azetidin-3-yl)piperidin-l-yl)(thiazol-2-yl)methanone, lj. A portion of TFA (20 mL) was added to a solution of compound li (10.8 mmol, 3.8 g) in 100 mL of CH2CI2. The solution was stirred at 20 °C for 5 h. The solvent was removed under vacuum and the residue was partitioned between CH2CI2 and IN aqueous NaOH. The organic layer was dried over MgS04 and concentrated to give 2.6 g (85% yield) of compound lj, which was used in the next reaction without purification. MS m/z 252.1 (M+H+).
G. 4-(l-{[3-Chloro-6-(trifluoromethyl)-l-benzothiophen-2- yl]carbonyl}azetidin-3-yl)-l-(l,3-thiazol-2-ylcarbonyl)piperidine, Cpd 29. To a stirring solution of compound lj (1.59 mmol, 0.40 g) and 3-chloro-6-
(trifluoromethyl)benzo[b]thiophene-2-carboxylic acid lk (1.75 mmol, 0.49 g) in 10 mL of CH2CI2 was added Et3N (6.37 mmol, 0.89 mL). After 20 min at 20 °C, HATU (1.91 mmol, 0.73 g) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 210 mg (26% yield) of Cpd 29. XH NMR (CD3OD, 400 MHz): δ = 8.40 (s, 1H), 8.08 (d, J=8.6 Hz, 1H), 7.93 (br. s., 1H), 7.75-7.85 (m, 2H), 5.31 (t, J=10.8 Hz, 1H), 4.63 (t, J=11.0 Hz, 1H), 4.37 (t, J=8.2 Hz, 1H), 4.28 (t, J=9.3 Hz, 1H), 4.10 (br. s., 1H), 4.00 (br. s., 1H), 3.15-3.29 (m, 1H), 2.82-3.00 (m, 1H), 2.47- 2.63 (m, 1H), 1.67-2.02 (m, 3H), 1.05-1.34 (m, 2H). MS m/z 514.0 (M+H+).
Following the procedure described above for Example 1 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Cpd Name and data
4-( 1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2- yl]carbonyl}azetidin-3-yl)-l-(lH-pyrrol-2-ylcarbonyl)piperidine.
XH NMR (CD3OD, 400 MHz): δ = 8.41 (s, 1H), 8.10 (d, J = 8.6
Hz, 1H), 7.83 (d, J = 8.1 Hz, 1H), 6.90 (dd, J = 2.4, 1.2 Hz, 1H),
41 6.54 (dd, J = 3.7, 1.2 Hz, 1H), 6.13 - 6.21 (m, 1H), 4.57 (t, J = 13.2
Hz, 2H), 4.33 - 4.42 (m, 1H), 4.29 (t, J = 9.4 Hz, 1H), 4.10 (dd, J =
9.4, 6.2 Hz, 1H), 3.94 - 4.04 (m, 1H), 3.05 (br. s., 2H), 2.49 - 2.63
(m, 1H), 1.70 - 1.98 (m, 3H), 1.06 - 1.25 (m, 2H). MS m/z 496.2
(M+H+).
1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} -
69 4-[l-(l,3-thiazol-2-ylcarbonyl)azetidin-3-yl]piperidine. MS m/z
514.0 (M+H+).
1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} -
70 4-[l-(l,3-thiazol-4-ylcarbonyl)azetidin-3-yl]piperidine. MS m/z
514.0 (M+H+).
1 - { [3 -Chloro-6-(trifluoromethyl)- 1 -benzothiophen-2-yl]carbonyl} -
71 4-[l-(lH-pyrrol-2-ylcarbonyl)azetidin-3-yl]piperidine. MS m/z
496.0 (M+H+).
Example 2
A. 3-Chloro-6-fluorobenzo[b]thiophene-2-carbonyl chloride, 2b. Thionyl chloride (73.7 mmol, 5.36 mL) was added to a mixture of 4-fluorocinnamic acid 2a (21.1 mmol, 3.5 g) and pyridine (2.53 mmol, 0.2 mL). The mixture was heated at 135 °C for 30 min and then cooled to room temperature. The crude mixture was triturated with hot hexanes to remove the solid pyridinium hydrochloride by-product. Compound 2b was isolated from the combined hexanes solutions.
B. 4-{l-[(3-Chloro-6-fluoro-l-benzothiophen-2-yl)carbonyl]azetidin-3-yl}- l-(l,3-thiazol-2-ylcarbonyl)piperidine, Cpd 8). A solution of compound 2b (0.45 mmol, 112 mg) in 4 mL of CH2CI2 was added to a solution of compound lj mono-TFA salt (0.41 mmol, 150 mg) in Et3N (2.46 mmol, 0.34 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 3 h. The crude product was purified by preparative reverse- phase chromatography to afford 18 mg (9% yield) of Cpd 8. XH NMR (CD3OD, 400 MHz): δ = 7.88-7.97 (m, 2H), 7.81 (d, J=2.9 Hz, 1H), 7.76 (dd, J=8.8, 2.2 Hz, 1H), 7.36 (td, J=9.0, 2.3 Hz, 1H), 5.22-5.37 (m, 1H), 4.55-4.70 (m, 1H), 4.32-4.44 (m, 1H), 4.23- 4.32 (m, 1H), 4.11 (br. s., 1H), 3.91-4.05 (m, 1H), 3.15-3.28 (m, 1H), 2.84-3.00 (m, 1H), 2.48-2.62 (m, 1H), 1.70-2.02 (m, 3H), 1.10-1.29 (m, 2H). MS m/z 464.1 (M+H+).
Following the procedure described above for Example 2 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compound of the present invention was prepared:
A. 3-Methyl-6-(trifluoromethyl)benzo[b]thiophene-2-carboxylate, 3c.
Methyl thioglycolate 3b (30.3 mmol, 2.76 mL) was added dropwise to a suspension of NaH (60% oil dispersion, 75.8 mmol, 3.03 g) in 10 mL of THF and 50 mL of DMSO at 20 °C. The mixture was stirred for 15 min and a solution of l-(2-fluoro-4- (trifluoromethyl)phenyl)ethanone 3a (24.3 mmol, 5.0 g) in 10 mL of DMSO was added. The reaction mixture was stirred at 20 °C for 4 h and water was added. The mixture was extracted with EtOAc. The organic layer was dried over MgS04 and concentrated to give compound 3c as a white solid.
B. 4-(l-{[3-Methyl-6-(trifluoromethyl)-l-benzothiophen-2- yl]carbonyl}azetidin-3-yl)-l-(l,3-thiazol-2-ylcarbonyl)piperidine, Cpd 20. To a stirring solution of compound lj mono-TFA salt (0.27 mmol, 100 mg) and compound 3c (0.30 mmol, 78 mg) in 4 mL of CH2C12 was added Et3N (1.09 mmol, 0.15 mL). After 20 min at 20 °C, HATU (0.33 mmol, 125 mg) was added and the mixture was stirred at 20 °C for 20h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 34 mg (25% yield) of Cpd 20. 'H NMR (CD3OD, 400 MHz): δ = 8.29 (s, 1H), 8.04 (d, J=8.6 Hz, 1H), 7.93 (br. s., 1H), 7.81 (d, J=3.2 Hz, 1H), 7.71 (d, J=8.3 Hz, 1H), 5.31 (br. s., 1H), 4.564.69 (m, 1H), 4.324.42 (m, 1H), 4.26 (t, J=9.2 Hz, 1H), 4.09 (br. s., 1H), 3.98 (br. s., 1H), 3.23 (br. s., 1H), 2.92 (br. s., 1H), 2.61 (s, 3H), 2.472.59 (m, 1H), 1.702.00 (m, 3H), 1.031.35 (m, J=9.3 Hz, 2H). MS m/z 494.1 (M+H+).
Following the procedure described above for Example 3, and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Cpd Name and data
4-( 1 - { [3 -Methyl-6-(trifluoromethyl)- 1 -benzothiophen-2-
14 yl]carbonyl}azetidin-3-yl)-l-(l,3-thiazol-4-ylcarbonyl)piperidine.
MS m/z 494.1 (M+H+).
4-( 1 - { [3 -Methyl-6-(trifluoromethyl)- 1 -benzothiophen-2-
26 yl]carbonyl}azetidin-3-yl)-l-(lH-pyrrol-2-ylcarbonyl)piperidine.
MS m/z 4Ί6.2 (M+H+).
1 - { [3-Methyl-6-(trifluoromethyl)- 1 -benzothiophen-2-
60 yl]carbonyl}-4-[l-(l,3-thiazol-2-ylcarbonyl)azetidin-3- yl]piperidine. MS m/z 494.1 (M+H+).
1 - { [3-Methyl-6-(trifluoromethyl)- 1 -benzothiophen-2-
61 yl]carbonyl}-4-[l-(l,3-thiazol-4-ylcarbonyl)azetidin-3- yl]piperidine. MS m/z 494.1 (M+H+).
1 - { [3-Methyl-6-(trifluoromethyl)- 1 -benzothiophen-2-
62 yl]carbonyl}-4-[l-(lH-pyrrol-2-ylcarbonyl)azetidin-3- yl]piperidine. MS m/z 476.2 (M+H+).
Example 4
A. 3'-(Trifluoromethyl)-[l,l'-biphenyl]-4-carboxylic acid, 4c. A portion of Pd(dppf)Ci2 (1.49 mmol, 1.09 g) was added to a suspension of 4-bromobenzoic acid 4a (14.9 mmol, 3.0 g), 3-trifluoromethylboronic acid 4b (17.9 mmol, 3.4 g), and CS2CO3 (37.3 mmol, 12.2 g) in 30 mL of dioxane and 7.5 mL of EtOH. The mixture was stirred at 80 °C for 2 h. After cooling, the solid was collected by filtration and washed with MeOH. The filtrate was concentrated and partitioned between EtOAc and IN aqueous HCl. The organic layer was washed with brine, dried over MgS04, and concentrated. CH2CI2 was added to the residue and the resulting solid was collected by filtration, washed with CH2CI2, and dried to give 3.58 g (86% yield) of compound 4c, which was used in the next step without further purification.
B. l-(l,3-Thiazol-2-ylcarbonyl)-4-(l-{[3'-(trifluoromethyl)biphenyl-4- yl]carbonyl}azetidin-3-yl)piperidine, Cpd 16. To a stirring solution of compound lj mono-TFA salt (0.27 mmol, 100 mg) and compound 4c (0.30 mmol, 80 mg) in 4 mL of CH2CI2 was added Et3N (1.09 mmol, 0.15 mL). After 20 min at 20 °C, HATU (0.33 mmol, 125 mg) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 57 mg (42% yield) of Cpd 16. ¾ NMR (CD3OD, 400 MHz): δ = 7.93 (br. s., 3H), 7.73-7.83 (m, 5H), 7.63-7.73 (m, 2H), 5.24-5.38 (m, 1H), 4.64 (t, J=10.8 Hz, 1H), 4.46 (t, J=8.2 Hz, 1H), 4.13-4.31 (m, 2H), 3.90-4.01 (m, 1H), 3.15- 3.29 (m, 1H), 2.84-2.99 (m, 1H), 2.44-2.60 (m, 1H), 1.73-2.00 (m, 3H), 1.08-1.32 (m, 2H). MS m/z 500.3 (M+H+).
Following the procedure described above for Example 4 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
4-14 4-15 4-16 4-17 4-18 4-19
4-121 4-122 4-123 4-124 4-125 4-T26 Following the procedure described above for Example 4, and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Cpd Name and data
1 -( 1 ,3 -Thiazol-2-ylcarbonyl)-4- [ 1 -( {4- [5 -
18 (trifluoromethyl)thiophen-2-yl]phenyl}carbonyl)azetidin-3- yl]piperidine. MS m/z 506.1 (M+H+).
4- { 1 - [(3 -Fluorobiphenyl-4-yl)carbonyl] azetidin-3 -yl} - 1 -( 1 H-
21
pyrrol-2-ylcarbonyl)piperidine. MS m/z 432.1 (M+H+).
1 -( 1 H-Pyrrol-2-ylcarbonyl)-4-( 1 - { [3 '-
22 (trifluoromethyl)biphenyl-4-yl]carbonyl} azetidin-3 - yl)piperidine. MS m/z 482.2 (M+H+).
4-{ l-[(3'-Fluorobiphenyl-3-yl)carbonyl]azetidin-3-yl}-l-
23
(lH-pyrrol-2-ylcarbonyl)piperidine. MS m/z 432.1 (M+H+). l-(lH-Pyrrol-2-ylcarbonyl)-4-[l-({4-[5-
24 (trifluoromethyl)thiophen-2-yl]phenyl}carbonyl)azetidin-3- yl]piperidine. MS m/z 488.3 (M+H+).
1 -[(3 -Fluorobiphenyl-4-yl)carbonyl]-4-[ 1 -( 1 ,3 -thiazol-2-
45
ylcarbonyl)azetidin-3-yl]piperidine. MS m/z 450.1 (M+H+).
1 -[(3 -Fluorobiphenyl-4-yl)carbonyl]-4-[ 1 -( 1 ,3 -thiazol-4-
46
ylcarbonyl)azetidin-3-yl]piperidine. MS m/z 450.1 (M+H+). l-[(3-Fluorobiphenyl-4-yl)carbonyl]-4-[l-(lH-pyrrol-2-
47
ylcarbonyl)azetidin-3-yl]piperidine. MS m/z 432.1 (M+H+).
4- [ 1 -( 1 ,3 -Thiazol-2-ylcarbonyl)azetidin-3 -yl]- 1 - { [3 '-
48 (trifluoromethyl)biphenyl-4-yl]carbonyl}piperidine.
MS m/z 500.3 (M+H+).
4- [ 1 -( 1 ,3 -Thiazol-4-ylcarbonyl)azetidin-3 -yl]- 1 - { [3 '-
49 (trifluoromethyl)biphenyl-4-yl]carbonyl}piperidine.
MS m/z 500.1 (M+H+).
4-[ 1 -( 1 H-Pyrrol-2-ylcarbonyl)azetidin-3 -yl] - 1 - { [3 '-
50 (trifluoromethyl)biphenyl-4-yl]carbonyl}piperidine.
MS m/z 482.2 (M+H+).
1 -[(3 '-Fluorobiphenyl-3-yl)carbonyl]-4-[ 1 -( 1 ,3 -thiazol-2-
51 ylcarbonyl)azetidin-3-yl]piperidine.
MS m/z 450.1 (M+H+). Cpd Name and data
l-[(3'-Fluorobiphenyl-3-yl)carbonyl]-4-[l-(l,3-thiazol-4-
52 ylcarbonyl)azetidin-3-yl]piperidine.
MS m/z 450.1 (M+H+).
l-[(3'-Fluorobiphenyl-3-yl)carbonyl]-4-[l-(lH-pyrrol-2-
53 ylcarbonyl)azetidin-3-yl]piperidine.
MS m/z 432.1 (M+H+).
4- [ 1 -( 1 ,3 -Thiazol-2-ylcarbonyl)azetidin-3 -yl] - 1 -( {4- [5 -
54 (trifluoromethyl)thiophen-2-yl]phenyl}carbonyl)piperidine.
MS m/z 506.1 (M+H+).
4- [ 1 -( 1 ,3 -Thiazol-4-ylcarbonyl)azetidin-3 -yl] - 1 -( {4- [5 -
55 (trifluoromethyl)thiophen-2-yl]phenyl}carbonyl)piperidine.
MS m/z 506.1 (M+H+).
4-[ 1 -( lH-Pyrrol-2-ylcarbonyl)azetidin-3 -yl]- 1 -( {4-[5-
56 (trifluoromethyl)thiophen-2-yl]phenyl}carbonyl)piperidine.
MS m/z 488.3 (M+H+).
Example 5
A. Methyl 4-(3-(trifluoromethyl)benzyl)benzoate, 5b. A portion of
Pd(dppf)Ci2 (0.87 mmol, 0.64 g) was added to a suspension of methyl 4- (bromomethyl)benzoate 5a (8.73 mmol, 2.0 g), 3-trifluoromethylboronic acid 4b (10.5 mmol, 1.99 g), and a2C03 (17.5 mmol, 1.85 g) in 20 mL of dioxane and 5 mL of water. The mixture was stirred at 80 °C for 3 h. After cooling, the solid was collected by filtration and washed with EtOAc. The filtrate was washed with IN aqueous HC1 and brine, dried over MgS04, and concentrated. The crude product was purified by flash column chromatography (silica gel, 0-10% EtOAc: heptane) to give 2.2 g (85% yield) of compound 5b. MS m/z 295.2 (M+H+).
B. 4-(3-(Trifluoromethyl)benzyl)benzoic acid, 5c. IN aqueous NaOH (12.9 mmol, 12.9 mL) was added to a suspension of compound 5b (6.46 mmol, 1.9 g) in 75 mL of EtOH. The mixture was stirred at 20 °C for 20 h. The reaction mixture was concentrated and the residue was acidified with IN aqueous HC1. The resulting solid was collected by filtration and dried to afford 1.6 g (87%) of compound 5c, which was used in the next reaction without further purification. MS m/z 281.1 (M+H+).
C. l-(l,3-Thiazol-2-ylcarbonyl)-4-[l-({4-[3- (trifluoromethyl)benzyl]phenyl}carbonyl)azetidin-3-yl]piperidine, Cpd 19. To a stirring solution of compound lj mono-TFA salt (0.27 mmol, 100 mg) and compound 5c (0.30 mmol, 84 mg) in 4 mL of CH2C12 was added Et3N (1.09 mmol, 0.15 mL). After 20 min at 20 °C, HATU (0.33 mmol, 125 mg) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 46 mg (33% yield) of Cpd 19. 'H NMR (CD3OD, 400 MHz): δ = 7.92 (br. s., 1H), 7.79 (d, J=3.2 Hz, 1H), 7.57-7.64 (m, J=8.1 Hz, 2H), 7.43-7.53 (m, 4H), 7.28-7.36 (m, J=8.1 Hz, 2H), 5.29 (t, J=10.0 Hz, 1H), 4.61 (t, J=10.9 Hz, 1H), 4.38 (t, J=8.6 Hz, 1H), 4.20 (t, J=9.3 Hz, 1H), 4.10 (s,
2H), 4.07-4.15 (m, 1H), 3.85-3.96 (m, 1H), 3.10-3.27 (m, 1H), 2.78-2.96 (m, 1H), 2.36- 2.53 (m, 1H), 1.66-1.93 (m, 3H), 1.03-1.29 (m, 2H). MS m/z 514.2 (M+H+).
Following the procedure described above for Example 5 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compound was prepared:
Following the procedure described above for Example 5, and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Cpd Name and data
4-[ 1 -( 1 ,3 -Thiazol-4-ylcarbonyl)azetidin-3 -yl] - 1 -( {4- [3 -
58 (trifluoromethyl)benzyl]phenyl}carbonyl)piperidine
MS m/z 514.2 (M+H+).
4-[ 1 -( 1 H-Pyrrol-2-ylcarbonyl)azetidin-3 -yl] - 1 -( {4- [3 -
59 (trifluoromethyl)benzyl]phenyl}carbonyl)piperidine
MS m/z 496.2 (M+H+).
4-[ 1 -( 1 ,3-Thiazol-2-ylcarbonyl)azetidin-3 -yl]- 1 -( {4-[4-
63 (trifluoromethyl)benzyl]phenyl}carbonyl)piperidine
MS m/z 514.2 (M+H+).
4-[ 1 -( 1 ,3 -Thiazol-4-ylcarbonyl)azetidin-3 -yl] - 1 -( {4- [4-
64 (trifluoromethyl)benzyl]phenyl}carbonyl)piperidine
MS m/z 514.2 (M+H+).
4-[ 1 -( 1 H-Pyrrol-2-ylcarbonyl)azetidin-3-yl]- 1 -( {4-[4-
65 (trifluoromethyl)benzyl]phenyl}carbonyl)piperidine
MS m/z 496.2 (M+H+).
Example 6
A. 2-Phenylbenzo[< ]oxazole-6-carboxylic acid, 6c. A solution of methyl 4- amino-3-hydroxybenzoate 6a (29.9 mmol, 5.0 g) and benzaldehyde 6b (29.9 mmol, 3.02 mL) in 150 mL of MeOH was stirred at 20 °C for 3 h. The solvent was removed under vacuum and the residue was mixed with 150 mL of acetonitrile. Lead (IV) acetate (29.9 mmol, 13.3 g) was added in one portion and the mixture was refluxed for 20 min. After cooling, the precipitate was removed by filtration and washed with acetonitrile. The filtrate and wash solutions were stirred with 3N aqueous NaOH (120 mmol, 40 mL) at 50 °C for 20 h. After cooling, the reaction mixture was acidified and filtered to give 6.0 g (79%) of compound 6c. MS m/z 240.0 (M+H+).
B. 2-Phenyl-6-({3-[l-(l,3-thiazol-2-ylcarbonyl)piperidin-4-yl]azetidin-l- yl}carbonyl)-l,3-benzoxazole, Cpd 28. To a stirring solution of compound lj (0.40 mmol, 100 mg) and compound 6c (0.44 mmol, 100 mg) in 4 mL of CH2CI2 was added Et3N (1.59 mmol, 0.22 mL). After 20 min at 20 °C, HATU (0.48 mmol, 180 mg) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 75 mg (39% yield) of Cpd 28. XH NMR (CD3OD, 400 MHz): δ = 8.23-8.32 (m, 2H), 8.01 (s, 1H), 7.89-7.97 (m, 1H), 7.77-7.85 (m, 2H), 7.73 (dd, J=8.3, 1.2 Hz, 1H), 7.54-7.68 (m, 3H), 5.30 (t, J=11.0 Hz, 1H), 4.64 (t, J=12.0 Hz, 1H), 4.43-4.54 (m, 1H), 4.18-4.34 (m, 2H), 3.92-4.05 (m, 1H), 3.16-3.29 (m, 1H), 2.84-2.99 (m, 1H), 2.47-2.60 (m, 1H), 1.72- 2.02 (m, 3H), 1.10-1.34 (m, 2H). MS m/z 473.1 (M+H+). Following the procedure described above for Example 6, and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Example 7
A. Methyl l-(4-fluorophenyl)-indole-5-carboxylate, 7c. A mixture of methyl indole-5-carboxylate 7a (0.5 g, 2.85 mmol), 1 -bromo-4-fluoro-benzene 7b (2 mL, 18.21 mmol), Cul (0.544 g, 2.85 mmol), and K2C03 (0.591 g, 4.28 mmol) was heated in a microwave reactor at 220 °C for 2.5 h. 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% EtO Ac/heptane) to give 0.58 g of compound 7c. MS m/z 270.1 (M+H+).
B. l-(4-Fluorophenyl)-indole-5-carboxylic acid, 7d. A mixture of methyl 1- (4-fluorophenyl)-indole-5-carboxylate 7c (0.58 g, 2.15 mmol) and LiOH'H20 (0.36 g, 8.6 mmol) in THF (15 mL) and H2O (10 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 0.5 g of compound 7d. MS m/z 256.2 (M+H+).
C. l-(4-Fluorophenyl)-5-({3-[l-(l,3-thiazol-2-ylcarbonyl)piperidin-4- yl]azetidin-l-yl}carbonyl)-lH-indole, Cpd 30. To a stirring solution of compound lj (0.40 mmol, 100 mg) and compound 7d (0.44 mmol, 110 mg) in 4 mL of CH2CI2 was added Et3N (1.59 mmol, 0.22 mL). After 20 min at 20 °C, HATU (0.48 mmol, 180 mg) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 165 mg (85% yield) of Cpd 30. XH NMR (CD3OD, 400 MHz): δ = 7.99 (s, 1H), 7.92 (br. s., 1H), 7.79 (d, J=3.2 Hz, 1H), 7.44-7.58 (m, 5H), 7.30 (t, J=8.7 Hz, 2H), 6.77 (d, J=3.4 Hz, 1H), 5.28 (br. s., 1H), 4.53-4.69 (m, 1H), 4.39-4.51 (m, 1H), 4.10-4.29 (m, 2H), 3.95 (br. s., 1H), 3.10-3.27 (m, 1H), 2.89 (t, J=10.5 Hz, 1H), 2.33-2.56 (m, 1H), 1.65-1.98 (m, J=10.8 Hz, 3H), 1.19 (br. s., 2H). MS m/z 489.1 (M+H+).
Following the procedure described above for Example 7, and substituting the appropriate reagents, starting materials, and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
Following the procedure described above for Example 7, and substituting the appropriate reagents, starting materials, and purification methods known to those skilled in the art, the following compounds of the invention were prepared:
Cpd Name and data
l-(4-Fluorophenyl)-5-({3-[l-(l,3-thiazol-4-ylcarbonyl)piperidin-4-
36
yl]azetidin-l-yl}carbonyl)-lH-indole. MS m/z 489.1 (M+H+).
5-( { 3 - [ 1 -( 1 ,3 -Thiazol-4-ylcarbonyl)piperidin-4-yl] azetidin- 1 -
37 yl}carbonyl)-l-[4-(trifluoromethyl)phenyl]-lH-indole. MS m/z 539.2 (M+H+).
l-(4-Fluorophenyl)-5-({3-[l-(lH-pyrrol-2-ylcarbonyl)piperidin-4-
42
yl]azetidin-l-yl}carbonyl)-lH-indole. MS m/z 472.2 (M+H+).
5-( {3 -[ 1 -( lH-Pyrrol-2-ylcarbonyl)piperidin-4-yl]azetidin- 1 -
43 yl}carbonyl)-l-[4-(trifluoromethyl)phenyl]-lH-indole. MS m/z 521.3 (M+H+).
l-(4-Fluorophenyl)-5-({4-[l-(l,3-thiazol-2-ylcarbonyl)azetidin-3-
72
yl]piperidin-l-yl}carbonyl)-lH-indole. MS m/z 489.1 (M+H+).
1 -(4-Fluorophenyl)-5-( {4- [ 1 -( 1 ,3 -thiazol-4-ylcarbonyl)azetidin-3 -
73
yl]piperidin-l-yl}carbonyl)-lH-indole. MS m/z 489.1 (M+H+). l-(4-Fluorophenyl)-5-({4-[l-(lH-pyrrol-2-ylcarbonyl)azetidin-3-
74
yl]piperidin-l-yl}carbonyl)-lH-indole. MS m/z 471.3 (M+H+).
5-({4-[l-(l,3-Thiazol-2-ylcarbonyl)azetidin-3-yl]piperidin-l-
75 yl}carbonyl)-l-[4-(trifluoromethyl)phenyl]-lH-indole. MS m/z 539.2 (M+H+).
5-({4-[l-(l,3-Thiazol-4-ylcarbonyl)azetidin-3-yl]piperidin-l-
76 yl}carbonyl)-l-[4-(trifluoromethyl)phenyl]-lH-indole. MS m/z 539.2 (M+H+).
5-({4-[l-(lH-Pyrrol-2-ylcarbonyl)azetidin-3-yl]piperidin-l-
77 yl}carbonyl)-l-[4-(trifluoromethyl)phenyl]-lH-indole. MS m/z 521.3 (M+H+).
Cpd Name and data
1 -(5 -Chloropyridin-2-yl)-5-( { 3 -[ 1 -( 1 ,3 -thiazol-2- ylcarbonyl)piperidin-4-yl] azetidin- 1 -yl} carbonyl)- 1 H-indole. XH NMR (CDC13) δ = 8.51 (d, J=2.5 Hz, 1H), 8.23 (d, J=8.8 Hz, 1H),
7.95 (d, J=1.4 Hz, 1H), 7.87 (br. s., 1H), 7.81 (dd, J=8.7, 2.6 Hz, 1H), 7.70 (d, J=3.5 Hz, 1H), 7.59 (dd, J=8.7, 1.5 Hz, 1H), 7.53 (d,
93
J=3.2 Hz, 1H), 7.45 (d, J=8.7 Hz, 1H), 6.77 (dd, J=2.9, 0.6 Hz, 1H), 5.40-5.54 (m, 1H), 4.73 (t, J=9.8 Hz, 1H), 4.37-4.49 (m, 1H), 4.21- 4.32 (m, 1H), 4.03-4.12 (m, 1H), 3.93-4.03 (m, 1H), 3.57-3.72 (m, 1H), 3.06-3.22 (m, 1H), 2.36-2.51 (m, 1H), 1.67-1.94 (m, 3H),
1.10-1.32 (m, 2H). MS m/z 506 (M+H+).
Example 8
A. Methyl l-(3,4-difluorophenyl)-indole-5-carboxylate, 8b. A mixture of methyl indole-5-carboxylate 7a (2 g, 11.4 mmol), l-iodo-3,4-difluoro-benzene 8a (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 110 °C for 7 h. 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 3.0 g of compound 8b. MS m/z 288.1 (M+H+).
B. l-(3,4-Difluorophenyl)-indole-5-carboxylic acid, 8c. A mixture of methyl l-(3,4-difluorophenyl)-indole-5-carboxylate 8b (3.0 g, 10.4 mmol) and LiOH (1.0 g,
41.8 mmol) in THF (120 mL) and H20 (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 EtOAc (2x). The organic solution was washed with brine, dried over Na2S04 and concentrated to give 2.85 g of compound 8c. MS m/z 274.2 (M+H+).
C. l-(3,4-Difluorophenyl)-5-({3-[4-(l,3-thiazol-2-ylcarbonyl)piperazin-l- yl]azetidin-l-yl}carbonyl)-lH-indole, Cpd 32. To a stirring solution of compound lj (0.40 mmol, 100 mg) and compound 8c (0.44 mmol, 120 mg) in 4 mL of CH2CI2 was added Et3N (1.59 mmol, 0.22 mL). After 20 min at 20 °C, HATU (0.48 mmol, 180 mg) was added and the mixture was stirred at 20 °C for 20 h. The solvent was removed and the crude residue was purified by preparative reverse-phase chromatography to give 118 mg (58% yield) of Cpd 32. XH NMR (CD3OD, 400 MHz): δ = 8.00 (s, 1H), 7.93 (br. s., 1H), 7.81 (d, J=2.9 Hz, 1H), 7.51-7.60 (m, 4H), 7.44-7.52 (m, 1H), 7.33-7.42 (m, 1H), 6.79 (d, J=3.2 Hz, 1H), 5.22-5.38 (m, 1H), 4.55-4.71 (m, 1H), 4.48 (t, J=8.8 Hz, 1H), 4.14-4.33 (m, 2H), 3.89-4.04 (m, 1H), 3.14-3.28 (m, 1H), 2.83-2.99 (m, 1H), 2.43-2.58 (m, 1H), 1.70-2.00 (m, 3H), 1.09-1.32 (m, 2H). MS m/z 507.1 (M+H+).
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:
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 compounds of the present invention were prepared:
Cpd Name and data
l-(3,4-Difluorophenyi)-5-({4-[l-(lH-pyrrol-2-
80 ylcarbonyl)azetidin-3 -yl]piperidin- 1 -yl} carbonyl)- 1 H-indole.
MS m/z 489.2 (M+H+).
1 -(2-Methylpyridin-4-yl)-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-lH-indole. ¾ NMR (CDC13) δ = 8.63 (d, J=5.4 Hz, IH), 8.00 (s, IH), 7.87 (br. s., IH), 7.70 (d, J=8.7 Hz, IH), 7.61 (dd, J=8.7, 1.4 Hz, IH), 7.53 (d, J=3.1 Hz, IH), 7.44 (d, J=3.4 Hz, IH),
95
7.24-7.36 (m, 2H), 6.79 (d, J=3.3 Hz, IH), 5.50 (d, J=12.5 Hz, IH), 4.64-4.84 (m, IH), 4.37-4.53 (m, IH), 4.21-4.37 (m, IH), 3.91-4.14 (m, 2H), 3.06-3.25 (m, IH), 2.73-2.89 (m, IH), 2.67 (s, 3H), 2.46 (d, J=8.1 Hz, IH), 1.65-1.94 (m, 3H), 1.09-1.35 (m, 2H).
Example 9
A. Methyl 4-((4-fluorophenyl)amino)-3-nitrobenzoate, 9c. A mixture of methyl 4-fluoro-3-nitrobenzoate 9a (1 g, 5.02 mmol), 4-fluoroaniline 9b (4.34 mL, 5.02 mmol), and DIPEA (1.04 mL, 6.03 mmol) in DMF (10 mL) was stirred at room temperature for 2 h. Water was added to the mixture, the resulting solid was collected by filtration, washed with water, and dried. The crude compound 9c was used in the next reaction without further purification.
B. Methyl 3-amino-4-((4-fluorophenyl)amino)benzoate, 9d. A mixture of compound 9c (1.4 g, 4.8 mmol) and SnCl2.2H20 (4.9 g, 21.7 mmol) in EtOH (50 mL) was stirred at 80°C. After 4 h, the mixture was cooled to room temperature and was slowly added to saturated aqueous aHC03. The solid was collected by filtration and washed with H20. The solid was triturated with EtOAc and the filtrate was concentrated. The crude compound 9d was used in the next reaction without further purification. MS m/z 261.1 (M+H+).
C. Methyl l-(4-fluorophenyl)-lH-benzo[d]imidazole-5-carboxylate, 9e. A mixture of compound 9d (0.18 g, 0.693 mmol) and trimethyl orthoformate (0.7 mL,
6.39 mmol) in DMF (2 mL) was refluxed for 5 h and then cooled to room temperature. Water was added to the mixture. The resulting solid was collected by filtration, washed with water, and dried. The crude compound 9e was used in the next reaction without further purification. MS m/z 271.1 (M+H+).
D. l-(4-Fluorophenyl)-lH-benzo[d]imidazole-5-carboxylic acid, 9f. To a solution of compound 9e (0.18 g, 0.666 mmol) in EtOH (lOmL) was added IN aqueous NaOH (2.5 mL, 2.5 mmol). The mixture was stirred at room temperature for 4 d. The solvent was evaporated and IN aqueous HC1 was added, followed by extraction with EtOAc. The organic layer was dried over MgS04 and concentrated. The crude compound 9f was purified by preparative reverse phase chromatography. MS m/z 257 '.1 (M+H+).
E. l-(4-Fluorophenyl)-5-({3-[l-(l,3-thiazol-2-ylcarbonyl)piperidin-4- yl]azetidin-l-yl}carbonyl)-lH-benzimidazole, Cpd 81. To a solution of compound lj (0.058 g, 0.178 mmol) and HATU (0.081 g, 0.214 mmol) in 3 mL of CH2C12 was added Et3N (0.099 mL, 0.713 mmol). The mixture was stirred at 20 °C for 30 min, and then compound 9f (0.050g, 0.196 mmol) was added. The reaction mixture was stirred at 20 °C for 20 h. Water (6 mL) was added and the mixture was extracted with EtOAc. The organic layer was dried over MgS04 and concentrated. The crude product was purified by preparative reverse phase chromatography to give 46 mg (47% yield) of Cpd 81. ¾ NMR (CD3OD) δ = 8.14 (s, 1H), 7.93 (br. s., 1H), 7.78-7.84 (m, 2H), 7.67-7.78 (m, 3H), 7.45 (t, J=8.7 Hz, 2H), 5.23-5.38 (m, 1H), 4.57-4.71 (m, 1H), 4.44- 4.54 (m, 1H), 4.29 (t, J=9.7 Hz, 1H), 4.22 (br. s., 1H), 4.00 (br. s., 1H), 3.17-3.27 (m, 1H), 2.85-2.99 (m, 1H), 2.48-2.61 (m, 1H), 1.70-2.02 (m, 3H), 1.09-1.34 (m, 2H) MS m/z 490.2 (M+H+).
Following the procedure described above for Example 9 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared.
Following the procedure described above for Example 9 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared:
Cpd Name and data
1 -(3 ,4-Difluorophenyl)-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2-
82 ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-lH- benzimidazole. MS m/z 508.2 (M+H+).
5-({3-[l-( 1 ,3 -Thiazol-2-ylcarbonyl)piperidin-4-yl] azetidin- 1 -
83 yl} carbonyl)- 1 -[4-(trifluoromethyl)phenyl]- 1 H-benzimidazole.
MS m/z 540.2 (M+H+).
5-({3-[l-( 1 ,3 -Thiazol-2-ylcarbonyl)piperidin-4-yl] azetidin- 1 -
84 yl}carbonyl)-l-(2,2,2-trifluoroethyl)-lH-benzimidazole. MS m/z 478.2 (M+H+).
5-({3-[l-( 1 ,3 -Thiazol-2-ylcarbonyl)piperidin-4-yl] azetidin- 1 -
85 yl}carbonyl)-l-(3,3,3-trifluoropropyl)-lH-benzimidazole. MS m/z 492.1 (M+H+).
Example 10
A. Methyl 2-methyl-l-(4-fluorophenyl)-lH-benzo[d]imidazole-5- carboxylate, 10a. The title compound 10a was prepared using the method described Example 9, substituting trimethyl orthoacetate for trimethyl orthoformate in Step C. The crude compound 10a was used in the next reaction without further purification. MS m/z 285.1 (M+H+). B. 2-Methyl-l-(4-fluorophenyl)-lH-benzo[d]imidazole-5-carboxylate, 10b.
The title compound 10b was prepared using the method described in Example 9, substituting compound 10a for compound 9e in Step D. The crude product 10b was used in the next reaction without further purification. MS m/z 271.2 (M+H+).
C. l-(4-Fluorophenyl)-2-methyl-5-({3-[l-(l,3-thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-lH-benzimidazole, Cpd 87. The title compound Cpd 87 was prepared using the method described in Example 9, substituting compound 10b for compound 9f in Step E. The crude product was purified by preparative reverse phase chromatography to give 23 mg (50% yield) of Cpd 87. XH NMR (CD3OD) δ = 8.08 (s, 1H), 7.93 (br. s., 1H), 7.76-7.84 (m, 2H), 7.68 (dd, J=8.7, 4.5 Hz, 2H), 7.50 (t, J=8.6 Hz, 2H), 7.43 (d, J=8.8 Hz, 1H), 5.31 (br. s., 1H), 4.64 (t, J=13.0 Hz, 1H), 4.45 (t, J=8.4 Hz, 1H), 4.29 (t, J=9.7 Hz, 1H), 4.15-4.24 (m, 1H), 4.00 (br. s., 1H), 3.17-3.27 (m, 1H), 2.83-2.99 (m, 1H), 2.71 (s, 3H), 2.55 (q, J=7.7 Hz, 1H), 1.74-2.00 (m, 3H), 1.15-1.31 (m, 2H). MS m/z 504.0 (M+H+).
Following the procedure described above for Example 10 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following intermediate compounds were prepared:
10-11 10-T2 10-13 10-14 10-T5 10-16
Following the procedure described above for Example 10 and substituting the appropriate reagents, starting materials and purification methods known to those skilled in the art, the following compounds of the present invention were prepared: Cpd Name and data
2-Methyl- 1 -phenyl-5 -( {3 - [ 1 -( 1 ,3 -thiazol-2-ylcarbonyl)piperidin-4-
86 yl]azetidin-l-yl}carbonyl)-lH-benzimidazole. MS m/z 486.1 (M+H+).
1 -(3 ,4-Difluorophenyl)-2-methyl-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-lH-
88
benzimidazole.
MS m/z 522.0 (M+H+).
2-Methyl-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2-ylcarbonyl)piperidin-4- yljazetidin- 1 -yl} carbonyl)- 1 -[4-(trifluoromethyl)phenyl]- 1H-
89
benzimidazole.
MS m/z 554.1 (M+H+).
2-Methyl-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2-ylcarbonyl)piperidin-4- yl] azetidin- 1 -yl} carbonyl)- 1 -(2,2,2-trifluoroethyl)- 1 H-
90
benzimidazole.
MS m/z 492.1 (M+H+).
2-Methyl-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2-ylcarbonyl)piperidin-4- yl] azetidin- 1 -yl} carbonyl)- 1 -(3,3,3 -trifluoropropyl)- 1H-
91
benzimidazole.
MS m/z 506.2 (M+H+).
l-(4,4-Difluorocyclohexyl)-2-methyl-5-({3-[l-(l,3-thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-lH-
92
benzimidazole.
MS m/z 528.3 (M+H+).
Example 11
A. Methyl l-(4-fluorophenyl)-2-oxo-2,3-dihydro-lH-benzo[d]imidazole-5- carboxylate, 11a. A mixture of compound 9d (0.20 g, 0.826 mmol) and 1, 1 '- carbonyldiimidazole (0.535 g, 3.3 mmol) in DMF (8 mL) was heated at 90°C for 2 h. The solvent was removed and the residue was triturated with water (15 mL). The resulting precipitate was collected by filtration and washed several times with water. The crude product 11a was used in the next reaction without further purification. MS m/z 287.1 (M+H+).
B. l-(4-Fluorophenyl)-2-oxo-2,3-dihydro-lH-benzo[d]imidazole-5- carboxylate, lib. The title compound lib was prepared using the method described in Example 9, substituting compound 11a for compound 9e in Step D. The crude product lib was used in the next reaction without further purification. MS m/z 273.1 (M+H+).
C. l-(4-Fluorophenyl)-5-({3-[l-(l,3-thiazol-2-ylcarbonyl)piperidin-4- yl]azetidin-l-yl}carbonyl)-l,3-dihydro-2H-benzimidazol-2-one, Cpd 97. The title compound Cpd 97 was prepared using the method described in Example 9, substituting compound lib for compound 9f in Step E. The crude product was purified by preparative reverse phase chromatography to give 51 mg (32% yield) of Cpd 97. 1H MR (CD3OD) 8 = 7.93 (br. s., 1H), 7.81 (d, J=3.2 Hz, 1H), 7.51-7.59 (m, 2H), 7.45 (s, 1H), 7.41 (dd, J=8.3, 1.2 Hz, 1H), 7.33 (t, J=8.7 Hz, 2H), 7.04 (d, J=8.1 Hz, 1H), 5.30 (br. s., 1H), 4.56-4.69 (m, 1H), 4.40-4.51 (m, 1H), 4.21-4.29 (m, 1H), 4.18 (br. s., 1H), 3.95 (br. s., 1H), 3.15-3.27 (m, 1H), 2.83-2.99 (m, 1H), 2.43- 2.58 (m, 1H), 1.71-2.00 (m, 3H), 1.22 (br. s., 2H). MS m/z 506.1 (M+H+). 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.
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 compounds of the present invention were prepared.
Cpd Name and data
l-Phenyl-5-({3-[l-(l,3-thiazol-2-ylcarbonyl)piperidin-4-
96 yl]azetidin-l-yl} carbonyl)- 1, 3 -dihydro-2H-benzimidazol-2-one.
MS m/z 488.1 (M+H+).
1 -(3 ,4-Difluorophenyl)-5 -( { 3 - [ 1 -( 1 ,3 -thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-l,3-dihydro-2H-
98
benzimidazol-2-one.
MS m/z 524.1 (M+H+).
5 -( { 3 - [ 1 -( 1 ,3 -Thiazol-2-ylcarbonyl)piperidin-4-yl] azetidin- 1 - yl} carbonyl)- 1 -[4-(trifluoromethyl)phenyl]- 1 ,3 -dihydro-2H- benzimidazol-2-one.
XH NMR (CD3OD) δ = 7.84-7.97 (m, 3H), 7.74-7.83 (m, 3H),
99
7.38-7.48 (m, 2H), 7.19 (d, J=8.3 Hz, 1H), 5.22-5.36 (m, 1H), 4.56-4.69 (m, 1H), 4.39-4.53 (m, 1H), 4.11-4.30 (m, 2H), 3.90- 4.01 (m, 1H), 2.83-3.00 (m, 1H), 2.43-2.59 (m, 1H), 1.68-2.01 (m, 3H), 1.13-1.29 (m, 2H).
5 -( { 3 - [ 1 -( 1 ,3 -Thiazol-2-ylcarbonyl)piperidin-4-yl] azetidin- 1 - yl} carbonyl)- 1 -(3 ,3,3-trifluoropropyl)- 1 ,3 -dihydro-2H-
100
benzimidazol-2-one.
MS m/z 508.2 (M+H+).
1 -(4,4-Difluorocyclohexyl)-5-( {3 -[ 1 -( 1 ,3-thiazol-2- ylcarbonyl)piperidin-4-yl]azetidin-l-yl}carbonyl)-l,3-dihydro-2H-
101
benzimidazol-2-one.
MS m/z 530.2 (M+H+).
Example 12
A. 3-(Pyridin-4-yl)azetidine, 12a. Compound lc was dissolved in a mixture of 3N aqueous HC1 and THF and stirred until compound lc was completely consumed.
The mixture was concentrated under reduced pressure and the aqueous residue was lyophilized to give compound 12a as the di-hydrochloride salt, which was used in the next step without further purification.
B. [l,l'-Biphenyl]-4-yl-(3-(pyridin-4-yl)azetidin-l-yl)methanone, 12c.
Compound 12a (4.5 mmol, 928 mg), [l, l'-biphenyl]-4-carboxylic acid 12b (4.95 mmol, 980 mg), HBTU (6.43 mmol, 2.44 g), and DIEA (20.2 mmol, 3.49 mL) were combined in DMF and stirred at 20 °C for 20 h. The crude reaction mixture was purified by preparative reverse-phase HPLC to give compound 12c. 1H NMR (CD3OD, 400 MHz): δ = 8.68 (d, J=6.6 Hz, 2H), 7.92 (d, J=6.8 Hz, 2H), 7.68-7.77 (m, 2H), 7.60-7.68 (m, 2H), 7.57 (dd, J=8.3, 1.2 Hz, 2H), 7.38 (t, J=7.5 Hz, 2H), 7.23-7.34 (m, 1H), 4.56-4.68 (m, 1H), 4.43-4.56 (m, 1H), 4.12-4.34 (m, 2H).
C. [l,l'-Biphenyl]-4-yl-(3-(piperidin-4-yl)azetidin-l-yl)methanone, 12d. Compound 12c and 10% palladium on carbon were combined in a 1 :5 mixture of IN aqueous HC1 and ethanol in a Parr pressure bottle. The mixture was purged with 2 and then shaken under a 55 psi ¾ atmosphere at 20 °C. When the reaction was complete, the catalyst was removed by filtration through a diatomaceous earth pad. The filtrate was concentrated and lyophilized to provide compound 12d in quantitative yield as the hydrochloride salt.
D. 4-[l-(Biphenyl-4-ylcarbonyl)azetidin-3-yl]-l-(l,3-thiazol-2- ylcarbonyl)piperidine, Cpd 2. A solution of compound 12d HC1 salt (0.28 mmol, 98 mg), thiazole-2-carboxylic acid lh (0.33 mmol, 43 mg), and HBTU (0.33 mmol, 126 mg) in 2.5 mL of DMF was stirred for 10 min. DIEA (1.1 mmol, 0.2 mL) was added and the mixture was stirred at 20 °C for 20 h. The crude reaction mixture was purified by preparative reverse-phase HPLC to give Cpd 2. XH NMR (CD3OD, 400 MHz): δ = 7.84 (br. s., 1H), 7.69-7.75 (m, 1H), 7.60-7.69 (m, 4H), 7.53-7.60 (m, 2H), 7.37 (t, J=7.5 Hz, 2H), 7.24-7.33 (m, 1H), 5.13-5.29 (m, 1H), 4.54 (t, J=11.6 Hz, 1H), 4.38 (t, J=8.7 Hz, 1H), 4.04-4.22 (m, 2H), 3.80-3.94 (m, 1H), 3.07-3.18 (m, 1H), 2.73-2.91 (m, 1H), 2.35-2.50 (m, 1H), 1.63-1.91 (m, 3H), 0.99-1.25 (m, 2H). MS m/z 432.0 (M+H+).
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 compounds of the present invention were prepared:
Example 13
A. (4-(l-Benzhydrylazetidin-3-yl)-4-hydroxypiperidin-l- yl)(phenyl)methanone, 13c. A solution of l-benzhydryl-3-iodoazetidine 13a (1.4 mmol, 490 mg) in 5 mL of THF was added to a stirring mixture of Sml2 (0.1 M THF solution, 3 mmol, 30 mL) and 1.7 mL of HMPA. After 5 min, a solution of 1- benzoylpiperidin-4-one 13b (3.1 mmol, 626 mg) in 5 mL of THF was added. The reaction mixture was stirred for 2 h. Saturated aqueous NH4C1 solution (20 mL) was added and the suspension was filtered through a diatomaceous earth pad. The solids were washed with chloroform and the combined organic layers were washed with brine, dried, and concentrated. The crude residue was purified by preparative reverse- phase chromatography to give 400 mg (55% yield) of compound 13c (mono-TFA salt) as a yellow oil. MS m/z 401.2 (M+H+).
B. (4-(Azetidin-3-yl)-4-hydroxypiperidin-l-yl)(phenyl)methanone, 13d.
Compound 13c mono-TFA salt (0.34 mmol, 180 mg) and 10% palladium on carbon (40 mg) were combined in a 30 mL of ethanol in a Parr pressure bottle. The mixture was purged with 2 and then shaken under a 50 psi EL atmosphere at 20 °C for 20 h. The catalyst was removed by filtration through a diatomaceous earth pad and the filtrate was concentrated. Water, CH2CI2, and aqueous HC1 were added and the mixture was frozen and lyophilized to give 82 mg (81% yield) of compound 13d (mono-HCl salt). MS m/z 261.1 (M+H+).
C. l-(Phenylcarbonyl)-4-(l-{[5-(trifluoromethyl)-l-benzothiophen-2- yl]carbonyl}azetidin-3-yl)piperidin-4-ol, Cpd 6. A solution of 5- (trifluoromethyl)benzo[b]thiophene-2-carboxylic acid 13e (0.30 mmol, 75 mg), DIEA (0.83 mmol, 0.15 mL), and HBTU (0.33 mmol, 126 mg) in 2 mL of DMF was stirred for 10 min. The HC1 salt of compound 13d (0.28 mmol, 82 mg) was added and the mixture was stirred at 20 °C for 20 h. The reaction mixture was filtered through 3 g of silica gel carbonate and 3 g of aminopropyl silica gel using CH3CN as eluant. The CH3CN was removed under reduced pressure and the residue was purified by preparative reverse-phase HPLC to give 45 mg (33% yield) of Cpd 6. XH NMR (CD3OD, 400 MHz): δ = 8.13-8.24 (m, 1H), 8.04 (d, J=8.6 Hz, 1H), 7.82-7.90 (m, 1H), 7.60 (d, J=8.6 Hz, 1H), 7.27-7.42 (m, 5H), 4.44-4.59 (m, 2H), 4.25-4.39 (m, J=15.0, 2.8 Hz, 1H), 3.98-4.19 (m, 2H), 3.42-3.54 (m, 1H), 3.32-3.42 (m, 1H), 2.79 (quin, J=7.4 Hz, 1H), 1.57-1.69 (m, 1H), 1.31-1.56 (m, 3H). MS m/z 489.1 (M+H+).
Example 14
A. (tert-Butyl 4-(l-benzhydrylazetidin-3-yl)-4-hydroxypiperidine-l- carboxylate, 14b. A solution of l-benzhydryl-3-iodoazetidine 13a (4.75 mmol, 1.66 g) in 10 mL of THF was added to a stirring mixture of Sml2 (0.1 M THF solution, 9.98 mmol, 99.8 mL) and 5.6 mL of HMPA. After 15 min, a solution of tert-butyl 4- oxopiperidine-1 -carboxylate 14a (4.75 mmol, 0.95 g) in 15 mL of THF was added. The reaction mixture was stirred for 18 h. Saturated aqueous NH4CI solution (40 mL) was added and the suspension was filtered through a diatomaceous earth pad. The solids were washed with chloroform and the combined organic layers were washed with brine, dried, and concentrated. The crude residue was purified by preparative reverse-phase chromatography to give 1.0 g (45% yield) of 90% pure compound 14b as a clear oil. MS m/z 423.3 (M+H+).
B. 4-(l-Benzhydrylazetidin-3-yl)piperidin-4-ol, 14c. Compound 14c was prepared according to the procedure described in Example 1, Step F, substituting compound 14b for compound li.
C. (4-(l-Benzhydi lazetidin-3-yl)-4-hydroxypiperidin-l-yl)(thiazol-2- yl)methanone, 14d. A solution of thiazole-2-carboxylic acid lh (1.79 mmol, 232 mg), DIEA (6.53 mmol, 1.12 mL), and HBTU (1.96 mmol, 742 mg) in 1 1 mL of DMF was stirred for 10 min. Compound 14c (1.63 mmol, 526 mg) was added and the mixture was stirred at 20 °C for 20 h. The reaction mixture was purified by preparative reverse- phase HPLC to give 298 mg (33% yield) of compound 14d as a buff powder. MS m/z 434.2 (M+H+).
D. 4 (4-(Azetidin-3-yl)-4-hydroxypiperidin-l-yl)(thiazol-2-yl)methanone, 14e. 1 -Chloroethyl chloroformate (7.06 mmol, 0.76 mL) was added dropwise to a solution of compound 14d in 20 mL of CH2CI2 in an ice-water bath. After 30 min, the cooling bath was removed and the reaction mixture was stirred for 2 h at 20 °C. The CH2CI2 was evaporated, 20 mL of MeOH was added, and the resulting solution was refluxed for 2 h. The solvent was evaporated and the residue was partitioned between CH2CI2 and 0.2 N aqueous HC1. The aqueous layer was lyophilized to give compound 14e, which was carried on to the next step without purification. MS m/z 267.7 (M+H+).
E. l-(l,3-Thiazol-2-ylcarbonyl)-4-(l-{[6-(trifluoromethyl)-l- benzothiophen-2-yl]carbonyl}azetidin-3-yl)piperidin-4-ol, Cpd 94. A solution of 6- (trifluoromethyl)benzo[b]thiophene-2-carboxylic acid 14f (0.30 mmol, 75 mg), DIEA (0.98 mmol, 0.17 mL), and HBTU (0.30 mmol, 112 mg) in 2 mL of DMF was stirred for 10 min. Compound 14e (0.25 mmol, 66 mg) was added and the mixture was stirred at 20 °C for 20 h. The reaction mixture was purified by preparative reverse-phase HPLC to give 77 mg (62% yield) of Cpd 94 as a white powder. XH NMR (DMSO-d6): δ = 8.39 (s, 1H), 8.29 (d, J=8.6 Hz, 1H), 8.02-8.08 (m, 1H), 8.00 (s, 2H), 7.76 (dd, J=8.6, 1.5 Hz, 1H), 4.99 (d, J=12.7 Hz, 1H), 4.46-4.56 (m, 2H), 4.25 (d, J=12.2 Hz, 1H), 4.06-4.15 (m, 1H), 3.97-4.06 (m, 1H), 3.17-3.27 (m, 2H), 2.74-2.86 (m, J=14.8, 7.3, 7.3 Hz, 1H), 1.36-1.66 (m, 4H). MS m/z 496.1 (M+H+).
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 L179S L186S) or purified wild type MGL (wt-MGLL 6H-11-313) were diluted separately into 20 mM PIPES buffer (pH = 7.0), containing 150 mM NaCl and 0.001% Tween 20. Compounds of Formula (I) were pre-dispensed (50 nL) into the assay plate using a Cartesian Hummingbird prior to adding 4MU-B (25 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 (I) 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
Cpd MGL mutant MGL wild type
inh IC50 (μΜ ) inh IC50 (μΜ )
1 0.079 0.258
2 <0.005 <0.005
3 0.0339
4 0.154
5 0.0876
6 0.918
7 0.0565
8 0.007 Cpd MGL mutant MGL wild type inh IC50 (μΜ ) inh IC50 (μΜ )
9 0.1 14
10 0.0155
11 0.498
12 0.01
13 <0.005
14 0.005
15 0.007
16 0.007
17 0.0173
18 0.0125
19 <0.005
20 <0.005
21 0.0124
22 0.008
23 0.368
24 0.0379
25 0.0120
26 <0.005
27 0.00900
28 0.0164
29 0.00600
30 <0.005
31 <0.005
32 <0.005
33 <0.005
34 0.0544
35 <0.005
36 <0.005
37 <0.005
38 0.005
39 <0.005
40 0.00900
41 <0.005
42 <0.005
43 <0.005
44 <0.005
45 <0.005
46 0.1 11
47 0.0379
48 <0.005
49 0.00600
50 <0.005
51 <0.005
52 0.0715
53 0.0466
54 <0.005 Cpd MGL mutant MGL wild type inh IC50 (μΜ ) inh IC50 (μΜ )
55 0.0130
56 <0.005
57 0.0170
58 <0.005
59 0.011
60 0.01
61 0.211
62 <0.005
63 <0.005
64 <0.005
65 <0.005
66 0.0192
67 0.0665
68 0.0621
69 <0.005
70 0.008
71 0.0120
72 0.0170
73 0.0120
74 0.0138
75 <0.005
76 0.0114
77 <0.005
78 <0.005
79 0.00900
80 <0.005
81 0.0232
82 0.0355
83 0.00916
84 0.175
85 0.280
86 0.0187
87 0.0316
88 0.00949
89 0.0341
90 0.173
91 0.237
92 0.163
93 <0.005
94 0.01
95 0.0140
96 0.0781
97 0.0340
98 0.0166
99 0.0173
100 0.101 Cpd MGL mutant MGL wild type
inh IC50 (μΜ ) inh IC50 (μΜ )
101 0.104
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 (I) (10 μΜ) were pre-incubated with rat brain homogenate (50 mg). After a 15-min incubation time at 37°C, CaCL: (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
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 μί. 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 Tm l.
1. 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.
2. Matulis, D., Kranz, J. K., Salemme, F. R., and Todd, M. J. (2005) Biochemistry
44, 5258-66.
The Ka values for compounds of Formula (I) 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
Cpd MGL mutant MGL mutant ThermoFluor Kd (μΜ ) ThermoFluor qKd
(μΜ) (using qNMR cone.)
14 0.0050
15 0.0090
16 0.0020
17 0.0990
18 0.0030
19 0.0090
20 0.0010 0.0013
21 0.0286
22 0.0066
23 0.500
24 0.0199
25 0.0275
26 0.0036
27 0.0133
28 0.01 11
29 0.0003 0.0012
30 0.0025
31 0.0026
32 0.0006
33 0.0626
34 0.198
35 0.0025 0.0057
36 0.0225
37 0.0160
38 0.0155
39 0.0489
40 0.0946
41 0.0041 0.0031
42 0.0245
43 0.0264
44 0.0213
45 0.0122
46 0.0200
47 0.0767 0.0333
48 0.0010
49 0.0010
50 0.0010
51 0.0180
52 0.0304 0.0322
53 0.0333
54 0.0018
55 0.0020
56 0.0055 0.0033 Cpd MGL mutant MGL mutant ThermoFluor Kd (μΜ ) ThermoFluor qKd
(μΜ) (using qNMR cone.)
57 0.0059
58 0.0069
59 0.0202 0.0094
60 0.0008 0.0005
61 0.0006 0.0005
62 0.0013 0.0006
63 0.0174
64 0.0189
65 0.0404
66 0.0067
67 0.0066
68 0.0145
69 0.0012
70 0.0006
71 0.0012
72 0.0027
73 0.0048
74 0.0080
75 0.0039
76 0.0083
77 0.0050
78 0.0023
79 0.0033
80 0.0049
81 0.0148
82 0.0228
83 0.0136
84 0.0663
85 0.0645
86 0.0106
87 0.0075
88 0.0133
89 0.0077
90 0.0237
91 0.0325
92 0.0034
93 0.0055
94 0.0249
95 0.0386
96 0.0247
97 0.0221
98 0.0436
99 0.0167 Cpd MGL mutant MGL mutant
ThermoFluor Kd (μΜ ) ThermoFluor qKd
(μΜ) (using qNMR
cone.)
100 0.0888
101 0.0039
In Vivo Methods
Example 4
CFA-Induced Paw Radiant Heat Hypersensitivity
Each rat was 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) was focused through the glass onto the plantar surface of each hind paw in turn. The thermal stimulus was automatically shut off by a photoelectric relay when the paw was moved or when the cut-off time was reached (20 sec for radiant heat at ~5 amps). An initial (baseline) response latency to the thermal stimulus was 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 was re-evaluated and compared to the animal's baseline response time. Only rats that exhibited at least a 25% reduction in response latency (i.e., are hyperalgesic) were included in further analysis. Immediately following the post-CFA latency assessment, the indicated test compound or vehicle was administered orally. Post- compound treatment withdrawal latency was assessed at fixed time intervals, typically 30, 60, 120, 180, and 300 min.
The percent reversal (%R) of hypersensitivity was 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 was 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 are 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.
Biological DataTable 4
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

A compound of Formula (I)
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is
i) C6-10 aryl is unsubstituted or substituted with a substituent selected from the group consisting of fluoro, chloro, Ci-4alkyl, Ci-4alkoxy, cyano, and trifluoromethyl; or
ii) an unsubstituted heteroaryl selected from the group consisting of
thiazolyl, isothiazolyl, and lH-pyrrolyl;
Group b) is selected from the group consisting of
i) Ce-io aryl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzothiazolyl, benzimidazolyl, benzothienyl, indazolyl, and indolyl; iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; and iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of bromo, chloro, fluoro, iodo, Ci-4alkyl, d^alkoxy, and ¾,;
provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, cyano, and fluoro; R is hydrogen or hydroxy;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
2. The compound of claim 1 wherein Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl.
3. The compound of claim 1 wherein Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, lH-pyrrol-2-yl, and lH-pyrrol-3-yl.
4. The compound of claim 1 wherein Group b) is selected from the group consisting of
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; and iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of Rb are unsubstituted or substituted with one or two substitutents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro.
5. The compound of claim 1 wherein Group b) is delected from the group consisting of
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; and
iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is and
Rb is selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro.
6. The compound of claim 1 wherein R is hydrogen.
7. A compound of Formul
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and IH-pyrrolyl;
Group b) is selected from the group consisting of
phenyl;
a heteroaryl selected from the group consisting of benzoxazolyl, benzimidazolyl, benzothienyl, and indolyl; iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl or fluoro; and iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoropropyl, 4,4-difluorocyclohexyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substitutents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
8. A compound of Formul
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is an unsubstituted phenyl or unsubstituted heteroaryl selected from the group consisting of thiazolyl, isothiazolyl, and lH-pyrrolyl;
Group b) is selected from the group consisting of
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; and iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl;
wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents independently selected from the group trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen or hydroxy;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
9. A compound of Formul
Formula (I)
wherein
Y and Z are independently selected from Group a) or Group b) such that one of Y and Z is Group a) and the other is Group b);
Group a) is an unsubstituted phenyl or an unsubstituted heteroaryl selected from the group consisting of thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, isothiazolyl, lH-pyrrol-2-yl, and lH-pyrrol-3-yl;
Group b) is selected from the group consisting of
i) phenyl;
ii) a heteroaryl selected from the group consisting of benzoxazolyl,
benzimidazolyl, benzothienyl, and indolyl;
iii) phenylmethyl-phenyl wherein the phenyl group of phenylmethyl is unsubstituted or substituted with trifluoromethyl; and
iv) 1 ,3 -dihydro-3 H-benzimidazol-2-on-yl; wherein Group b) other than phenylmethyl-phenyl is unsubstituted or substituted with one or two substitutents independently selected from the group consisting of chloro, fluoro, methyl, and ¾; provided that no more than one substituent is ¾; and
Rb is selected from the group consisting of trifluoromethyl, thienyl, pyridinyl, and phenyl; wherein said thienyl, pyridinyl, and phenyl of ¾ are unsubstituted or substituted with one or two substituents independently selected from the group consisting of trifluoromethyl, methyl, chloro, and fluoro;
R is hydrogen or hydroxy;
and enantiomers, diastereomers, and pharmaceutically acceptable salts thereof.
A compound of Formul
Formula (I)
selected from the group consisting of
the compound wherein Y is thiazol-4-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is isothiazol-5-yl, Z is biphenyl-4-yl, and R is H; the compound wherein Y is lH-pyrrol-3-yl, Z is biphenyl-4-yl, and R is H; the compound wherein Y is thiazol-5-yl, Z is biphenyl-4-yl, and R is H;
the compound wherein Y is phenyl, Z is 5-trifluoromethyl-benzothien-2-yl, and R is OH;
the compound wherein Y is thiazol-4-yl, Z is 3-chloro-6-fluoro-benzothien-2-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-fluoro-benzothien-2-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 4-(3-trifluoromethylphenyl)- phenyl, and R is H; the compound wherein Y is thiazol-4-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 4-(5-trifluoromethylthien-2-yl)- phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 4-(3- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 3-methyl-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 4-(3-trifluoromethylphenyl)- phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 4-(5-trifluoromethylthien-2-yl)- phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 4-(3- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 3-methyl-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 2-fluoro-4-phenyl-phenyl, and
R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 4-(3-trifluoromethylphenyl)- phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 3-(3-fluorophenyl)-phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 4-(5-trifluoromethylthien-2- yl)-phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 4-(3- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 3-methyl-6-trifluoromethyl- benzothien-2-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is 4-(4- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 2-phenyl-benzoxazol-6- yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-lH- indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-lH-indol-
5-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 4-(4- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 2-phenyl-benzoxazol-6- yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(4-fluorophenyl)-lH-indol-5-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(4-trifluoromethylphenyl)-lH- indol-5-yl, and R is H;
the compound wherein Y is thiazol-4-yl, Z is l-(3,4-difluorophenyl)-lH-indol-
5-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 4-(4- trifluoromethylphenylmethyl)-phenyl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 2-phenyl-benzoxazol-6- yl, and
R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 3-chloro-6-trifluoromethyl- benzothien-2-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is l-(4-fluorophenyl)-lH-indol-5- yl, and R is H; the compound wherein Y is lH-pyrrol-2-yl, Z is 1 -(4-trifluoromethylphenyl)- lH-indol-5-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is l-(3,4-difluorophenyl)-lH- indol-5-yl, and R is H;
the compound wherein Y is 2-fluoro-4-phenyl-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 2-fluoro-4-phenyl-phenyl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is lH-pyrrol-2-yl, Z is 2-fluoro-4-phenyl-phenyl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenyl)-phenyl, Z is thiazol-2- yl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenyl)-phenyl, Z is thiazol-4- yl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenyl)-phenyl, Z is lH-pyrrol- 2-yl, and R is H;
the compound wherein Y is 3-(3-fluorophenyl)-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 3-(3-fluorophenyl)-phenyl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 3-(3-fluorophenyl)-phenyl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is 4-(5-trifluoromethyl-thien-2-yl)-phenyl, Z is
thiazol-2-yl, and R is H;
the compound wherein Y is 4-(5-trifluoromethyl-thien-2-yl)-phenyl, Z is
thiazol-4-yl, and R is H;
the compound wherein Y is 4-(5-trifluoromethyl-thien-2-yl)-phenyl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 4-(3-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-4-yl, and R is H; the compound wherein Y is 4-(3-trifluoromethylphenylmethyl)-phenyl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 3-methyl-6-trifluoromethyl-benzothien-2-yl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 4-(4-trifluoromethylphenylmethyl)-phenyl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 2-phenyl-benzoxazol-6-yl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is 3-chloro-6-trifluoromethyl-benzothien-2-yl, Z is lH-pyrrol-2-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is l-(4-fluorophenyl)-lH-indol-5-yl, Z is lH-pyrrol-2- yl, and R is H; the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is thiazol-2-yl, and R is H;
the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is thiazol-4-yl, and R is H;
the compound wherein Y is l-(4-trifluoromethylphenyl)-lH-indol-5-yl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is thiazol-
2-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is thiazol- 4-yl, and R is H;
the compound wherein Y is l-(3,4-difluorophenyl)-lH-indol-5-yl, Z is 1H- pyrrol-2-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(2,2,2-trifluoroethyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-phenyl-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-2-methyl-lH- benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-2-methyl- lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-2- methyl-lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(2,2,2-trifluoroethyl)-2-methyl- lH-benzimidazol-5-yl, and R is H; the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-2- methyl-lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4,4-difluorocyclohexyl)-2- methyl-lH-benzimidazol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(5-chloropyridin-2-yl)-lH- indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is 6-trifluoromethyl-benzothien-2-yl, and R is OH;
the compound wherein Y is thiazol-2-yl, Z is l-(2-methylpyridin-4-yl)-lH- indol-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-phenyl-l,3-dihydro-3H- benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-fluorophenyl)-l,3-dihydro-
3H-benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,4-difluorophenyl)-l,3- dihydro-3H-benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4-trifluoromethylphenyl)-l,3- dihydro-3H-benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(3,3,3-trifluoropropyl)-l,3- dihydro-3H-benzimidazol-2-on-5-yl, and R is H;
the compound wherein Y is thiazol-2-yl, Z is l-(4,4-difluorocyclohexyl)-l,3- dihydro-3H-benzimidazol-2-on-5-yl, and R is H; and pharmaceutically acceptable salt forms thereof.
1 1. A pharmaceutical composition comprising the compound of claim 1 or 10 and a member selected from the group consisting of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient, and a pharmaceutically acceptable diluent.
12. A pharmaceutical composition of claim 1 1, wherein the composition is a solid oral dosage form.
13. A pharmaceutical composition of claim 1 1, wherein the composition is selected from the group consisting of a syrup, an elixir, and a suspension.
14. 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 10 to the subject.
15. The method of claim 14 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.
16. A compound that is Fo
EP11784828.3A 2010-10-22 2011-10-20 Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors Withdrawn EP2629851A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US40587610P 2010-10-22 2010-10-22
PCT/US2011/057085 WO2012054716A1 (en) 2010-10-22 2011-10-20 Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors

Publications (1)

Publication Number Publication Date
EP2629851A1 true EP2629851A1 (en) 2013-08-28

Family

ID=44993878

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11784828.3A Withdrawn EP2629851A1 (en) 2010-10-22 2011-10-20 Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors

Country Status (12)

Country Link
EP (1) EP2629851A1 (en)
JP (1) JP2013540159A (en)
KR (1) KR20130142137A (en)
CN (1) CN103260703A (en)
AR (1) AR083542A1 (en)
AU (1) AU2011316970A1 (en)
BR (1) BR112013009858A2 (en)
CA (1) CA2815350A1 (en)
IL (1) IL225769A0 (en)
RU (1) RU2013123275A (en)
TW (1) TW201305136A (en)
WO (1) WO2012054716A1 (en)

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201524952A (en) * 2013-03-15 2015-07-01 Araxes Pharma Llc Covalent inhibitors of KRAS G12C
US9745319B2 (en) 2013-03-15 2017-08-29 Araxes Pharma Llc Irreversible covalent inhibitors of the GTPase K-Ras G12C
UY35464A (en) 2013-03-15 2014-10-31 Araxes Pharma Llc KRAS G12C COVALENT INHIBITORS.
JO3805B1 (en) 2013-10-10 2021-01-31 Araxes Pharma Llc Inhibitors of kras g12c
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
JO3556B1 (en) 2014-09-18 2020-07-05 Araxes Pharma Llc Combination therapies for treatment of cancer
ES2826443T3 (en) 2014-09-25 2021-05-18 Araxes Pharma Llc KRAS G12C Mutant Protein Inhibitors
WO2016049568A1 (en) 2014-09-25 2016-03-31 Araxes Pharma Llc Methods and compositions for inhibition of ras
EP3279191B1 (en) 2015-03-30 2020-09-02 Takeda Pharmaceutical Company Limited Heterocyclic compound
TW201702232A (en) 2015-04-10 2017-01-16 亞瑞克西斯製藥公司 Substituted quinazoline compounds and methods of use thereof
CA2982360A1 (en) 2015-04-15 2016-10-20 Liansheng Li Fused-tricyclic inhibitors of kras and methods of use thereof
US10144724B2 (en) 2015-07-22 2018-12-04 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10975071B2 (en) 2015-09-28 2021-04-13 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10875842B2 (en) 2015-09-28 2020-12-29 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2017058768A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
WO2017058792A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
WO2017058805A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
EP3356339A1 (en) 2015-09-28 2018-08-08 Araxes Pharma LLC Inhibitors of kras g12c mutant proteins
WO2017058728A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
WO2017070256A2 (en) 2015-10-19 2017-04-27 Araxes Pharma Llc Method for screening inhibitors of ras
TW201726656A (en) 2015-11-16 2017-08-01 亞瑞克西斯製藥公司 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
WO2017100546A1 (en) 2015-12-09 2017-06-15 Araxes Pharma Llc Methods for preparation of quinazoline derivatives
US10463753B2 (en) 2016-02-19 2019-11-05 Lundbeck La Jolla Research Center, Inc. Radiolabeled monoacylglycerol lipase occupancy probe
WO2017172979A1 (en) 2016-03-30 2017-10-05 Araxes Pharma Llc Substituted quinazoline compounds and methods of use
EA036637B1 (en) 2016-03-31 2020-12-02 Такеда Фармасьютикал Компани Лимитед Heterocyclic spiro compounds inhibiting monoacylglycerol lipase
JP6832342B2 (en) 2016-03-31 2021-02-24 武田薬品工業株式会社 Heterocyclic compound
US10646488B2 (en) 2016-07-13 2020-05-12 Araxes Pharma Llc Conjugates of cereblon binding compounds and G12C mutant KRAS, HRAS or NRAS protein modulating compounds and methods of use thereof
WO2018064510A1 (en) 2016-09-29 2018-04-05 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
WO2018068017A1 (en) 2016-10-07 2018-04-12 Araxes Pharma Llc Heterocyclic compounds as inhibitors of ras and methods of use thereof
US11279689B2 (en) 2017-01-26 2022-03-22 Araxes Pharma Llc 1-(3-(6-(3-hydroxynaphthalen-1-yl)benzofuran-2-yl)azetidin-1 yl)prop-2-en-1-one derivatives and similar compounds as KRAS G12C modulators for treating cancer
EP3573970A1 (en) 2017-01-26 2019-12-04 Araxes Pharma LLC 1-(6-(3-hydroxynaphthalen-1-yl)quinazolin-2-yl)azetidin-1-yl)prop-2-en-1-one derivatives and similar compounds as kras g12c inhibitors for the treatment of cancer
EP3573964A1 (en) 2017-01-26 2019-12-04 Araxes Pharma LLC Benzothiophene and benzothiazole compounds and methods of use thereof
US11274093B2 (en) 2017-01-26 2022-03-15 Araxes Pharma Llc Fused bicyclic benzoheteroaromatic compounds and methods of use thereof
JP7327802B2 (en) 2017-01-26 2023-08-16 アラクセス ファーマ エルエルシー Fused hetero-heterobicyclic compounds and methods of use thereof
JP2020521741A (en) 2017-05-25 2020-07-27 アラクセス ファーマ エルエルシー Compounds for the treatment of cancer and methods of their use
BR112019024674A2 (en) 2017-05-25 2020-06-16 Araxes Pharma Llc COVALENT KRAS INHIBITORS
EP3630747A1 (en) 2017-05-25 2020-04-08 Araxes Pharma LLC Quinazoline derivatives as modulators of mutant kras, hras or nras
AU2018338855B2 (en) 2017-09-29 2022-04-28 Takeda Pharmaceutical Company Limited Heterocyclic compound
CA3111309A1 (en) 2018-09-28 2020-04-02 Janssen Pharmaceutica Nv Monoacylglycerol lipase modulators
CN109879797A (en) * 2019-01-10 2019-06-14 安徽昊帆生物有限公司 N- benzyl-tetrahydropyridines and preparation method thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9812037D0 (en) * 1998-06-04 1998-07-29 Pfizer Ltd Piperidones
US8106208B2 (en) * 2006-05-18 2012-01-31 Albireo Ab Benzamide compounds that act as NK receptor antagonists
FR2915198B1 (en) * 2007-04-18 2009-12-18 Sanofi Aventis TRIAZOLOPYRIDINE CARBOXAMIDE AND TRIAZOLOPYRIDINE -CARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE.
CA2722449C (en) * 2008-04-25 2016-11-08 Carsten Schubert Crystal structure of monoacylglycerol lipase (mgll)
ES2775000T3 (en) * 2009-04-02 2020-07-23 Merck Patent Gmbh Piperidine and piperazine derivatives as autotaxin inhibitors
BRPI1014876A2 (en) * 2009-04-22 2016-04-12 Janssen Pharmaceutica Nv "Azetidinyl diamides as monoacylglycerol lipase inhibitors."

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2012054716A1 *

Also Published As

Publication number Publication date
IL225769A0 (en) 2013-06-27
WO2012054716A1 (en) 2012-04-26
AR083542A1 (en) 2013-03-06
AU2011316970A1 (en) 2013-05-09
BR112013009858A2 (en) 2016-07-26
JP2013540159A (en) 2013-10-31
TW201305136A (en) 2013-02-01
RU2013123275A (en) 2014-11-27
KR20130142137A (en) 2013-12-27
CN103260703A (en) 2013-08-21
CA2815350A1 (en) 2012-04-26

Similar Documents

Publication Publication Date Title
EP2629851A1 (en) Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors
EP2611774B1 (en) Di-azetidinyl diamide as monoacylglycerol lipase inhibitors
US8513423B2 (en) Piperidin-4-yl-azetidine diamides as monoacylglycerol lipase inhibitors
AU2011307252B2 (en) Oxopiperazine-azetidine amides and oxodiazepine-azetidine amides as monoacylglycerol lipase inhibitors
US8415341B2 (en) Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitors
US8748417B2 (en) Amino-pyrrolidine-azetidine diamides as monoacylglycerol lipase inhibitors
RU2558141C2 (en) Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitors
KR20120034617A (en) Azetidinyl diamides as monoacylglycerol lipase inhibitors
WO2010124122A1 (en) Heteroaromatic and aromatic piperazinyl azetidinyl amides as monoacylglycerol lipase inhibitors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130515

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 409/14 20060101ALI20140226BHEP

Ipc: C07D 413/14 20060101ALI20140226BHEP

Ipc: C07D 401/14 20060101ALI20140226BHEP

Ipc: C07D 205/04 20060101ALI20140226BHEP

Ipc: C07D 211/16 20060101ALI20140226BHEP

Ipc: A61K 31/4523 20060101ALI20140226BHEP

Ipc: C07D 417/12 20060101ALI20140226BHEP

Ipc: A61P 29/00 20060101AFI20140226BHEP

Ipc: C07D 401/04 20060101ALI20140226BHEP

Ipc: C07D 417/14 20060101ALI20140226BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140505

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150410

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20150821