US20200308190A1 - Heterocyclic compounds as monoacylglycerol lipase inhibitors - Google Patents

Heterocyclic compounds as monoacylglycerol lipase inhibitors Download PDF

Info

Publication number
US20200308190A1
US20200308190A1 US16/844,262 US202016844262A US2020308190A1 US 20200308190 A1 US20200308190 A1 US 20200308190A1 US 202016844262 A US202016844262 A US 202016844262A US 2020308190 A1 US2020308190 A1 US 2020308190A1
Authority
US
United States
Prior art keywords
compound
carbonyl
oxazin
alkyl
hydrogen
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.)
Abandoned
Application number
US16/844,262
Inventor
Charles Bell
Joerg Benz
Luca Gobbi
Uwe Grether
Katrin Groebke Zbinden
Benoit Hornsperger
Buelent Kocer
Carsten Kroll
Bernd Kuhn
Marius Daniel Rinaldo LUTZ
Fionn O'Hara
Hans Richter
Martin Ritter
Didier Rombach
Martin KURATLI
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.)
F Hoffmann La Roche AG
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Publication of US20200308190A1 publication Critical patent/US20200308190A1/en
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KURATLI, Martin
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BELL, CHARLES, BENZ, JOERG, GOBBI, LUCA, GRETHER, UWE, GROEBKE ZBINDEN, KATRIN, HORNSPERGER, BENOIT, KOCER, BUELENT, KROLL, Carsten, KUHN, BERND, LUTZ, Marius Daniel Rinaldo, O'HARA, FIONN, RICHTER, HANS, RITTER, MARTIN, ROMBACH, DIDIER
Priority to US17/749,496 priority Critical patent/US11802133B2/en
Priority to US18/475,065 priority patent/US20240150373A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to organic compounds useful for therapy or prophylaxis in a mammal, and in particular to monoacylglycerol lipase (MAGL) inhibitors for the treatment or to prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer, mental disorders, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine and/or depression in a mammal.
  • MLM monoacylglycerol lipase
  • Endocannabinoids are signaling lipids that exert their biological actions by interacting with cannabinoid receptors (CBRs), CB1 and CB2. They modulate multiple physiological processes including neuroinflammation, neurodegeneration and tissue regeneration (Iannotti, F. A., et al., Progress in lipid research 2016, 62, 107-28).
  • CBRs cannabinoid receptors
  • CB1 and CB2 cannabinoid receptors
  • DAGL diacyglycerol lipases
  • MAGL monoacylglycerol lipase
  • MAGL is expressed throughout the brain and in most brain cell types, including neurons, astrocytes, oligodendrocytes and microglia cells (Chanda, P. K., et al., Molecular pharmacology 2010, 78, 996; Viader, A., et al., Cell reports 2015, 12, 798).
  • 2-AG hydrolysis results in the formation of arachidonic acid (AA), the precursor of prostaglandins (PGs) and leukotrienes (LTs).
  • Oxidative metabolism of AA is increased in inflamed tissues.
  • the cyclo-oxygenase which produces PGs
  • the 5-lipoxygenase which produces LTs.
  • PGE2 is one of the most important. These products have been detected at sites of inflammation, e.g. in the cerebrospinal fluid of patients suffering from neurodegenerative disorders and are believed to contribute to inflammatory response and disease progression.
  • mice lacking MAGL exhibit dramatically reduced 2-AG hydrolase activity and elevated 2-AG levels in the nervous system while other arachidonoyl-containing phospho- and neutral lipid species including anandamide (AEA), as well as other free fatty acids, are unaltered.
  • AEA arachidonoyl-containing phospho- and neutral lipid species including anandamide
  • levels of AA and AA-derived prostaglandins and other eicosanoids including prostaglandin E2 (PGE2), D2 (PGD2), F2 (PGF2), and thromboxane B2 (TXB2), are strongly decreased.
  • Phospholipase A 2 (PLA 2 ) enzymes have been viewed as the principal source of AA, but cPLA 2 -deficient mice have unaltered AA levels in their brain, reinforcing the key role of MAGL in the brain for AA production and regulation of the brain inflammatory process.
  • Neuroinflammation is a common pathological change characteristic of diseases of the brain including, but not restricted to, neurodegenerative diseases (e.g. multiple sclerosis, Alzheimer's disease. Parkinson disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy and mental disorders such as anxiety and migraine).
  • neurodegenerative diseases e.g. multiple sclerosis, Alzheimer's disease. Parkinson disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy and mental disorders such as anxiety and migraine.
  • LPS lipopolysaccharide
  • LPS treatment also induces a widespread elevation in pro-inflammatory cytokines including interleukin-1-a (IL-1-a), IL-1b, IL-6, and tumor necrosis factor-a (TNF-a) that is prevented in MgII ⁇ / ⁇ mice.
  • IL-1-a interleukin-1-a
  • IL-6 IL-6
  • TNF-a tumor necrosis factor-a
  • Neuroinflammation is characterized by the activation of the innate immune cells of the central nervous system, the microglia and the astrocytes. It has been reported that anti-inflammatory drugs can suppress in preclinical models the activation of glia cells and the progression of disease including Alzheimer's disease and multiple sclerosis (Lleo A., Cell Mol Life Sci. 2007, 64, 1403). Importantly, genetic and/or pharmacological disruption of MAGL activity also blocks LPS-induced activation of microglial cells in the brain (Nomura, D. K., et al., Science 2011, 334, 809).
  • MAGL activity was shown to be protective in several animal models of neurodegeneration including, but not restricted to, Alzheimer's disease, Parkinson's disease and multiple sclerosis.
  • an irreversible MAGL inhibitor has been widely used in preclinical models of neuroinflammation and neurodegeneration (Long, J. Z., et al., Nature chemical biology 2009, 5, 37).
  • Systemic injection of such inhibitor recapitulates the MgII ⁇ / ⁇ mice phenotype in the brain, including an increase in 2-AG levels, a reduction in AA levels and related eicosanoids production, as well as the prevention of cytokines production and microglia activation following LPS-induced neuroinflammation (Nomura, D. K., et al., Science 2011, 334, 809), altogether confirming that MAGL is a druggable target.
  • 2-AG has been reported to show beneficial effects on pain with, for example, anti-nociceptive effects in mice (Ignatowska-Jankowska B. et al., J. Pharmacol. Exp. Ther. 2015, 353, 424.) and on mental disorders, such as depression in chronic stress models (Zhong P. et al., Neuropsychopharmacology 2014, 39, 1763.).
  • oligodendrocytes (OLs), the myelinating cells of the central nervous system, and their precursors (OPCs) express the cannabinoid receptor 2 (CB2) on their membrane.
  • CB2 cannabinoid receptor 2
  • 2-AG is the endogenous ligand of CB1 and CB2 receptors. It has been reported that both cannabinoids and pharmacological inhibition of MAGL attenuate OLs's and OPCs's vulnerability to excitotoxic insults and therefore may be neuroprotective (Bernal-Chico, A., et al., Glia 2015, 63, 163.).
  • MAGL inhibition increases the number of myelinating OLs in the brain of mice, suggesting that MAGL inhibition may promote differentiation of OPCs in myelinating OLs in vivo (Alpar, A., et al., Nature communications 2014, 5, 4421.). Inhibition of MAGL was also shown to promote remyelination and functional recovery in a mouse model of progressive multiple sclerosis (Feliu A. et al., Journal of Neuroscience 2017, 37 (35), 8385.).
  • MAGL as an important decomposing enzyme for both lipid metabolism and the endocannabinoids system, additionally as a part of a gene expression signature, contributes to different aspects of tumourigenesis (Qin, H., et al., Cell Biochem. Biophys. 2014, 70, 33: Nomura D K et al., Cell 2009, 140(1), 49-61; Nomura D K et al., Chem. Biol. 2011, 18(7), 846-856).
  • suppressing the action and/or the activation of MAGL is a promising new therapeutic strategy for the treatment or prevention of neuroinflammation, neurodegenerative diseases, pain, cancer and mental disorders. Furthermore, suppressing the action and/or the activation of MAGL is a promising new therapeutic strategy for providing neuroprotection and myelin regeneration. Accordingly, there is a high unmet medical need for new MAGL inhibitors.
  • the present invention provides a compound of formula (I)
  • the present invention provides a process of manufacturing the urea compounds of formula (I) described herein, comprising:
  • the present invention provides a compound of formula (I) as described herein, when manufactured according to the processes described herein.
  • the present invention provides a compound of formula (I) as described herein, for use as therapeutically active substance.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) as described herein and a therapeutically inert carrier.
  • the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for inhibiting monoacylglycerol lipase (MAGL) in a mammal.
  • a compound of formula (I) as described herein or of a pharmaceutical composition described herein for inhibiting monoacylglycerol lipase (MAGL) in a mammal.
  • the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for the treatment or to prophylaxis of multiple sclerosis.
  • the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein for use in a method of inhibiting monoacylglycerol lipase in a mammal.
  • the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein for use in the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein, for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • multiple sclerosis Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for inhibiting monoacylglycerol lipase in a mammal.
  • the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • multiple sclerosis Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • the present invention provides a method for inhibiting monoacylglycerol lipase in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • alkyl refers to a mono- or multivalent, e.g., a mono- or bivalent, linear or branched saturated hydrocarbon group of 1 to 12 carbon atoms.
  • the alkyl group contains 1 to 6 carbon atoms (“C 1-6 -alkyl”), e.g., 1, 2, 3, 4, 5, or 6 carbon atoms.
  • the alkyl group contains 1 to 3 carbon atoms, e.g., 1, 2 or 3 carbon atoms.
  • alkyl examples include methyl, ethyl, propyl, 2-propyl (isopropyl), n-butyl, iso-butyl, sec-butyl, tert-butyl, and 2,2-dimethylpropyl.
  • Particularly preferred, yet non-limiting examples of alkyl are methyl and tert-butyl.
  • alkoxy refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In some preferred embodiments, the alkoxy group contains 1 to 6 carbon atoms (“C 1-6 -alkoxy”). In other embodiments, the alkoxy group contains 1 to 4 carbon atoms. In still other embodiments, the alkoxy group contains 1 to 3 carbon atoms. Some non-limiting examples of alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy and tert-butoxy. A particularly preferred, yet non-limiting example of alkoxy is methoxy.
  • halogen refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).
  • halogen refers to fluoro (F), chloro (Cl) or bromo (Br).
  • Particularly preferred, yet non-limiting examples of “halogen” or “halo” are fluoro (F) and chloro (Cl).
  • cycloalkyl refers to a saturated or partly unsaturated monocyclic or bicyclic hydrocarbon group of 3 to 10 ring carbon atoms. In some preferred embodiments, the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 8 ring carbon atoms.
  • “Bicyclic cycloalkyl” refers to cycloalkyl moieties consisting of two saturated carbocycles having two carbon atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom.
  • the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 6 ring carbon atoms, e.g., of 3, 4, 5 or 6 carbon atoms.
  • cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • heterocyclyl and “heterocycloalkyl” are used herein interchangeably and refer to a saturated or partly unsaturated mono- or bicyclic, preferably monocyclic ring system of 3 to 10 ring atoms, preferably 3 to 8 ring atoms, wherein 1, 2, or 3 of said ring atoms are heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • 1, 2, or 3 of said ring atoms are heteroatoms selected from N, O and S, the remaining ring atoms being carbon.
  • 1 to 2 of said ring atoms are selected from N and O, the remaining ring atoms being carbon.
  • Bicyclic heterocyclyl refers to heterocyclic moieties consisting of two cycles having two ring atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom.
  • monocyclic heterocyclyl groups include azetidin-3-yl, azetidin-2-yl, oxetan-3-yl, oxetan-2-yl, l-piperidyl, 2-piperidyl, 3-piperidyl, 4-piperidyl, 2-oxopyrrolidin-1-yl, 2-oxopyrrolidin-3-yl, 5-oxopyrrolidin-2-yl, 5-oxopyrrolidin-3-yl, 2-oxo-1-piperidyl, 2-oxo-3-piperidyl, 2-oxo-4-piperidyl, 6-oxo-2-piperidyl, 6-oxo-3-piperidyl, morpholino, morpholin-2-yl and morpholin-3-yl.
  • aryl refers to a monocyclic, bicyclic, or tricyclic carbocyclic ring system having a total of 6 to 14 ring members, preferably, 6 to 12 ring members, and more preferably 6 to 10 ring members, and wherein at least one ring in the system is aromatic.
  • aryl include phenyl and 9H-fluorenyl (e.g, 9H-fluoren-9-yl).
  • a particularly preferred, yet non-limiting example of aryl is phenyl.
  • heteroaryl refers to a mono- or multivalent, monocyclic or bicyclic ring system having a total of 5 to 14 ring members, preferably, 5 to 12 ring members, and more preferably 5 to 10 ring members, wherein at least one ring in the system is aromatic, and at least one ring in the system contains one or more heteroatoms.
  • heteroaryl refers to a 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • heteroaryl refers to a 5-10 membered heteroaryl comprising 1 to 2 heteroatoms independently selected from O, S and N.
  • heteroaryl examples include thiazolyl (e.g. thiazol-2-yl); oxazolyl (e.g. oxazol-2-yl); 5,6-dihydro-4H-cyclopenta[d]thiazol-2-yl; 1,2,4-oxadiazol-5-yl; pyridyl (e.g, 2-pyridyl); pyrazolyl (e.g. pyrazol-1-yl); imidazolyl (e.g. imidazole-1-yl); benzoxazolyl (e.g. benzoxazol-2-yl) and oxazolo[5,4-c]pyridin-2-yl.
  • thiazolyl e.g. thiazol-2-yl
  • oxazolyl e.g. oxazol-2-yl
  • 5,6-dihydro-4H-cyclopenta[d]thiazol-2-yl 1,2,4-o
  • hydroxy refers to an —OH group.
  • cyano refers to a —CN (nitrile) group.
  • haloalkyl refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a halogen atom, preferably fluoro.
  • haloalkyl refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a halogen atom, most preferably fluoro.
  • Particularly preferred, yet non-limiting examples of haloalkyl are trifluoromethyl (CF 3 ) and trifluoroethyl (e.g, 2,2,2-trifluoroethyl).
  • haloalkoxy refers to an alkoxy group, wherein at least one of the hydrogen atoms of the alkoxy group has been replaced by a halogen atom, preferably fluoro.
  • haloalkoxy refers to an alkoxy group wherein 1, 2 or 3 hydrogen atoms of the alkoxy group have been replaced by a halogen atom, most preferably fluoro.
  • a particularly preferred, yet non-limiting example of haloalkoxy is trifluoromethoxy (—OCF 3 ).
  • hydroxyalkyl refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a hydroxy group.
  • hydroxyalkyl refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms, most preferably 1 hydrogen atom of the alkyl group have been replaced by a hydroxy group.
  • Preferred, yet non-limiting examples of hydroxyalkyl are hydroxymethyl and hydroxyethyl (e.g, 2-hydroxyethyl).
  • a particularly preferred, yet non-limiting example of hydroxyalkyl is hydroxymethyl.
  • haloaryl refers to an aryl group, wherein at least one of the hydrogen atoms of the aryl group has been replaced by a halogen atom.
  • haloaryl refers to an aryl group wherein 1, 2 or 3 hydrogen atoms, more preferably 1 or 2 hydrogen atoms, most preferably 1 hydrogen atom of the aryl group have been replaced by a halogen atom.
  • a particularly preferred, yet non-limiting example of haloaryl is chlorophenyl, in particular 4-chlorophenyl.
  • aryloxy refers to an aryl group, as previously defined, attached to the parent molecular moiety via an oxygen atom.
  • a preferred, yet non-limiting example of aryloxy is phenoxy.
  • haloaryloxy refers to a haloaryl group, as previously defined, attached to the parent molecular moiety via an oxygen atom.
  • a preferred, yet non-limiting example of haloaryloxy is 4-fluorophenoxy.
  • salts refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable.
  • the salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, in particular hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like.
  • salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like.
  • Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyimine resins and the like.
  • Particular pharmaceutically acceptable salts of compounds of formula (I) are hydrochloride salts.
  • ester refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms.
  • Representative examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates. Examples of pharmaceutically acceptable prodrug types are described in Higuchi and Stella. Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987.
  • protective group denotes the group which selectively blocks a reactive site in a multifunctional compound such that a chemical reaction can be carried out selectively at another unprotected reactive site in the meaning conventionally associated with it in synthetic chemistry.
  • Protective groups can be removed at the appropriate point.
  • Exemplary protective groups are amino-protective groups, carboxy-protective groups or hydroxy-protective groups.
  • Particular protective groups are the tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), fluorenylmethoxycarbonyl (Fmoc) and benzyl (Bn).
  • protective groups are the tert-butoxycarbonyl (Boc) and the fluorenylmethoxycarbonyl (Fmoc). More particular protective group is the tert-butoxycarbonyl (Boc).
  • Exemplary protective groups and their application in organic synthesis are described, for example, in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014, John Wiley & Sons, N.Y.
  • urea forming reagent refers to a chemical compound that is able to render a first amine to a species that will react with a second amine, thereby forming an urea derivative.
  • Non-limiting examples of urea forming reagents include bis(trichloromethyl) carbonate, phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate and 1,1′-carbonyldiimidazole.
  • the urea forming reagents described in G. Sartori et al., Green Chemistry 2000, 2, 140 are incorporated herein by reference.
  • the compounds of formula (I) can contain several asymmetric centers and can be present in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates.
  • the compound of formula (I) according to the invention is a cis-enantiomer of formula (Ia) or (Ib), respectively, as described herein.
  • the asymmetric carbon atom can be of the “R” or “S” configuration.
  • MAGL refers to the enzyme monoacylglycerol lipase.
  • the terms “MAGL” and “monoacylglycerol lipase” are used herein interchangeably.
  • treatment includes: (1) inhibiting the state, disorder or condition (e.g. arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); and/or (2) relieving the condition (i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms).
  • the benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician.
  • a medicament is administered to a patient to treat a disease, the outcome may not always be effective treatment.
  • prophylaxis as used herein includes: preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a mammal and especially a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition.
  • neuroinflammation as used herein relates to acute and chronic inflammation of the nervous tissue, which is the main tissue component of the two parts of the nervous system; the brain and spinal cord of the central nervous system (CNS), and the branching peripheral nerves of the peripheral nervous system (PNS).
  • Chronic neuroinflammation is associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis.
  • Acute neuroinflammation usually follows injury to the central nervous system immediately, e.g., as a result of traumatic brain injury (TBI).
  • TBI traumatic brain injury
  • TBI traumatic brain injury
  • intracranial injury relates to damage to the brain resulting from external mechanical force, such as rapid acceleration or deceleration, impact, blast waves, or penetration by a projectile.
  • neurodegenerative diseases relates to diseases that are related to the progressive loss of structure or function of neurons, including death of neurons.
  • Examples of neurodegenerative diseases include, but are not limited to, multiple sclerosis, Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis.
  • mental disorders also called mental illnesses or psychiatric disorders
  • psychiatric disorders relates to behavioral or mental patterns that may cause suffering or a poor ability to function in life. Such features may be persistent, relapsing and remitting, or occur as a single episode. Examples of mental disorders include, but are not limited to, anxiety and depression.
  • pain relates to an unpleasant sensory and emotional experience associated with actual or potential tissue damage.
  • pain include, but are not limited to, nociceptive pain, chronic pain (including idiopathic pain), neuropathic pain including chemotherapy induced neuropathy, phantom pain and phsychogenic pain.
  • a particular example of pain is neuropathic pain, which is caused by damage or disease affecting any part of the nervous system involved in bodily feelings (i.e., the somatosensory system).
  • “pain” is neuropathic pain resulting from amputation or thoracotomy.
  • “pain” is chemotherapy induced neuropathy.
  • neurotoxicity relates to toxicity in the nervous system. It occurs when exposure to natural or artificial toxic substances (neurotoxins) alter the normal activity of the nervous system in such a way as to cause damage to nervous tissue.
  • neurotoxicity include, but are not limited to, neurotoxicity resulting from exposure to substances used in chemotherapy, radiation treatment, drug therapies, drug abuse, and organ transplants, as well as exposure to heavy metals, certain foods and food additives, pesticides, industrial and/or cleaning solvents, cosmetics, and some naturally occurring substances.
  • cancer refers to a disease characterized by the presence of a neoplasm or tumor resulting from abnormal uncontrolled growth of cells (such cells being “cancer cells”).
  • cancer explicitly includes, but is not limited to, hepatocellular carcinoma, colon carcinogenesis and ovarian cancer.
  • mammal as used herein includes both humans and non-humans and includes but is not limited to humans, non-human primates, canines, felines, murines, bovines, equines, and porcines. In a particularly preferred embodiment, the term “mammal” refers to humans.
  • the present invention provides a compound of formula (I)
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ia):
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ib):
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ic):
  • said compound of formula (Ic) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Id):
  • said compound of formula (Id) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein p is 1 or 2.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is aryl substituted with R 4 , R 5 and R 6 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is phenyl substituted with R 4 , R 5 and R 6 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from phenyl, 4-tert-butylthiazol-2-yl, 4-tert-butyloxazol-2-yl, 2-chloro-4-fluoro-phenyl, 4-(trifluoromethyl)phenyl, 4-(trifluoromethoxy)phenyl, 4-chlorophenyl, 5,6-dihydro-4H-cyclopenta[d]thiazol-2-yl, 3-phenyl-1,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)-2-pyridyl, 4-(trifluoromethyl)pyrazol-1-yl, 2-fluoro-4-(trifluoromethyl)phenyl, 2,4-difluorophenyl, 4-chloro-3-fluoro-phenyl, 4-cyanophenyl, 4,4-difluoro-1
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is hydrogen or methyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is hydrogen.
  • the present invention provides a compound of formula (I) as described to herein, or a pharmaceutically acceptable salt thereof, wherein R 2 is hydrogen or C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 2 is hydrogen or methyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from hydrogen, halogen, C 1-6 -alkyl and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from hydrogen, halogen and C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from hydrogen, fluoro and methyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from hydrogen, methyl, fluoro and trifluoromethyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from hydrogen, halogen, halo-C 1-6 -alkoxy and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from halogen, halo-C 1-6 -alkoxy and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from chloro, OCF 3 and CF 3 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from hydrogen, chloro, fluoro, OCF 3 and CF 3 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, halogen, C 1-6 -alkyl, C 1-6 -alkoxy, heterocycloalkyl, C 3-10 -cycloalkyl, heteroaryl and haloaryl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, halogen, heterocycloalkyl, C 3-10 -cycloalkyl and haloaryl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, fluoro, chloro, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, methyl, methoxy, cyano, fluoro, chloro, pyrolidinyl, morpholinyl, pyrazolyl, cyclopentyl, cyclopropyl and 4-chlorophenyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 6 is hydrogen or halogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 6 is hydrogen or fluoro.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 6 is hydrogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is selected from hydrogen, C 1-6 -alkyl, aryl and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is tert-butyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is selected from hydrogen, tert-butyl, phenyl and CF 3 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen or C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen or methyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 9 is hydrogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is halogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is fluoro.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is halogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is fluoro.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 and R 11 are both fluoro.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 12 is hydrogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein p is 2.
  • the present invention provides a compound of formula (I) as described to herein, or a pharmaceutically acceptable salt thereof, wherein q is 0 or 1.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein q is 0.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from hydrogen, halogen, hydroxy, cyano, C 1-6 -alkyl, C 1-6 -alkanoyl, SF 5 , C 1-6 -alkoxy, halo-C 1-6 -alkoxy, halo-C 1-6 -alkyl, C 3-10 -cycloalkyl, 3- to 14-membered heterocyclyl, 3- to 14-membered heterocycloalkyl substituted with R 14 and R 15 , 5- to 14-membered heteroaryl, C 6 -C 14 -aryloxy, and halo-C 6 -C 14 -aryl.
  • R 4 is selected from hydrogen, halogen, hydroxy, cyano, C 1-6 -alkyl, C 1-6 -alkanoyl, SF 5 , C 1-6 -alkoxy, halo-C 1-6 -
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from halogen, SF 5 , C 1-6 -alkyl, C 1-6 -alkoxy, halo-C 1-6 -alkoxy, halo-C 1-6 -alkyl, C 3-10 -cycloalkyl, and 3- to 14-membered heterocycloalkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from chloro, SF 5 , methyl, methoxy, OCF 3 , CF 3 , cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, halogen, C 1-6 -alkyl, halo-C 1-6 -alkyl, C 1-6 -alkoxy, 3- to 14-membered heterocycloalkyl, C 3-10 -cycloalkyl, 5- to 14-membered heteroaryl, and halo-C 6 -C 14 -aryl.
  • R 5 is selected from hydrogen, cyano, halogen, C 1-6 -alkyl, halo-C 1-6 -alkyl, C 1-6 -alkoxy, 3- to 14-membered heterocycloalkyl, C 3-10 -cycloalkyl, 5- to 14-membered heteroaryl, and halo-C 6 -C 14 -aryl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, halogen, C 1-6 -alkyl, halo-C 1-6 -alkyl, 3- to 14-membered heterocycloalkyl, C 3-10 -cycloalkyl and halo-C 6 -C 14 -aryl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF 3 , pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is selected from hydrogen, C 1-6 -alkyl, C 6 -C 14 -aryl, halo-C 6 -C 14 -aryl, halo-C 6 -C 14 -aryloxy, and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is C 1-6 -alkyl or halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 7 is tert-butyl or CF 3 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is selected from hydrogen, halogen, C 1-6 -alkyl, and halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen or halo-C 1-6 -alkyl.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen or CF 3 .
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from C 1-6 -alkyl, C 1-6 -alkoxy, and halogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 15 is hydrogen or halogen.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from the compounds disclosed in Table 1.
  • the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from:
  • the present invention provides pharmaceutically acceptable salts or esters of the compounds of formula (I) as described herein.
  • the present invention provides pharmaceutically acceptable salts of the compounds according to formula (I) as described herein, especially hydrochloride salts.
  • the present invention provides pharmaceutically acceptable esters of the compounds according to formula (I) as described herein.
  • the present invention provides compounds according to formula (I) as described herein.
  • the compounds of formula (I) are isotopically-labeled by having one or more atoms therein replaced by an atom having a different atomic mass or mass number.
  • isotopically-labeled (i.e., radiolabeled) compounds of formula (I) are considered to be within the scope of this disclosure.
  • isotopes that can be incorporated into the compounds of formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as, but not limited to, 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 CL, 123 I, and 125 I, respectively.
  • Certain isotopically-labeled compounds of formula (I) for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e.
  • a compound of formula (I) can be enriched with 1, 2, 5, 10, 25, 50, 75, 90, 95, or 99 percent of a given isotope.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • one of the starting materials, intermediates or compounds of formula (I) contain one or more functional groups which are not stable or are reactive under the reaction conditions of one or more reaction steps
  • appropriate protective groups as described e.g., in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014. John Wiley & Sons, N.Y.
  • Such protective groups can be removed at a later stage of the synthesis using standard methods described in the literature.
  • compounds of formula (I) can be obtained as mixtures of diastereomers or enantiomers, which can be separated by methods well known in the art e.g., chiral HPLC, chiral SFC or chiral crystallization. Racemic compounds can e.g., be separated into their antipodes via diastereomeric salts by crystallization with optically pure acids or by separation of the antipodes by specific chromatographic methods using either a chiral adsorbent or a chiral eluent. It is equally possible to separate starting materials and intermediates containing stereogenic centers to afford diastereomerically/enantiomerically enriched starting materials and intermediates.
  • the compounds of formula (I) can be manufactured by the methods given below, by the methods given in the examples or by analogous methods.
  • Appropriate reaction conditions for the individual reaction steps are known to a person skilled in the art.
  • reaction conditions described in literature affecting the described reactions see for example: Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 2 nd Edition , Richard C. Larock, John Wiley & Sons, New York, N.Y. 1999). It was found convenient to carry out the reactions in the presence or absence of a solvent. There is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or the reagents involved and that it can dissolve the reagents, at least to some extent.
  • the described reactions can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. It is convenient to carry out the described reactions in a temperature range between ⁇ 78° C. to reflux.
  • the time required for the reaction may also vary widely, depending on many factors, notably the reaction temperature and the nature of the reagents. However, a period of from 0.5 hours to several days will usually suffice to yield the described intermediates and compounds.
  • the reaction sequence is not limited to the one displayed in the schemes, however, depending on the starting materials and their respective reactivity, the sequence of reaction steps can be freely altered.
  • urea forming reagent such as bis(trichloromethyl) carbonate using a suitable base and solvent such as, e.g. sodium bicarbonate in DCM, to give compounds of formula I (step a).
  • a urea forming reagent such as bis(trichloromethyl) carbonate using a suitable base and solvent such as, e.g. sodium bicarbonate in DCM
  • urea forming reagents include but are not limited to phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate or 1,1′-carbonyldiimidazole.
  • 3-aminopiperidin-4-ol derivatives 3 in which “PG” signifies a suitable protective group such as a Cbz or Boc protective group can be acylated for example with acyl chlorides 4 in which R 1 is as defined herein and “LG” signifies a suitable leaving group (e.g., Cl or Br), using a suitable base such as sodium or potassium carbonate, sodium hydroxide or sodium acetate in an appropriate solvent such as THF, water, acetone or mixtures thereof, to provide intermediates 5 (step a).
  • Intermediates 5 can be cyclized to intermediates 6 using methods well known in the art, for example by treatment of 5 with sodium hydride in THF or potassium tert-butoxide in IPA and water (step b). Reactions of that type are described in literature (e.g. Z. Rafinski et al., J. Org. Chem. 2015, 80, 7468; S. Dugar et al., Synthesis 2015, 47(5), 712; WO2005/066187).
  • Intermediates 1 can be obtained as mixtures of diastereomers and enantiomers, respectively, or as single stereoisomers depending on whether racemic mixtures or enantiomerically pure forms of cis- or trans-3-aminopiperidin-4-ol derivatives 3 or intermediates 4 are employed in their syntheses.
  • Intermediates 3 are commercially available and their synthesis has also been described in literature (e.g. WO2005/066187; WO2011/0059118; WO2016/185279).
  • Optically pure cis-configured intermediates 1B and 1C can be obtained for example according to Scheme 3 by chiral separation of commercially available rac-(4aR,8aS)-4a,5,6,7,8,8a-hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one (1A) (optionally in form of a salt such as, e.g. a hydrochloride salt) using methods known in the art, e.g. by diastereomeric salt crystallization or by chiral chromatography (step a).
  • a salt such as, e.g. a hydrochloride salt
  • intermediates 2 are intermediates of type B.
  • Intermediates of type B in which A, m, n and R 2 are as described herein can be prepared by methods well known by a person skilled in the art and as exemplified by the general synthetic procedures outlined in Scheme 4.
  • Ketones 7, either commercially available or prepared by methods known in the art, can be subjected for example to a Wittig reaction with alkylidene triphenylphosphoranes of type 8a in a suitable solvent such as, e.g. THF. Methyl-THF or DMSO to give intermediates 9 (step a).
  • Phosphoranes 8a can be formed by treating the corresponding phosphonium salts with a suitable base such as BuLi. NaH, or KOtBu in a suitable solvent such as THF, dioxane or Methyl-THF and may be isolated or used in situ.
  • Phosphonium salts in turn are readily available from an aryl/heteroaryl/heterocyclic-substituted alkylhalide (with halide being Cl, Br and iodo) and triphenylphosphine in a suitable solvent such as toluene. Heating may be applied to accelerate the reaction or drive the reaction to completion (e.g. H. J. Cristau, F. Plisset in PATAI'S Chemistry of Functional Groups, Editor(s): Frank R. Hartley, 07th August 2006, Series Editor(s): Prof Saul Patai).
  • intermediates 9 can be obtained using a Homer-Wadsworth-Emmons (HWE) reaction using ketones 7 and phosphonates 8b, wherein R a is alkyl, for example methyl or ethyl.
  • Phosphonates 8b are in situ ⁇ -metalated using a suitable base and solvent such as NaH, nBuLi or KOtBu in THF (step a).
  • Phosphonates 8b are readily prepared using for example the Arbuzov reaction by alkylation of an aryl/heteroaryl/heterocyclic halide (with halide being Cl, Br and iodo) with commercially available trialkyl phosphite (e.g. Chem. Rev. 1984, 84, 577).
  • intermediates 2 are intermediates of type C.
  • compounds 11a,b can be subjected to a reductive amination reaction with aldehydes of type 13 using a suitable reducing agent and solvent such as NaBH 3 CN in MeOH, AcOH or mixtures thereof to give intermediates 14a,b (step a).
  • a suitable reducing agent and solvent such as NaBH 3 CN in MeOH, AcOH or mixtures thereof
  • compounds of formula I are compounds of type Ie.
  • compounds 1 can be coupled with mono-protected piperazine or 1,4-diazepane derivatives lib in which PG signifies a suitable protective group such as a Cbz or Boc protective group applying for example the conditions outlined under Scheme 1, step a, to give intermediates 16 (step c).
  • PG signifies a suitable protective group such as a Cbz or Boc protective group applying for example the conditions outlined under Scheme 1, step a, to give intermediates 16 (step c).
  • step d Removal of the protective group by published methods or as described under Scheme 5, step c, furnishes intermediates 15 (step d).
  • intermediates 2 are intermediates of type D.
  • Intermediates of type D in which A, m, n and R 2 are as described herein and R 3 is selected from hydrogen, halogen, C 1-6 -alkoxy, C 1-6 -alkyl and halo-C 1-6 -alkyl, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 7.
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 18 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 19 (step a).
  • Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739 ; Chem. Rev. 2009, 109 (6), 2551).
  • step c furnishes intermediates D (step b).
  • intermediates 19 may be prepared from alcohols 17 that can be alkylated with compounds 20 in which LG is a suitable leaving group such as chlorine, bromine, iodine.
  • LG is a suitable leaving group such as chlorine, bromine, iodine.
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • OSO 2 aryl e.g. p-toluenesulfonate using a suitable base in an appropriate solvent (e.g. sodium hydride in DMF) at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • intermediates 19 may be synthesized via alkylation of alcohols of type 18 with compounds 21 under the conditions described under step c (step d).
  • intermediates 2 are intermediates of type E.
  • Intermediates of type E in which A, m, n, R 2 and R 3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 8.
  • R 3 is a hydroxy group a suitable protective group strategy known to those skilled in the art may be applied.
  • Intermediates 23 can be reacted with compounds of type 24, either commercially available or synthesized by methods known in the art and as described below to yield intermediates 25 (step b).
  • Compounds 25 can be further converted into compounds 26 by a deoxyfluorination reaction using a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine trihydrofluoride and TEA or DBU) in a suitable solvent such as DCM or ACN (step d).
  • a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine tri
  • step c furnishes intermediates E (step e).
  • intermediates 2 are intermediates of type F.
  • Intermediates F in which A, m, n, R 2 and R 3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 9.
  • R 3 is a hydroxy group a suitable protective group strategy known to those skilled in the art may be applied.
  • Intermediates 30 can be reacted with compounds of type 31, either commercially available or synthesized by methods known in the art and as described below to yield intermediates 32 (step b).
  • magnesium e.g. magnesium turnings optionally in the presence of catalytic amounts of iodine, powder in the presence of LiCl or Rieke magnesium, organic halides
  • halogen-magnesium exchange by treating 34 in which Hal
  • Compounds 32 can be further converted into compounds 35 by a deoxyfluorination reaction using a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine trihydrofluoride and TEA or DBU) in a suitable solvent such as DCM or ACN (step e).
  • a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine tri
  • step c Removal of the protective group from intermediates 35 applying literature methods and as described for example under Scheme 3, step c, furnishes intermediates F (step f).
  • intermediates 2 are intermediates of type G.
  • Intermediates of type G in which A, m, n, R 2 are as described herein and R 3 is hydrogen, C 1-6 -alkoxy, C 1-6 -alkyl and halo-C 1-6 -alkyl, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 10.
  • Intermediates 38 may be prepared from alcohols 36 in which PG is a suitable protective group such as a Cbz, Boc or Bn, that can be alkylated with compounds 37 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base, such as sodium hydride, potassium tert-butoxide, in an appropriate solvent (e.g. in DMF or THF) at temperatures between 0° C. and the boiling temperature of the solvent (step a).
  • PG is a suitable protective group such as a Cbz, Boc or Bn
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methane
  • step c furnishes intermediates G (step b).
  • intermediates 2 are intermediates of type H.
  • Intermediates of type H in which A, m, n, R 2 and R 3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 11.
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 39 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 41 (step a).
  • Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739 ; Chem. Rev. 2009, 109 (6), 2551).
  • step c furnishes intermediates H (step b).
  • intermediates 41 may be prepared from alcohols 17 that can be alkylated with compounds 40 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs 2 CO 3 , NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • OSO 2 aryl e.g. p-toluenesulfon
  • step a Reacting intermediates H with intermediates 1, for example using the conditions described under scheme 1, step a, affords compounds of type If, wherein A, R 1 , R 2 , R 3 , m and n are as defined herein.
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 42, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give compounds ID (step a).
  • Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739 ; Chem. Rev. 2009, 109 (6), 2551).
  • compounds ID may be directly prepared from alcohols 17 that can be alkylated with compounds 43 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs 2 CO 3 , NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step b).
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • OSO 2 aryl e.g. p-toluenesulfon
  • intermediates 2 are intermediates of type J.
  • Intermediates of type J in which A, m, n, R 2 and R 3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 13.
  • Intermediates 46 may be prepared from heterocycloalkyls or heteroaryls 45 that can be alkylated with compounds 44 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs 2 CO 3 , K 2 CO 3 , NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step a).
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • OSO 2 aryl
  • step c furnishes intermediates J (step b).
  • compounds of type Ig may be directly prepared from heterocycloalkyls or heteroaryls 45 that can be alkylated with compounds 47 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs 2 CO 3 , K 2 CO 3 , NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (Scheme 14).
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • intermediates 2 are intermediates of type K.
  • Intermediates of type K in which A, m, n, R 2 and R 3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 15.
  • Alcohols of type 48 can be subjected to a Mitsunobu reaction with intermediates 39 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 49 (step a).
  • Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739 ; Chem. Rev. 2009, 109 (6), 2551).
  • step c furnishes intermediates K (step b).
  • intermediates 49 may be prepared from alcohols 48 that can be alkylated with compounds 40 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO 2 alkyl (e.g. methanesulfonate), OSO 2 fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO 2 aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs 2 CO 3 , NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • LG is a suitable leaving group such as chlorine, bromine, iodine
  • OSO 2 alkyl e.g. methanesulfonate
  • OSO 2 fluoroalkyl e.g. trifluoromethanesulfonate
  • OSO 2 aryl e.g. p-toluenesulfon
  • intermediates 2 are intermediates of type L.
  • Intermediates of type L in which A, m, n, R 2 and R 3 are as described herein can be prepared by methods well known by a person skilled in the art and as exemplified by the general synthetic procedures outlined in Scheme 16.
  • Intermediates 51 can be prepared for example from aldehydes 50, either commercially available or prepared by methods known in the art, using a Wittig reaction or Homer-Wadsworth-Emmons (HWE) reaction using alkylidene triphenylphosphoranes of type 8a and phosphonates 8b, respectively, as described under step a in Scheme 4 (step a).
  • HWE Homer-Wadsworth-Emmons
  • the present invention provides a process of manufacturing the urea compounds of formula (I) described herein, comprising:
  • urea forming reagent is selected from bis(trichloromethyl) carbonate, phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate and 1,1′-carbonyldiimidazole, preferably wherein said urea forming reagent is bis(trichloromethyl) carbonate.
  • the present invention provides a compound of formula (I) as described herein, when manufactured according to any one of the processes described herein.
  • compositions of the present invention are MAGL inhibitors.
  • the present invention provides the use of compounds of formula (I) as described herein for inhibiting MAGL in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in a method of inhibiting MAGL in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for inhibiting MAGL in a mammal.
  • the present invention provides a method for inhibiting MAGL in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the 4-NPA assay was carried out in 384 well assay plates (black with clear bottom, non-binding surface treated, Coming Ref 3655) in a total volume of 40 ⁇ L.
  • Compound dilutions were made in 100% DMSO (VWR Chemicals 23500.297) in a polypropylene plate in 3-fold dilution steps to give a final concentration range in the assay from 25 ⁇ M to 1.7 nM.
  • 1 ⁇ L compound dilutions (100% DMSO) were added to 19 ⁇ L MAGL (recombinant wild-type) in assay buffer (50 mM TRIS (GIBCO, 15567-027), 1 mM EDTA (Fluka, 03690-100 ml)).
  • the plate was shaked for 1 min at 2000 rpm (Variomag Teleshake) and then incubated for 15 min at RT.
  • 20 ⁇ L 4-Nitrophenlyacetate (Sigma N-8130) in assay buffer with 6% EtOH was added.
  • the final concentrations in the assay were 1 nM MAGL and 300 ⁇ M 4-Nitrophenylacetate.
  • the absorbance at 405 nm was measured for a fist time (Molecular Devices, SpectraMax Paradigm).
  • a second measurement was then done after incubation for 80 min at RT. From the two measurements, the slope was calculated by subtracting the first from the second measurement.
  • the 2-AG assay was carried out in 384 well assay plates (PP. Greiner Cat #784201) in a total volume of 20 ⁇ L. Compound dilutions were made in 100% DMSO (VWR Chemicals 23500.297) in a polypropylene plate in 3-fold dilution steps to give a final concentration range in the assay from 12.5 ⁇ M to 0.8 ⁇ M. 0.25 ⁇ L compound dilutions (100% DMSO) were added to 9 ⁇ L MAGL in assay buffer (50 mM TRIS (GIBCO, 15567-027), 1 mM EDTA (Fluka, 03690-100 ml), 0.01% (v/v) Tween.
  • a C18 SPE cartridge (G9205A) was used in an acetonitrile/water liquid setup.
  • the mass spectrometer was operated in negative electrospray mode following the mass transitions 303.1 ⁇ 259.1 for arachidonic acid and 311.1 ⁇ 267.0 for d8-arachidonic acid.
  • the activity of the compounds was calculated based on the ratio of intensities [arachidonic acid/d8-arachidonic acid].
  • the present invention provides compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein, wherein said compounds of formula (I) and their pharmaceutically acceptable salts or esters have IC 50 's for MAGL inhibition below 25 ⁇ M, preferably below 10 ⁇ M, more preferably below 5 ⁇ M as measured in the MAGL assay described herein.
  • compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein have IC 50 (MAGL inhibition) values between 0.0000001 ⁇ M and 25 ⁇ M, particular compounds have IC 50 values between 0.000005 ⁇ M and 10 ⁇ M, further particular compounds have IC 50 values between 0.00005 ⁇ M and 5 ⁇ M, as measured in the MAGL assay described herein.
  • IC 50 MAGL inhibition
  • the present invention provides compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein, wherein said compounds of formula (I) and their pharmaceutically acceptable salts or esters have an IC 50 for MAGL below 25 ⁇ M, preferably below 10 ⁇ M, more preferably below 5 ⁇ M as measured in an assay comprising the steps of:
  • the present invention provides compounds of formula (I) as described herein for use as therapeutically active substance.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of cancer in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of cancer in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of cancer in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis in a mammal.
  • the present invention provides a method for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of neurodegenerative diseases in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression and/or pain in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a method for the treatment or prophylaxis of multiple sclerosis in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) as described herein and a therapeutically inert carrier.
  • the compounds of formula (I) and their pharmaceutically acceptable salts and esters can be used as medicaments (e.g. in the form of pharmaceutical preparations).
  • the pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragées, hard and soft gelatin capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays) or rectally (e.g. in the form of suppositories).
  • the administration can also be effected parentally, such as intramuscularly or intravenously (e.g. in the form of injection solutions).
  • the compounds of formula (I) and their pharmaceutically acceptable salts and esters can be processed with pharmaceutically inert, inorganic or organic adjuvants for the production of tablets, coated tablets, dragées and hard gelatin capsules.
  • Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts etc. can be used, for example, as such adjuvants for tablets, dragées and hard gelatin capsules.
  • Suitable adjuvants for soft gelatin capsules are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols, etc.
  • Suitable adjuvants for the production of solutions and syrups are, for example, water, polyols, saccharose, invert sugar, glucose, etc.
  • Suitable adjuvants for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc.
  • Suitable adjuvants for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols, etc.
  • the pharmaceutical preparations can contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances.
  • the dosage can vary in wide limits and will, of course, be fitted to the individual requirements in each particular case.
  • the compounds of formula (I) or their pharmaceutically acceptable salts and esters can be used for the treatment or prophylaxis of type 2 diabetes related microvascular complications (such as, but not limited to diabetic retinopathy, diabetic neuropathy and diabetic nephropathy), coronary artery disease, obesity and underlying inflammatory diseases, chronic inflammatory and autoimmune/inflammatory diseases.
  • type 2 diabetes related microvascular complications such as, but not limited to diabetic retinopathy, diabetic neuropathy and diabetic nephropathy
  • coronary artery disease such as, but not limited to diabetic retinopathy, diabetic neuropathy and diabetic nephropathy
  • obesity underlying inflammatory diseases
  • chronic inflammatory and autoimmune/inflammatory diseases such as, chronic inflammatory and autoimmune/inflammatory diseases.
  • the pure enantiomers can be separated by methods described herein or by methods known to the man skilled in the art, such as e.g., chiral chromatography (e.g., chiral SFC) or crystallization.
  • a microwave vial was heat gun-dried and charged with bis(trichloromethyl) carbonate (26.6 mg, 89.6 ⁇ mol) and sodium bicarbonate (32.3 mg, 384 mol).
  • the flask was placed under argon and DCM (1 mL) was added to give a suspension.
  • the suspension was cooled by an ice-bath and 4-((2-chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine; hydrochloride salt (36.1 mg, 121 mol, BB15) was added. The mixture was stirred at 0° C. for 15 min and at RT overnight.
  • reaction mixture was cooled down in an-ice bath and DCM (500 ⁇ L) and DIPEA (49.7 mg, 67.1 ⁇ L, 384 ⁇ mol) followed by (4aR,8aS)-6-(4-((2-chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (21.1 mg, 47.5 ⁇ mol, BB1a) were added. The resulting off-white suspension was stirred at room temperature for 7 h. The reaction mixture was poured on water, DCM was added and the layers were separated. The aqueous layer was extracted twice with DCM.
  • Step b) (4aS,8aR)-6-(4-(((6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 42) and (4aR 8aS)-6-(4-(((6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 43)
  • example 117 The stereoisomers of example 117 were separated by preparative chiral HPLC (Reprosil Chiral NR column) using an isocratic mixture of EtOH (containing 0.05% of NH 4 OAc): n-heptane (40:60) to provide examples 103 and 104 as single isomers and example 105 as mixture of two stereoisomers. The fractions were evaporated to provide the desired compounds as colorless solids.
  • BB7 a crude racemic product
  • the crude material was directly submitted for a chiral SFC separation to yield enantiomer BB7b (2.72 g, second eluting to enantiomer) as a yellow solid and enantiomer BB7a (3.25 g, first eluting enantiomer) as a light beige solid but contaminated with BB7b.
  • a further SFC chiral separation was carried out to yield 2.71 g of BB7a.
  • MS (ESI): m/z 322.2 [M+H] + for both enantiomers.
  • Trifluoroacetic acid (2 g, 1.35 mL, 17.5 mmol) was added to a solution of tert-butyl 3-((2-chloro-4-(trifluoromethyl)phenoxy)methyl)azetidine-1-carboxylate (320 mg, 875 ⁇ mol) in DCM (4.37 mL) and the solution was stirred at RT for 2 h. The solvent was removed under reduced pressure and the resulting pale oil (470 mg) was diluted with EtOAc and washed with aq. Na 2 CO 3 solution.
  • the organic layers were washed twice with H 2 O, dried over MgSO 4 , filtered, treated with silica gel and evaporated.
  • the compound was purified by silica gel chromatography on a 120 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (50 to 100 in 30 min.) to provide the compound as a light yellow solid (2.48 g) which could be used in the following step without further purification.
  • Step a) Benzyl 3-(((2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate
  • benzyl 3-(aminomethyl)azetidine-1-carboxylate 0.5 g, 2.27 mmol
  • NEt 3 689 mg, 949 ⁇ L, 6.81 mmol
  • 2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethan-1-one 554 mg, 391 ⁇ L, 2.27 mmol
  • dry DCM 15 mL
  • Titanium tetrachloride 1M in DCM (1.13 mL, 1.13 mmol) was added via a syringe and the flask was cooled in an ice bath (exothermic).
  • the reaction was basified with sat. NaHCO 3 .
  • the obtained insoluble material was filtered over celite and the filtrate was extracted with DCM.
  • the organic layers were combined, washed with brine, dried over Na 2 SO 4 and concentrated.
  • the crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in n-heptane) to give the desired compound as a colorless oil (1 g).
  • Step b) Benzyl 3-(((1-(2,4-dichlorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate
  • tert-butyl 4-(hydroxymethyl)piperidine-1-carboxylate 80.7 mg, 375 ⁇ mol was dissolved in DMF (1.5 mL), then NaH in Oil 60% (18 mg, 450 ⁇ mol) was added at room temperature, the mixture was stirred for 20 min, then 5-bromo-3-methyl-2-(trifluoromethyl)pyridine (90 mg, 60 ⁇ L, 375 ⁇ mol) was added, and it was stirred for 2 hr at RT, yielding a brown solution. 10 mL sat. NH 4 C were added, it was extracted with water/ethyl acetate, dried over MgSO 4 , the solvent was removed at 40° C./150 mbar.
  • Step a Benzyl 3-(((1-(2-chloro-4-fluorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate
  • benzyl 3-(aminomethyl)azetidine-1-carboxylate 0.5 g, 2.27 mmol
  • triethylamine 689 mg, 949 ⁇ L, 6.81 mmol
  • 1-(2-chloro-4-fluoro-phenyl)-2,2,2-trifluoro-ethanone 519 mg, 2.27 mmol
  • dry DCM 15 mL
  • Titanium tetrachloride 1M in DCM (1.13 mL, 1.13 mmol) was added via a syringe to the ice-cooled flask (exothermic).
  • the reaction was stirred overnight at RT, carefully quenched with a methanolic solution of sodium cyanoborohydride (428 mg, 6.81 mmol) in methanol (4.36 g, 5.51 mL, 136 mmol)+Acetic Acid (0.1 mL) and stirred overnight at RT.
  • the reaction was basified with sat. NaHCO 3 .
  • the insoluble material obtained was filtered away over celite, the filtrate was extracted with DCM, the organic layers were combined, washed with brine, dried over Na 2 SO 4 and concentrated.
  • tert-butyl-3-hydroxy-2-methylazetidine-1-carboxylate (215 mg, 1.15 mmol) was dissolved in DMF (5 mL) to give a colorless solution.
  • sodium hydride (60% dispersion in mineral oil) (41.8 mg, 1.05 mmol) was added.
  • the reaction mixture was stirred at 0° C. for 15 min.
  • 1-(bromomethyl)-4-(trifluoromethyl)benzene (0.250 g, 1.05 mmol) was added at 0° C.
  • the reaction mixture was stirred at RT overnight.
  • the reaction mixture was poured onto 20 mL sat.
  • intermediates BB20, BB25 and BB61 of the following table were prepared from the commercially available phen9ols. Where trifluoroacetate salts are indicated, the crude product resulting from concentration of the reaction mixture was used directly without further neutralization or purification.
  • step a the following building blocks were prepared from the respective starting materials.
  • tert-butyl 3-(3-bromo-4-chlorophenoxy)azetidine-1-carboxylate (0.075 g, 0.165 mmol), cyclopropylboronic acid (0.021 g, 0.248 mmol) and K 2 CO 3 (0.046 g, 0.331 mmol) were mixed in dioxane (1.6 mL). Then, water (0.4 mL) was added followed by bis(triphenylphosphine)palladium (II) chloride (0.012 g, 0.016 mmol) and the reaction mixture heated at 130° C. under microwave irradiation for 1 hour.
  • (+)-(4aR,8aS)-6-[3-[3-bromo-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (BB 205, 0.2 g, 0.420 mmol), Zn(CN) 2 (0.098 g, 0.840 mmol), Zn (0.027 g, 0.420 mmol), dppf (0.232 g, 0.420 mmol).
  • Hünig's base (0.108 g, 0.840 mmol) were mixed in DMA (10 mL) and the mixture was degassed.
  • (+)-(4aR,8aS)-6-[3-[3-bromo-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (BB203, 0.2 g, 0.420 mmol), 2-azaspiro[3.3]heptane (CAS RN 665-04-03, 0.117 g, 0.630 mmol), BINAP (0.052 g, 0.080 mmol) and K 2 CO 3 (0.173 g, 1.25 mmol) were mixed in DMF (10 mL) and the mixture was degassed.
  • Example 265 The following examples listed in the table below were prepared in analogy to the procedure described for the preparation of Example 265 by using the indicated intermediates and/or commercially available compounds and using the mentioned purification method such as reversed-phase HPLC or silica gel flash chromatography.
  • tert-butyl 4-(hydroxymethyl)azepane-1-carboxylate (480 mg, 2.09 mmol) was dissolved in THF (10 ml). Subsequently, 2-chloro-4-fluorophenol (337 mg, 251 ⁇ l, 2.3 mmol) and triphenylphosphine (604 mg, 2.3 mmol) were added and the clear solution was stirred for 5 min at rt. The mixture was cooled to 0° C. and DEAD (401 mg, 365 ⁇ l, 2.3 mmol) was added in portions over 10 min.
  • Triphenylphosphine (1.84 g, 7 mmol) and 1-(bromomethyl)-4-(trifluoromethyl)benzene (1.61 g, 6.74 mmol) were dissolved in xylene (35 ml).
  • the reaction mixture was heated to reflux at 155° C. for 3.5 h and then cooled to room temperature.
  • the precipitated white crystalline solid was collected by filtration, washed with diethyl ether and dried in vacuo.
  • the final compound (3.30 g, 6.58 mmol, 97.7% yield) was obtained as a white powder and directly used on the next step.
  • Step b tert-butyl (E)-4-(4-(trifluoromethyl)benzylidene)azepane-1-carboxylate
  • Step c tert-butyl 4-(4-(trifluoromethyl)benzyl)azepane-1-carboxylate
  • the compound was prepared in analogy to BB99, step a, and used in the next step without further purification.
  • the sulfoxide intermediate was isolated from the synthesis of the according sulfone building block BB99, step b.
  • the product was obtained as a white lyophilized powder (50 mg, 13.9%)
  • MS (ESI): m/z 328.1 [M ⁇ 56+H] + .
  • step a) tert-Butyl 3-((2-fluoro-6-(trifluoromethyl)benzyl)thio)azetidine-1-carboxylate
  • Step a tert-Butyl 4-[3-bromo-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The invention provides new heterocyclic compounds having the general formula
Figure US20200308190A1-20201001-C00001
wherein A, L, X, m, n, R1 and R2 are as described herein, compositions including the compounds, processes of manufacturing the compounds and methods of using the compounds.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of International Application No. PCT/EP2019/071520, filed Aug. 12, 2019, which claims priority to EP Application No. 18188681.3, filed Aug. 13, 2018, the disclosure of each of which is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to organic compounds useful for therapy or prophylaxis in a mammal, and in particular to monoacylglycerol lipase (MAGL) inhibitors for the treatment or to prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer, mental disorders, multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine and/or depression in a mammal.
  • BACKGROUND OF THE INVENTION
  • Endocannabinoids (ECs) are signaling lipids that exert their biological actions by interacting with cannabinoid receptors (CBRs), CB1 and CB2. They modulate multiple physiological processes including neuroinflammation, neurodegeneration and tissue regeneration (Iannotti, F. A., et al., Progress in lipid research 2016, 62, 107-28). In the brain, the main endocannabinoid, 2-arachidonoylglycerol (2-AG), is produced by diacyglycerol lipases (DAGL) and hydrolyzed by the monoacylglycerol lipase, MAGL. MAGL hydrolyses 85% of 2-AG; the remaining 15% being hydrolysed by ABHD6 and ABDH12 (Nomura, D. K., et al., Science 2011, 334, 809). MAGL is expressed throughout the brain and in most brain cell types, including neurons, astrocytes, oligodendrocytes and microglia cells (Chanda, P. K., et al., Molecular pharmacology 2010, 78, 996; Viader, A., et al., Cell reports 2015, 12, 798). 2-AG hydrolysis results in the formation of arachidonic acid (AA), the precursor of prostaglandins (PGs) and leukotrienes (LTs). Oxidative metabolism of AA is increased in inflamed tissues. There are two principal enzyme pathways of arachidonic acid oxygenation involved in inflammatory processes, the cyclo-oxygenase which produces PGs and the 5-lipoxygenase which produces LTs. Of the various cyclooxygenase products formed during inflammation, PGE2 is one of the most important. These products have been detected at sites of inflammation, e.g. in the cerebrospinal fluid of patients suffering from neurodegenerative disorders and are believed to contribute to inflammatory response and disease progression. Mice lacking MAGL (MgII−/−) exhibit dramatically reduced 2-AG hydrolase activity and elevated 2-AG levels in the nervous system while other arachidonoyl-containing phospho- and neutral lipid species including anandamide (AEA), as well as other free fatty acids, are unaltered. Conversely, levels of AA and AA-derived prostaglandins and other eicosanoids, including prostaglandin E2 (PGE2), D2 (PGD2), F2 (PGF2), and thromboxane B2 (TXB2), are strongly decreased. Phospholipase A2 (PLA2) enzymes have been viewed as the principal source of AA, but cPLA2-deficient mice have unaltered AA levels in their brain, reinforcing the key role of MAGL in the brain for AA production and regulation of the brain inflammatory process.
  • Neuroinflammation is a common pathological change characteristic of diseases of the brain including, but not restricted to, neurodegenerative diseases (e.g. multiple sclerosis, Alzheimer's disease. Parkinson disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy and mental disorders such as anxiety and migraine). In the brain, production of eicosanoids and prostaglandins controls the neuroinflammation process. The pro-inflammatory agent lipopolysaccharide (LPS) produces a robust, time-dependent increase in brain eicosanoids that is markedly blunted in MgII−/− mice. LPS treatment also induces a widespread elevation in pro-inflammatory cytokines including interleukin-1-a (IL-1-a), IL-1b, IL-6, and tumor necrosis factor-a (TNF-a) that is prevented in MgII−/− mice.
  • Neuroinflammation is characterized by the activation of the innate immune cells of the central nervous system, the microglia and the astrocytes. It has been reported that anti-inflammatory drugs can suppress in preclinical models the activation of glia cells and the progression of disease including Alzheimer's disease and multiple sclerosis (Lleo A., Cell Mol Life Sci. 2007, 64, 1403). Importantly, genetic and/or pharmacological disruption of MAGL activity also blocks LPS-induced activation of microglial cells in the brain (Nomura, D. K., et al., Science 2011, 334, 809).
  • In addition, genetic and/or pharmacological disruption of MAGL activity was shown to be protective in several animal models of neurodegeneration including, but not restricted to, Alzheimer's disease, Parkinson's disease and multiple sclerosis. For example, an irreversible MAGL inhibitor has been widely used in preclinical models of neuroinflammation and neurodegeneration (Long, J. Z., et al., Nature chemical biology 2009, 5, 37). Systemic injection of such inhibitor recapitulates the MgII−/− mice phenotype in the brain, including an increase in 2-AG levels, a reduction in AA levels and related eicosanoids production, as well as the prevention of cytokines production and microglia activation following LPS-induced neuroinflammation (Nomura, D. K., et al., Science 2011, 334, 809), altogether confirming that MAGL is a druggable target.
  • Consecutive to the genetic and/or pharmacological disruption of MAGL activity, the endogenous levels of the MAGL natural substrate in the brain, 2-AG, are increased. 2-AG has been reported to show beneficial effects on pain with, for example, anti-nociceptive effects in mice (Ignatowska-Jankowska B. et al., J. Pharmacol. Exp. Ther. 2015, 353, 424.) and on mental disorders, such as depression in chronic stress models (Zhong P. et al., Neuropsychopharmacology 2014, 39, 1763.).
  • Furthermore, oligodendrocytes (OLs), the myelinating cells of the central nervous system, and their precursors (OPCs) express the cannabinoid receptor 2 (CB2) on their membrane. 2-AG is the endogenous ligand of CB1 and CB2 receptors. It has been reported that both cannabinoids and pharmacological inhibition of MAGL attenuate OLs's and OPCs's vulnerability to excitotoxic insults and therefore may be neuroprotective (Bernal-Chico, A., et al., Glia 2015, 63, 163.). Additionally, pharmacological inhibition of MAGL increases the number of myelinating OLs in the brain of mice, suggesting that MAGL inhibition may promote differentiation of OPCs in myelinating OLs in vivo (Alpar, A., et al., Nature communications 2014, 5, 4421.). Inhibition of MAGL was also shown to promote remyelination and functional recovery in a mouse model of progressive multiple sclerosis (Feliu A. et al., Journal of Neuroscience 2017, 37 (35), 8385.).
  • Finally, in recent years, metabolism is talked highly important in cancer research, especially the lipid metabolism. Researchers believe that the de novo fatty acid synthesis plays an important role in tumor development. Many studies illustrated that endocannabinoids have anti-tumorigenic actions, including anti-proliferation, apoptosis induction and anti-metastatic effects. MAGL as an important decomposing enzyme for both lipid metabolism and the endocannabinoids system, additionally as a part of a gene expression signature, contributes to different aspects of tumourigenesis (Qin, H., et al., Cell Biochem. Biophys. 2014, 70, 33: Nomura D K et al., Cell 2009, 140(1), 49-61; Nomura D K et al., Chem. Biol. 2011, 18(7), 846-856).
  • In conclusion, suppressing the action and/or the activation of MAGL is a promising new therapeutic strategy for the treatment or prevention of neuroinflammation, neurodegenerative diseases, pain, cancer and mental disorders. Furthermore, suppressing the action and/or the activation of MAGL is a promising new therapeutic strategy for providing neuroprotection and myelin regeneration. Accordingly, there is a high unmet medical need for new MAGL inhibitors.
  • SUMMARY OF THE INVENTION
  • In a first aspect, the present invention provides a compound of formula (I)
  • Figure US20200308190A1-20201001-C00002
      • or a pharmaceutically acceptable salt thereof, wherein A, L, X, m, n, R1 and R2 are as described herein.
  • In one aspect, the present invention provides a process of manufacturing the urea compounds of formula (I) described herein, comprising:
      • reacting a first amine of formula 1, wherein R1 is as described herein, preferably wherein R1 is hydrogen,
  • Figure US20200308190A1-20201001-C00003
      • with a second amine 2, wherein A, L, m, n, X and R2 are as described herein
  • Figure US20200308190A1-20201001-C00004
      • in the presence of a base and a urea forming reagent,
        form said compound of formula (I).
  • In a further aspect, the present invention provides a compound of formula (I) as described herein, when manufactured according to the processes described herein.
  • In a further aspect, the present invention provides a compound of formula (I) as described herein, for use as therapeutically active substance.
  • In a further aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) as described herein and a therapeutically inert carrier.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for inhibiting monoacylglycerol lipase (MAGL) in a mammal.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein or of a pharmaceutical composition described herein for the treatment or to prophylaxis of multiple sclerosis. Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a further aspect, the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein for use in a method of inhibiting monoacylglycerol lipase in a mammal.
  • In a further aspect, the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein for use in the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In a further aspect, the present invention provides a compound of formula (I) as described herein or a pharmaceutical composition described herein, for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for inhibiting monoacylglycerol lipase in a mammal.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In a further aspect, the present invention provides the use of a compound of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a further aspect, the present invention provides a method for inhibiting monoacylglycerol lipase in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • In a further aspect, the present invention provides a method for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • In a further aspect, the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein or of a pharmaceutical composition described herein to the mammal.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein, unless incompatible therewith. All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive. The invention is not restricted to the details of any foregoing embodiments. The invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed.
  • The term “alkyl” refers to a mono- or multivalent, e.g., a mono- or bivalent, linear or branched saturated hydrocarbon group of 1 to 12 carbon atoms. In some preferred embodiments, the alkyl group contains 1 to 6 carbon atoms (“C1-6-alkyl”), e.g., 1, 2, 3, 4, 5, or 6 carbon atoms. In other embodiments, the alkyl group contains 1 to 3 carbon atoms, e.g., 1, 2 or 3 carbon atoms. Some non-limiting examples of alkyl include methyl, ethyl, propyl, 2-propyl (isopropyl), n-butyl, iso-butyl, sec-butyl, tert-butyl, and 2,2-dimethylpropyl. Particularly preferred, yet non-limiting examples of alkyl are methyl and tert-butyl.
  • The term “alkoxy” refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In some preferred embodiments, the alkoxy group contains 1 to 6 carbon atoms (“C1-6-alkoxy”). In other embodiments, the alkoxy group contains 1 to 4 carbon atoms. In still other embodiments, the alkoxy group contains 1 to 3 carbon atoms. Some non-limiting examples of alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy and tert-butoxy. A particularly preferred, yet non-limiting example of alkoxy is methoxy.
  • The term “halogen” or “halo” refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I). Preferably, the term “halogen” or “halo” refers to fluoro (F), chloro (Cl) or bromo (Br). Particularly preferred, yet non-limiting examples of “halogen” or “halo” are fluoro (F) and chloro (Cl).
  • The term “cycloalkyl” as used herein refers to a saturated or partly unsaturated monocyclic or bicyclic hydrocarbon group of 3 to 10 ring carbon atoms. In some preferred embodiments, the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 8 ring carbon atoms. “Bicyclic cycloalkyl” refers to cycloalkyl moieties consisting of two saturated carbocycles having two carbon atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom. Preferably, the cycloalkyl group is a saturated monocyclic hydrocarbon group of 3 to 6 ring carbon atoms, e.g., of 3, 4, 5 or 6 carbon atoms. Some non-limiting examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • The terms “heterocyclyl” and “heterocycloalkyl” are used herein interchangeably and refer to a saturated or partly unsaturated mono- or bicyclic, preferably monocyclic ring system of 3 to 10 ring atoms, preferably 3 to 8 ring atoms, wherein 1, 2, or 3 of said ring atoms are heteroatoms selected from N, O and S, the remaining ring atoms being carbon. Preferably, 1 to 2 of said ring atoms are selected from N and O, the remaining ring atoms being carbon. “Bicyclic heterocyclyl” refers to heterocyclic moieties consisting of two cycles having two ring atoms in common, i.e., the bridge separating the two rings is either a single bond or a chain of one or two ring atoms, and to spirocyclic moieties, i.e., the two rings are connected via one common ring atom. Some non-limiting examples of monocyclic heterocyclyl groups include azetidin-3-yl, azetidin-2-yl, oxetan-3-yl, oxetan-2-yl, l-piperidyl, 2-piperidyl, 3-piperidyl, 4-piperidyl, 2-oxopyrrolidin-1-yl, 2-oxopyrrolidin-3-yl, 5-oxopyrrolidin-2-yl, 5-oxopyrrolidin-3-yl, 2-oxo-1-piperidyl, 2-oxo-3-piperidyl, 2-oxo-4-piperidyl, 6-oxo-2-piperidyl, 6-oxo-3-piperidyl, morpholino, morpholin-2-yl and morpholin-3-yl.
  • The term “aryl” refers to a monocyclic, bicyclic, or tricyclic carbocyclic ring system having a total of 6 to 14 ring members, preferably, 6 to 12 ring members, and more preferably 6 to 10 ring members, and wherein at least one ring in the system is aromatic. Some non-limiting examples of aryl include phenyl and 9H-fluorenyl (e.g, 9H-fluoren-9-yl). A particularly preferred, yet non-limiting example of aryl is phenyl.
  • The term “heteroaryl” refers to a mono- or multivalent, monocyclic or bicyclic ring system having a total of 5 to 14 ring members, preferably, 5 to 12 ring members, and more preferably 5 to 10 ring members, wherein at least one ring in the system is aromatic, and at least one ring in the system contains one or more heteroatoms. Preferably, “heteroaryl” refers to a 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. Most preferably, “heteroaryl” refers to a 5-10 membered heteroaryl comprising 1 to 2 heteroatoms independently selected from O, S and N. Some preferred, yet non-limiting examples of heteroaryl include thiazolyl (e.g. thiazol-2-yl); oxazolyl (e.g. oxazol-2-yl); 5,6-dihydro-4H-cyclopenta[d]thiazol-2-yl; 1,2,4-oxadiazol-5-yl; pyridyl (e.g, 2-pyridyl); pyrazolyl (e.g. pyrazol-1-yl); imidazolyl (e.g. imidazole-1-yl); benzoxazolyl (e.g. benzoxazol-2-yl) and oxazolo[5,4-c]pyridin-2-yl.
  • The term “hydroxy” refers to an —OH group.
  • The term “cyano” refers to a —CN (nitrile) group.
  • The term “haloalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a halogen atom, preferably fluoro. Preferably. “haloalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms of the alkyl group have been replaced by a halogen atom, most preferably fluoro. Particularly preferred, yet non-limiting examples of haloalkyl are trifluoromethyl (CF3) and trifluoroethyl (e.g, 2,2,2-trifluoroethyl).
  • The term “haloalkoxy” refers to an alkoxy group, wherein at least one of the hydrogen atoms of the alkoxy group has been replaced by a halogen atom, preferably fluoro. Preferably, “haloalkoxy” refers to an alkoxy group wherein 1, 2 or 3 hydrogen atoms of the alkoxy group have been replaced by a halogen atom, most preferably fluoro. A particularly preferred, yet non-limiting example of haloalkoxy is trifluoromethoxy (—OCF3).
  • The term “hydroxyalkyl” refers to an alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by a hydroxy group. Preferably, “hydroxyalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms, most preferably 1 hydrogen atom of the alkyl group have been replaced by a hydroxy group. Preferred, yet non-limiting examples of hydroxyalkyl are hydroxymethyl and hydroxyethyl (e.g, 2-hydroxyethyl). A particularly preferred, yet non-limiting example of hydroxyalkyl is hydroxymethyl.
  • The term “haloaryl” refers to an aryl group, wherein at least one of the hydrogen atoms of the aryl group has been replaced by a halogen atom. Preferably, “haloaryl” refers to an aryl group wherein 1, 2 or 3 hydrogen atoms, more preferably 1 or 2 hydrogen atoms, most preferably 1 hydrogen atom of the aryl group have been replaced by a halogen atom. A particularly preferred, yet non-limiting example of haloaryl is chlorophenyl, in particular 4-chlorophenyl.
  • The term “aryloxy” refers to an aryl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. A preferred, yet non-limiting example of aryloxy is phenoxy.
  • The term “haloaryloxy” refers to a haloaryl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. A preferred, yet non-limiting example of haloaryloxy is 4-fluorophenoxy.
  • The term “pharmaceutically acceptable salt” refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, in particular hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like. In addition these salts may be prepared by addition of an inorganic base or an organic base to the free acid. Salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like. Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyimine resins and the like. Particular pharmaceutically acceptable salts of compounds of formula (I) are hydrochloride salts.
  • The term “pharmaceutically acceptable ester” refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Representative examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates. Examples of pharmaceutically acceptable prodrug types are described in Higuchi and Stella. Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987.
  • The term “protective group” (PG) denotes the group which selectively blocks a reactive site in a multifunctional compound such that a chemical reaction can be carried out selectively at another unprotected reactive site in the meaning conventionally associated with it in synthetic chemistry. Protective groups can be removed at the appropriate point. Exemplary protective groups are amino-protective groups, carboxy-protective groups or hydroxy-protective groups. Particular protective groups are the tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), fluorenylmethoxycarbonyl (Fmoc) and benzyl (Bn). Further particular protective groups are the tert-butoxycarbonyl (Boc) and the fluorenylmethoxycarbonyl (Fmoc). More particular protective group is the tert-butoxycarbonyl (Boc). Exemplary protective groups and their application in organic synthesis are described, for example, in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014, John Wiley & Sons, N.Y.
  • The term “urea forming reagent” refers to a chemical compound that is able to render a first amine to a species that will react with a second amine, thereby forming an urea derivative. Non-limiting examples of urea forming reagents include bis(trichloromethyl) carbonate, phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate and 1,1′-carbonyldiimidazole. The urea forming reagents described in G. Sartori et al., Green Chemistry 2000, 2, 140 are incorporated herein by reference.
  • The compounds of formula (I) can contain several asymmetric centers and can be present in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates. In a preferred embodiment, the compound of formula (I) according to the invention is a cis-enantiomer of formula (Ia) or (Ib), respectively, as described herein.
  • According to the Cahn-Ingold-Prelog Convention, the asymmetric carbon atom can be of the “R” or “S” configuration.
  • The abbreviation “MAGL” refers to the enzyme monoacylglycerol lipase. The terms “MAGL” and “monoacylglycerol lipase” are used herein interchangeably.
  • The term “treatment” as used herein includes: (1) inhibiting the state, disorder or condition (e.g. arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); and/or (2) relieving the condition (i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms). The benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician. However, it will be appreciated that when a medicament is administered to a patient to treat a disease, the outcome may not always be effective treatment.
  • The term “prophylaxis” as used herein includes: preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a mammal and especially a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition.
  • The term “neuroinflammation” as used herein relates to acute and chronic inflammation of the nervous tissue, which is the main tissue component of the two parts of the nervous system; the brain and spinal cord of the central nervous system (CNS), and the branching peripheral nerves of the peripheral nervous system (PNS). Chronic neuroinflammation is associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis. Acute neuroinflammation usually follows injury to the central nervous system immediately, e.g., as a result of traumatic brain injury (TBI).
  • The term “traumatic brain injury” (“TBI”, also known as “intracranial injury”), relates to damage to the brain resulting from external mechanical force, such as rapid acceleration or deceleration, impact, blast waves, or penetration by a projectile.
  • The term “neurodegenerative diseases” relates to diseases that are related to the progressive loss of structure or function of neurons, including death of neurons. Examples of neurodegenerative diseases include, but are not limited to, multiple sclerosis, Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis.
  • The term “mental disorders” (also called mental illnesses or psychiatric disorders) relates to behavioral or mental patterns that may cause suffering or a poor ability to function in life. Such features may be persistent, relapsing and remitting, or occur as a single episode. Examples of mental disorders include, but are not limited to, anxiety and depression.
  • The term “pain” relates to an unpleasant sensory and emotional experience associated with actual or potential tissue damage. Examples of pain include, but are not limited to, nociceptive pain, chronic pain (including idiopathic pain), neuropathic pain including chemotherapy induced neuropathy, phantom pain and phsychogenic pain. A particular example of pain is neuropathic pain, which is caused by damage or disease affecting any part of the nervous system involved in bodily feelings (i.e., the somatosensory system). In one embodiment, “pain” is neuropathic pain resulting from amputation or thoracotomy. In one embodiment, “pain” is chemotherapy induced neuropathy.
  • The term “neurotoxicity” relates to toxicity in the nervous system. It occurs when exposure to natural or artificial toxic substances (neurotoxins) alter the normal activity of the nervous system in such a way as to cause damage to nervous tissue. Examples of neurotoxicity include, but are not limited to, neurotoxicity resulting from exposure to substances used in chemotherapy, radiation treatment, drug therapies, drug abuse, and organ transplants, as well as exposure to heavy metals, certain foods and food additives, pesticides, industrial and/or cleaning solvents, cosmetics, and some naturally occurring substances.
  • The term “cancer” refers to a disease characterized by the presence of a neoplasm or tumor resulting from abnormal uncontrolled growth of cells (such cells being “cancer cells”). As used herein, the term cancer explicitly includes, but is not limited to, hepatocellular carcinoma, colon carcinogenesis and ovarian cancer.
  • The term “mammal” as used herein includes both humans and non-humans and includes but is not limited to humans, non-human primates, canines, felines, murines, bovines, equines, and porcines. In a particularly preferred embodiment, the term “mammal” refers to humans.
  • Compounds of the Invention
  • In a first aspect, the present invention provides a compound of formula (I)
  • Figure US20200308190A1-20201001-C00005
      • or a pharmaceutically acceptable salt thereof,
      • wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CH2O—, —CH2OCH2—, —CF2CH2—, and —CH2CF2—; or
      • (ii) X is N; m is 1; n is 1 or 2; and L is —(CH2)p— or —CF2CH2—;
      • p is selected from 1, 2 and 3:
      • A is selected from:
        • (i) aryl substituted with R4, R5 and R6;
        • (ii) heteroaryl substituted with R7, R8 and R9; and
        • (iii) heterocycloalkyl substituted with R10, R11 and R12;
      • R1 is hydrogen or C1-6-alkyl;
      • R2 is selected from hydrogen, C1-6-alkyl and hydroxy-C1-6-alkyl;
      • R3 is selected from hydrogen, halogen, hydroxy, C1-6-alkoxy, C1-6-alkyl and halo-C1-6-alkyl;
      • each of R4, R5, R6, R7, R8, R9, R10, R11 and R12 is independently selected from hydrogen, halogen, cyano, hydroxy, C1-6-alkyl, halo-C1-6-alkyl, hydroxy-C1-6-alkyl, halo-C1-5-alkyl-CH(OH)—, C1-6-alkoxy, halo-C1-6-alkoxy, SF5, CH3SO2, C3-10-cycloalkyl, C3-10-cycloalkyl substituted with R13, heterocycloalkyl, heterocycloalkyl substituted with R14, heteroaryl, aryl and haloaryl; and
      • each of R13 and R14 is independently C1-6-alkyl, halo-C1-6-alkyl or hydroxy.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ia):
  • Figure US20200308190A1-20201001-C00006
      • wherein A, L, X, m, n, R1 and R2 are as defined herein. Preferably, said compound of formula (Ia) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ib):
  • Figure US20200308190A1-20201001-C00007
      • wherein A, L, X, m, n, R1 and R2 are as defined herein. Preferably, said compound of formula (Ib) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ic):
  • Figure US20200308190A1-20201001-C00008
  • wherein A, L, X, m, n, R1 and R2 are as defined herein. Preferably, said compound of formula (Ic) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Id):
  • Figure US20200308190A1-20201001-C00009
  • wherein A, L, X, m, n, R1 and R2 are as defined herein. Preferably, said compound of formula (Id) has an enantiomeric excess (ee) of >80%, more preferably >90%, in particular >99%.
  • In one embodiment, there is provided a compound of formula (I) as described herein, wherein when X is C—R3 and R3 is hydroxy or halogen, L is not —O— or —CH2O—.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CH2O— and —CH2OCH2—; or
      • (ii) X is N; m and n are both 1; and L is —(CH2)p—.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3:
      • m and n are both 0; or
      • m and n are both 1; and
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—.
  • In another preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
      • R3 is selected from hydrogen, C1-6-alkyl and halogen; and
      • p is 1 or 2.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2—;
      • R3 is hydrogen; and
      • p is 1 or 2.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein p is 1 or 2.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
      • (i) aryl substituted with R4, R5 and R6; and
      • (ii) heteroaryl substituted with R7, R8 and R9.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
      • (i) phenyl substituted with R4, R5 and R6; and
      • (ii) oxazolyl substituted with R7, R8 and R9.
  • In another preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is aryl substituted with R4, R5 and R6.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is phenyl substituted with R4, R5 and R6.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
      • (i) aryl substituted with R4, R5 and R6; and
      • (ii) heteroaryl substituted with R7, R8 and R9;
    • R4 is selected from hydrogen, halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl;
    • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, C1-6-alkoxy, heterocycloalkyl, C3-10-cycloalkyl, heteroaryl and haloaryl;
    • R6 is hydrogen or halogen;
    • R7 is selected from hydrogen, C1-6-alkyl, aryl and halo-C1-6-alkyl;
    • R8 is hydrogen or C1-6-alkyl; and
    • R9 is hydrogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
      • (i) aryl substituted with R4, R5 and R6; and
      • (ii) heteroaryl substituted with R7, R8 and R9;
    • R4 is selected from halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl;
    • R5 is selected from hydrogen, cyano, halogen, heterocycloalkyl, C3-10-cycloalkyl and haloaryl;
    • R6 is hydrogen:
    • R7 is C1-6-alkyl;
    • R8 is hydrogen; and
    • R9 is hydrogen.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein: A is selected from:
      • (i) phenyl substituted with R4, R5 and R6; and
      • (ii) oxazolyl substituted with R7, R8 and R9;
    • R4 is selected from chloro OCF3 and CF3;
    • R5 is selected from hydrogen, cyano, fluoro, chloro, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl;
    • R6 is hydrogen:
    • R7 is tert-butyl;
    • R8 is hydrogen; and
    • R9 is hydrogen.
  • In a further particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from phenyl, 4-tert-butylthiazol-2-yl, 4-tert-butyloxazol-2-yl, 2-chloro-4-fluoro-phenyl, 4-(trifluoromethyl)phenyl, 4-(trifluoromethoxy)phenyl, 4-chlorophenyl, 5,6-dihydro-4H-cyclopenta[d]thiazol-2-yl, 3-phenyl-1,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)-2-pyridyl, 4-(trifluoromethyl)pyrazol-1-yl, 2-fluoro-4-(trifluoromethyl)phenyl, 2,4-difluorophenyl, 4-chloro-3-fluoro-phenyl, 4-cyanophenyl, 4,4-difluoro-1-piperidyl. 5-tert-butyloxazol-2-yl. 4-methoxy-2-fluoro-phenyl, 2-chloro-4(trifluoromethyl)phenyl, 6-(trifluoromethyl)-3-pyridyl, 3-(trifluoromethyl)phenyl, 2-chloro-4-(trifluoromethoxy)phenyl, 4-chloro-2-fluoro-phenyl, 4-fluoro-2-(trifluoromethyl)phenyl, 2-pyrrolidin-1-yl-4-(trifluoromethyl)phenyl, 4-fluoro-2-cyano-phenyl, 2-cyclopentyl-4-(trifluoromethyl)phenyl, 2-chloro-4-cyanophenyl, 4-(trifluoromethyl)imidazol-1-yl, 4-fluoro-2-methyl-phenyl, 4-tert-butylpyrazol-1-yl, 1,3-benzoxazol-2-yl, 4-chloro-3-(4-chlorophenyl)phenyl, 2-(1H-pyrazol-4-vl)-4-(trifluoromethyl)phenyl, 2,4-dichlorophenyl, 3-methoxy-4-(trifluoromethyl)phenyl, 5-methyl-6-(trifluoromethyl)-3-pyridyl, 3-chlorophenyl, 2-chlorophenyl, 2-cyclopropyl-4-(trifluoromethyl)phenyl, 2-methyl-4-(trifluoromethyl)phenyl, 3-fluoro-5-(trifluoromethyl)phenyl, 2-fluoro-6-(trifluoromethyl)phenyl, 3-chloro-4-(trifluoromethyl)phenyl, 2,4-difluoro-5-(trifluoromethyl)phenyl, 2-fluoro-5-(trifluoromethyl)phenyl, 2-methoxy-4-(trifluoromethyl)phenyl, 4-chloro-2-(trifluoromethyl)phenyl, 4-chloro-3-(trifluoromethyl)phenyl. 4-chloro-3-cyclopropyl-phenyl, 4-chloro-3-morpholino-phenyl, 2-cyano-4-(trifluoromethyl)phenyl, oxazolo[5,4-c]pyridin-2-yl, 4-Methyl-3-(trifluoromethyl)phenyl, 3-cyclopropyl-4-(trifluoromethyl)phenyl, 2-fluoro-4-methyl-phenyl, 4-methoxy-2-(trifluoromethyl)phenyl, 4-methyl-2-(trifluoromethyl)phenyl, 3,4-dichlorophenyl, 2,5-dichlorophenyl, 5-methyl-6-(trifluoromethyl)-3-pyridyl, 4,5-bis(trifluoromethyl)-2-pyridyl, 2-fluoro-4-(trifluoromethyl)-phenyl. 2-fluoro-4-(pentafluoro-lambda6-sulfanyl)phenyl, 2,4-bis(trifluoromethyl)phenyl, 2-methyl-3-(trifluoromethyl)phenyl, 2-methyl-4-(trifluoromethoxy)phenyl, 3-chloro-4-(trifluoromethyl)phenyl, 3-cyclopropyl-4-chloro-phenyl, 2-chloro-3-(trifluoromethyl)phenyl, 2-chloro-3-cyclopropyl-phenyl, 3-(2-azaspiro[3.3]heptan-2-yl)-4-(trifluoromethyl)phenyl, 2-chloro-3-(2-azaspiro[3.3]heptan-2-yl)-phenyl, 2-chloro-3-(5-oxa-2-azaspiro[3.5]nonan-2-yl)phenyl, 2-fluoro-4-(trifluoromethtly)phenyl, 2-fluoro-4-methyl-phenyl, 2-fluoro-6-(trifluoromethyl)phenyl, and 2-(trifluoromethyl)-4-methyl-phenyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen or methyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described to herein, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen or C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen or methyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen, C1-6-alkyl and halo-C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen and C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, fluoro and methyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, methyl, fluoro and trifluoromethyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from chloro, OCF3 and CF3.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, chloro, fluoro, OCF3 and CF3.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, C1-6-alkoxy, heterocycloalkyl, C3-10-cycloalkyl, heteroaryl and haloaryl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, heterocycloalkyl, C3-10-cycloalkyl and haloaryl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, fluoro, chloro, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, methyl, methoxy, cyano, fluoro, chloro, pyrolidinyl, morpholinyl, pyrazolyl, cyclopentyl, cyclopropyl and 4-chlorophenyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen or halogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen or fluoro.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is selected from hydrogen, C1-6-alkyl, aryl and halo-C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is tert-butyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is selected from hydrogen, tert-butyl, phenyl and CF3.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or methyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R9 is hydrogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R10 is halogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R10 is fluoro.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R11 is halogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R11 is fluoro.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R10 and R11 are both fluoro.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R12 is hydrogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CH2O— and —CH2OCH2—; or
      • (ii) X is N; m and n are both 1; and L is —(CH2)p—;
      • p is 1 or 2;
      • A is selected from:
        • (i) aryl substituted with R4, R5 and R6:
        • (ii) heteroaryl substituted with R7, R8 and R9; and
        • (iii) heterocycloalkyl substituted with R10, R11 and R12;
      • R1 is hydrogen or C1-6-alkyl;
      • R2 is selected from hydrogen or C1-6-alkyl;
      • R3 is selected from hydrogen, halogen, C1-6-alkyl and halo-C1-6-alkyl;
      • R4 is selected from hydrogen, halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl;
      • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, C1-6-alkoxy, heterocycloalkyl, C3-10-cycloalkyl, heteroaryl and haloaryl;
      • R6 is hydrogen or halogen:
      • R7 is selected from hydrogen, C1-6-alkyl, aryl and halo-C1-6-alkyl;
      • R8 is hydrogen or C1-6-alkyl;
      • R9 is hydrogen;
      • R10 is halogen;
      • R11 is halogen, and
      • R12 is hydrogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3:
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
      • p is 1 or 2;
      • A is selected from:
        • (i) aryl substituted with R4, R5 and R6; and
        • (ii) heteroaryl substituted with R7, R8 and R9;
      • R1 is hydrogen;
      • R2 is selected from hydrogen or C1-6-alkyl;
      • R1 is selected from hydrogen, halogen and C1-6-alkyl;
      • R4 is selected from halogen, halo-C1-6-alkoxy and halo-C1-6-alkyl;
      • R5 is selected from hydrogen, cyano, halogen, heterocycloalkyl, C3-10-cycloalkyl and haloaryl;
      • R6 is hydrogen;
      • R7 is C1-6-alkyl;
      • R8 is hydrogen; and
      • R9 is hydrogen.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3:
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
      • p is 1 or 2;
      • A is selected from:
        • (i) phenyl substituted with R4, R5 and R6; and
        • (ii) oxazolyl substituted with R7, R8 and R9;
      • R1 is hydrogen;
      • R2 is selected from hydrogen or methyl;
      • R1 is selected from hydrogen, fluoro and methyl;
      • R4 is selected from chloro, OCF3 and CF3;
      • R5 is selected from hydrogen, cyano, fluoro, chloro, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl;
      • R6 is hydrogen:
      • R7 is tert-butyl;
      • R8 is hydrogen; and
      • R9 is hydrogen.
  • In one aspect, the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CH2OCH2—, —CF2CH2—, —CH2═CH2—, —(CR16R17)q—CH2O—, and —CH2CF2—; or
      • (ii) X is N; m is 1; n is 1 or 2; and L is —(CH2)p— or —CF2CH2—;
      • p is selected from 1, 2 and 3:
      • q is 0 or 1;
      • A is selected from:
        • (i) C6-C14-aryl substituted with R4, R5 and R6;
        • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9; and
        • (iii) 3- to 14-membered heterocycloalkyl substituted with R10, R11 and R12;
      • R1 is hydrogen or C1-6-alkyl;
      • R2 is selected from hydrogen, C1-6-alkyl and hydroxy-C1-6-alkyl;
      • R3 is selected from hydrogen, halogen, hydroxy, C1-6-alkoxy, C1-6-alkyl and halo-C1-6-alkyl;
      • each of R4, R5, R6, R7, R8, R9, R10, R11 and R12 is independently selected from hydrogen, halogen, cyano, hydroxy, C1-6-alkyl, halo-C1-6-alkyl, hydroxy-C1-6-alkyl, C1-6-alkanoyl, halo-C3-5-alkyl-CH(OH)—, C1-6-alkoxy, halo-C1-6-alkoxy, SF5, CH3SO2, C3-10-cycloalkyl, C3-10-cycloalkyl substituted with R13, 3- to 14-membered heterocycloalkyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, C6-C14-aryl, C6-C14-aryloxy, halo-C6-C14-aryl, and halo-C6-C14-aryloxy;
      • each of R13, R14, and R15 is independently selected from C3-6-alkyl, C1-6-alkoxy, halo-C1-6-alkyl, halo-C1-6-alkoxy, halogen, and hydroxy; and
      • R16 and R17, taken together with the carbon atom to which they are attached, form a C3-10-cycloalkyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CF2CH2—, —CH2═CH2—, —(CR16R17)q—CH2O—, and —CH2OCH2—; or
      • (ii) X is N; m and n are both 1; and L is —(CH2)p—.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein p is 2.
  • In one embodiment, the present invention provides a compound of formula (I) as described to herein, or a pharmaceutically acceptable salt thereof, wherein q is 0 or 1.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein q is 0.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
      • (i) C6-C14-aryl substituted with R4, R5 and R6; and
      • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
      • (i) phenyl substituted with R4, R5 and R6;
      • (ii) oxazolyl substituted with R7, R8 and R9; and
      • (iii) pyridyl substituted with R7, R8 and R9.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, halogen, hydroxy, cyano, C1-6-alkyl, C1-6-alkanoyl, SF5, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, 3- to 14-membered heterocyclyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, C6-C14-aryloxy, and halo-C6-C14-aryl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from halogen, SF5, C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, and 3- to 14-membered heterocycloalkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from chloro, SF5, methyl, methoxy, OCF3, CF3, cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, C1-6-alkoxy, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl, 5- to 14-membered heteroaryl, and halo-C6-C14-aryl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl and halo-C6-C14-aryl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF3, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is selected from hydrogen, C1-6-alkyl, C6-C14-aryl, halo-C6-C14-aryl, halo-C6-C14-aryloxy, and halo-C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is C1-6-alkyl or halo-C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R7 is tert-butyl or CF3.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or halo-C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or CF3.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from C1-6-alkyl, C1-6-alkoxy, and halogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R15 is hydrogen or halogen.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —(CR16R17)q—CH2O—, —CH2OCH2—, —CF2CH2—, and —CH2═CH2—; or
      • (ii) X is N; m and n are both 1; and L is —(CH2)p—;
      • p is 1 or 2;
      • q is 0 or 1;
      • A is selected from:
        • (i) C6-C14-aryl substituted with R4, R5 and R6;
        • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9; and
      • (iii) 3- to 14-membered heterocycloalkyl substituted with R10, R11 and R12;
      • R1 is hydrogen or C1-6-alkyl;
      • R2 is hydrogen or C1-6-alkyl;
      • R3 is selected from hydrogen, halogen, C1-6-alkyl and halo-C1-6-alkyl;
      • R4 is selected from hydrogen, halogen, cyano, SF5, C1-6-alkyl, C1-6-alkanoyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, 3- to 14-membered heterocycloalkyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, halo-C6-C14-aryl, and C6-C14-aryloxy;
      • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, C1-6-alkoxy, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl, 5- to 14-membered heteroaryl and halo-C6-C14-aryl;
      • R6 is hydrogen or halogen;
      • R7 is selected from hydrogen, C1-6-alkyl, C6-C14-aryl, halo-C6-C14-aryl, halo-C6-C14-aryloxy, and halo-C1-6-alkyl;
      • R8 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl;
      • R9 is hydrogen;
      • R10 is halogen;
      • R11 is halogen;
      • R12 is hydrogen;
      • R14 is selected from halogen, C1-6-alkyl, and C1-6-alkoxy;
      • R15 is hydrogen or halogen; and
      • R16 and R17, taken together with the carbon atom to which they are attached, form a C3-10-cycloalkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
      • p is 1 or 2;
      • A is selected from:
        • (i) C6-C14-aryl substituted with R4, R5 and R6; and
        • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9;
      • R1 is hydrogen;
      • R2 is hydrogen or C1-6-alkyl;
      • R3 is selected from hydrogen, halogen and C1-6-alkyl;
      • R4 is selected from halogen, SF5, C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, and 3- to 14-membered heterocycloalkyl;
      • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl and halo-C6-C14-aryl;
      • R6 is hydrogen;
      • R7 is C1-6-alkyl or halo-C1-6-alkyl;
      • R8 is hydrogen or halo-C1-6-alkyl; and
      • R9 is hydrogen.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
      • p is 1 or 2;
      • A is selected from:
        • (i) phenyl substituted with R4, R5 and R6;
        • (ii) oxazolyl substituted with R7, R8 and R9; and
        • (iii) pyridyl substituted with R7, R8 and R9;
      • R1 is hydrogen;
      • R2 is hydrogen or methyl;
      • R3 is selected from hydrogen, fluoro and methyl;
      • R4 is selected from chloro, SF5, methyl, methoxy, OCF3, CF3, cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl;
      • R5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF3, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl;
      • R6 is hydrogen:
      • R7 is selected from tert-butyl, methyl, and CF3;
      • R8 is hydrogen or CF3; and
      • R9 is hydrogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3; and
      • R3 is selected from hydrogen, halogen and C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3; and
      • R3 is selected from hydrogen, fluoro and methyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • (i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —(CR16R17)q—CH2O—, —CH2OCH2—, —CF2CH2—, and —CH2═CH2—; or
      • (ii) X is N; m and n are both 1; and L is —(CH2)p—;
      • p is 1 or 2;
      • q is 0 or 1;
      • R3 is selected from hydrogen, halogen and C1-6-alkyl; and
      • to R16 and R17, taken together with the carbon atom to which they are attached, form a C3-10-cycloalkyl.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—:
      • p is 1 or 2; and
      • R3 is selected from hydrogen, halogen and C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—R3;
      • m and n are both 0; or
      • m and n are both 1;
      • L is selected from —(CH)p—, —O—, —OCH2— and —CH2O—;
      • p is 1 or 2; and
      • R3 is selected from hydrogen, fluoro and methyl.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • A is selected from:
        • (i) C6-C14-aryl substituted with R4, R5 and R6;
        • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9; and
        • (iii) 3- to 14-membered heterocycloalkyl substituted with R10, R11 and R12;
      • R4 is selected from hydrogen, halogen, cyano, SF5, C1-6-alkyl, C1-6-alkanoyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, 3- to 14-membered heterocycloalkyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, to 14-membered heteroaryl, halo-C6-C14-aryl, and C6-C14-aryloxy
      • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, C1-6-alkoxy, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl, 5- to 14-membered heteroaryl and halo-C6-C14-aryl;
      • R6 is hydrogen or halogen;
      • R7 is selected from hydrogen, C1-6-alkyl, C6-C14-aryl, halo-C6-C14-aryl, halo-C6-C14-aryloxy, and halo-C1-6-alkyl;
      • R8 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl;
      • R9 is hydrogen;
      • R10 is halogen;
      • R11 is halogen;
      • R12 is hydrogen;
      • R14 is selected from halogen, C1-6-alkyl, and C1-6-alkoxy; and
      • R15 is hydrogen or halogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • A is selected from:
        • (i) C6-C14-aryl substituted with R4, R5 and R6; and
        • (ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9;
      • R4 is selected from halogen, SF5, C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, and 3- to 14-membered heterocycloalkyl;
      • R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl and halo-C6-C14-aryl;
      • R6 is hydrogen;
      • R7 is C1-6-alkyl or halo-C1-6-alkyl;
      • R8 is hydrogen or halo-C1-6-alkyl; and
      • R9 is hydrogen.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • A is selected from:
        • (i) phenyl substituted with R4, R5 and R6;
        • (ii) oxazolyl substituted with R7, R8 and R9; and
        • (iii) pyridyl substituted with R7, R8 and R9;
      • R4 is selected from chloro, SF5, methyl, methoxy, OCF3, CF3, cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl;
      • R5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF3, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl;
      • R6 is hydrogen;
      • R7 is tert-butyl, methyl, and CF3;
      • R8 is hydrogen or CF3; and
      • R9 is hydrogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—H;
      • m and n are both 0;
      • L is —CH2O—;
      • A is C6-C14-aryl substituted with R4, R5 and R6;
      • R1, R2, and R6 are all hydrogen;
      • R4 is halo-C1-6-alkyl; and
      • R5 is halogen or C1-6-alkyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—H;
      • m and n are both 0:
      • L is —CH2O—:
      • A is C6-C14-aryl substituted with R4, R5 and R6;
      • R1, R2, and R6 are all hydrogen;
      • R4 is halo-C1-6-alkyl; and
      • R5 is halogen.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—H;
      • m and n are both 0;
      • L is —CH2O—;
      • A is phenyl substituted with R4, R5 and R6;
      • R1, R2, and R6 are all hydrogen;
      • R4 is CF3; and
      • R5 is fluoro or methyl.
  • In a particularly preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:
      • X is C—H:
      • m and n are both 0;
      • L is —CH2O—;
      • A is phenyl substituted with R4, R5 and R6;
      • R1, R2, and R6 are all hydrogen;
      • R4 is CF3; and
      • R5 is fluoro.
  • In one embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from the compounds disclosed in Table 1.
  • In a preferred embodiment, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from:
    • (+)-(4aR,8aS)-6-(4-((4-(tert-Butyl)oxazol-2-yl)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)-(4aR,8aS)-6-[4-[[4-(Trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (+)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • rac-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4-fluorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-(4-(Trifluoromethoxy)benzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-(4-Chloro-3-fluorobenzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-(2-Chloro-4-(trifluoromethyl)phenoxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-(3-(Trifluoromethyl)phenoxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-[4-[[2-Chloro-4-(trifluoromethoxy)phenoxy]methyl]piperidine-1-carbonyl]4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4(trifluoromethyl)phenoxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-]1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6(4((4-Chloro-2-fluorophenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((4-Fluoro-2-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((2-Fluoro-4-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-(2-(Pyrrolidin-1-yl)-4-(trifluoromethyl)benzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((2-Chloro-4-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-[4-[[2-Cyclopentyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (+)- or (−)-3-Chloro-4-((1-((4aR,8aS)-3-oxooctahydro-2H-pyrido[4,3-b][1,4]oxazine-6-carbonyl)piperidin-4-yl)methoxy)benzonitrile;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4(trifluoromethyl)phenoxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((4′,6-Dichloro-[1,1′-biphenyl]-3-yl)oxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(cis-4-((2-Chloro-4-(trifluorophenoxy)methyl)-3-methylpiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethoxy)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (+)- or (−)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)-4-methylpiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-[3-[(2,4-Dichlorophenyl)methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[4-[[2-Fluoro-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[4-[[2-Cyclopropyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[[3-Chloro-4-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[[2-Fluoro-5-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[2-[2-Fluoro-6-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-(3-(2-fluoro-4-(trifluoromethyl)phenethyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • 6-(3-((2,4-bis(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-[4-[3-chloro-4-(trifluoromethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-(3-methyl-4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-(3-((3,4-dichlorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-(3-((2,5-dichlorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • rac-(4aR,8aS)-6-(2-methyl-3-((4-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-(3-(((4,5-bis(trifluoromethyl)pyridin-2-yl)oxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • rac-(4aR,8aS)-6-(3-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)-2-methylazetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-(3-((2-fluoro-4-(pentafluoro-16-sulfaneyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxain-3(4H)-one;
    • (4aR,8aS)-6-(3-((4-methyl-2-(trifluoromethoxy)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
    • (4aR,8aS)-6-[4-[3-cyclopropyl-4-(trifluoromethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[2-(2-fluoro-4-methyl-phenyl)ethyl]azetidine-1-carbonyl-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[2-[4-methoxy-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[3-(2-azaspiro[3.3]heptan-2-yl)-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[2-[4-methy-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-(3-((4-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxain-3(4H)-one;
    • (4aR,8aS)-6-(3-((2-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b[[1,4]oxazin-3(4H)-one;
    • rac-(4aR,8aS)-6-[2-methyl-3-[[2-methyl-4-(trifluoromethoxy)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • rac-(4aR,8aS)-6-[2-methyl-3-[[2-methyl-3-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-(4-chloro-3-cyclopropyl-phenoxy)azetidine-1-carbonyl]-4,4#a!,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[4-[2-cloro-3-(trifluoroethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-(2-chloro-3-cyclopropyl-phenoxy)azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[3-(2-azaspiro[3.3]heptan-2-yl)-2-chloro-phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[2-chloro-3-(5-oxa-2-azaspiro[3.5]nonan-2-yl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-[3-[(E)-2-(2-fluoro-4-methyl-phenyl)vinyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
    • (4aR,8aS)-6-(3-((E)-2-fluoro-6-(trifluoromethyl)styryl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one; and
    • (4aR,8aS)-6-(3-((4-methyl-3-(trifluoromethyl)benzyl)ox)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one.
  • In one embodiment, the present invention provides pharmaceutically acceptable salts or esters of the compounds of formula (I) as described herein. In a particular embodiment, the present invention provides pharmaceutically acceptable salts of the compounds according to formula (I) as described herein, especially hydrochloride salts. In a further particular embodiment, the present invention provides pharmaceutically acceptable esters of the compounds according to formula (I) as described herein. In yet a further particular embodiment, the present invention provides compounds according to formula (I) as described herein.
  • In some embodiments, the compounds of formula (I) are isotopically-labeled by having one or more atoms therein replaced by an atom having a different atomic mass or mass number. Such isotopically-labeled (i.e., radiolabeled) compounds of formula (I) are considered to be within the scope of this disclosure. Examples of isotopes that can be incorporated into the compounds of formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as, but not limited to, 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36CL, 123I, and 125I, respectively. Certain isotopically-labeled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e., 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. For example, a compound of formula (I) can be enriched with 1, 2, 5, 10, 25, 50, 75, 90, 95, or 99 percent of a given isotope.
  • Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements.
  • Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • Processes of Manufacturing
  • The preparation of compounds of formula (I) of the present invention may be carried out in sequential or convergent synthetic routes. Syntheses of the invention are shown in the following general schemes. The skills required for carrying out the reaction and purification of the resulting products are known to those persons skilled in the art. The substituents and indices used in the following description of the processes have the significance given herein, unless indicated to the contrary.
  • If one of the starting materials, intermediates or compounds of formula (I) contain one or more functional groups which are not stable or are reactive under the reaction conditions of one or more reaction steps, appropriate protective groups (as described e.g., in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014. John Wiley & Sons, N.Y.) can be introduced before the critical step applying methods well known in the art. Such protective groups can be removed at a later stage of the synthesis using standard methods described in the literature.
  • If starting materials or intermediates contain stereogenic centers, compounds of formula (I) can be obtained as mixtures of diastereomers or enantiomers, which can be separated by methods well known in the art e.g., chiral HPLC, chiral SFC or chiral crystallization. Racemic compounds can e.g., be separated into their antipodes via diastereomeric salts by crystallization with optically pure acids or by separation of the antipodes by specific chromatographic methods using either a chiral adsorbent or a chiral eluent. It is equally possible to separate starting materials and intermediates containing stereogenic centers to afford diastereomerically/enantiomerically enriched starting materials and intermediates. Using such diastereomerically/enantiomerically enriched starting materials and intermediates in the synthesis of compounds of formula (I) will typically lead to the respective diastereomerically/enantiomerically enriched compounds of formula (I). A person skilled in the art will acknowledge that in the synthesis of compounds of formula (I)—insofar not desired otherwise—an “orthogonal protection group strategy” will be applied, allowing the cleavage of several protective groups one at a time each without affecting other protective groups in the molecule. The principle of orthogonal protection is well known in the art and has also been described in literature (e.g. Barany and R. B. Merrifield, J. Am. Chem. Soc. 1977, 99, 7363; H. Waldmann et al., Angew. Chem. Int. Ed. Engl. 1996, 35, 2056).
  • A person skilled in the art will acknowledge that the sequence of reactions may be varied depending on reactivity and nature of the intermediates.
  • In more detail, the compounds of formula (I) can be manufactured by the methods given below, by the methods given in the examples or by analogous methods. Appropriate reaction conditions for the individual reaction steps are known to a person skilled in the art. Also, for reaction conditions described in literature affecting the described reactions see for example: Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 2nd Edition, Richard C. Larock, John Wiley & Sons, New York, N.Y. 1999). It was found convenient to carry out the reactions in the presence or absence of a solvent. There is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or the reagents involved and that it can dissolve the reagents, at least to some extent. The described reactions can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. It is convenient to carry out the described reactions in a temperature range between −78° C. to reflux. The time required for the reaction may also vary widely, depending on many factors, notably the reaction temperature and the nature of the reagents. However, a period of from 0.5 hours to several days will usually suffice to yield the described intermediates and compounds. The reaction sequence is not limited to the one displayed in the schemes, however, depending on the starting materials and their respective reactivity, the sequence of reaction steps can be freely altered.
  • If starting materials or intermediates are not commercially available or their synthesis not described in literature, they can be prepared in analogy to existing procedures for close analogues or as outlined in the experimental section.
  • The following abbreviations are used in the present text:
  • AcOH=acetic acid, ACN=acetonitrile, Bn=benzyl, Boc=tert-butyloxycarbonyl, CAS RN=chemical abstracts registration number, Cbz=benzyloxycarbonyl, Cs2CO3=cesium carbonate, CO=carbon monoxide, CuCl=copper(I) chloride, CuCN=copper(I) cyanide, CuI=copper(I) iodide, DAST=(diethylamino)sulfur trifluoride, DBU=1,8-diazabicyclo[5,4,0]undec-7-ene, DCM=dichloromethane, DEAD=diethyl azodicarboxylate, DIAD=diisopropyl azodicarboxylate, DMAP=4-dimethylaminopyridine, DME=dimethoxyethane, DMEDA=N,N′-dimethylethylenediamine, DMF=N,N-dimethylformamide, DIPEA=N,N-diisopropylethylamine, dppf=1,1 bis(diphenyl phosphino)ferrocene, EDC.HCl=N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride, EI=electron impact, ESI=electrospray ionization, EtOAc=ethyl acetate, EtOH=ethanol, h=hour(s), FA=formic acid, H2O=water, H2SO4=sulfuric acid, HATU=1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium-3-oxide hexafluorophosphate, HBTU=O-benzotriazole-N,N,N′,N′-tetramethyl-uronium-hexafluoro-phosphate, HCl=hydrogen chloride, HOBt=1-hydroxy-1H-benzotriazole; HPLC=high performance liquid chromatography, iPrMgCl=isopropylmagnesium chloride, I2=iodine, IPA=2-propanol, ISP=ion spray positive (mode), ISN=ion spray negative (mode), K2CO3=potassium carbonate, KHCO3=potassium bicarbonate, KI=potassium iodide, KOH=potassium hydroxide, K3PO4=potassium phosphate tribasic, LiAlH4 or LAH=lithium aluminium hydride, LiHMDS=lithium bis(trimethylsilyl)amide, LiOH=lithium hydroxide, MgSO4=magnesium sulfate, min=minute(s), mL=milliliter, MPLC=medium pressure liquid chromatography, MS=mass spectrum, nBuLi=n-butyllithium, NaBH3CN=sodium cyanoborohydride, NaH=sodium hydride, NaHCO3=sodium hydrogen carbonate, NaNO2=sodium nitrite, NaBH(OAc)3=sodium triacetoxyborohydride, NaOH=sodium hydroxide, Na2CO3=sodium carbonate, Na2SO4=sodium sulfate, Na2S2O3=sodium thiosulfate, NBS=N-bromosuccinimide, nBuLi=n-butyllithium, NEt3=triethylamine (TEA), NH4Cl=ammonium chloride, NMP=N-methyl-2-pyrrolidone, OAc=Acetoxy, T3P=propylphosphonic anhydride, PE=petroleum ether, PG=protective group, Pd—C=palladium on activated carbon, PdCl2(dppf)-CH2Cl2=1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex, Pd2(dba)3=tris(dibenzylideneacetone)dipalladium(0), Pd(OAc)2=palladium(II) acetate, Pd(OH)2=palladium hydroxide, Pd(PPh3)4=tetrakis(triphenylphosphine)palladium(0), PTSA=p-toluenesulfonic acid, R=any group, RT=room temperature, SFC=Supercritical Fluid Chromatography, S-PHOS=2-dicyclohexylphosphino-2′,6′-dimethoxybiphenyl, TBAI=tetra butyl ammonium iodine, TEA=triethylamine, TFA=trifluoroacetic acid, THF=tetrahydrofuran, TMEDA=N,N,N′,N′-tetramethylethylenediamine, ZnCl2=zinc chloride, Hal=halogen.
  • Compounds of formula I wherein A, L, X, m, n, R1 and R2 are as described herein can be synthesized in analogy to literature procedures and/or as depicted for example in Scheme 1.
  • Figure US20200308190A1-20201001-C00010
  • Accordingly, 4a,5,6,7,8,8a-hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-ones 1 are reacted with intermediates 2 in the presence of a urea forming reagent, such as bis(trichloromethyl) carbonate using a suitable base and solvent such as, e.g. sodium bicarbonate in DCM, to give compounds of formula I (step a). Further urea forming reagents include but are not limited to phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate or 1,1′-carbonyldiimidazole. Reactions of this type and the use of these reagents are widely described in literature (e.g. G. Sartori et al., Green Chemistry 2000, 2, 140). A person skilled in the art will acknowledge that the order of the addition of the reagents can be important in this type of reactions due to the reactivity and stability of the intermediary formed carbamoyl chlorides, as well as for avoiding formation of undesired symmetrical urea by-products.
  • Intermediates 1 may be synthesized as depicted for example in Scheme 2 and/or in analogy to methods described in literature.
  • Figure US20200308190A1-20201001-C00011
  • Thus, 3-aminopiperidin-4-ol derivatives 3 in which “PG” signifies a suitable protective group such as a Cbz or Boc protective group can be acylated for example with acyl chlorides 4 in which R1 is as defined herein and “LG” signifies a suitable leaving group (e.g., Cl or Br), using a suitable base such as sodium or potassium carbonate, sodium hydroxide or sodium acetate in an appropriate solvent such as THF, water, acetone or mixtures thereof, to provide intermediates 5 (step a). Intermediates 4 are either commercially available or can be prepared according to literature methods in achiral (R1=H), racemic (R1 not H) or enantiomerically pure form (R1 not H).
  • Intermediates 5 can be cyclized to intermediates 6 using methods well known in the art, for example by treatment of 5 with sodium hydride in THF or potassium tert-butoxide in IPA and water (step b). Reactions of that type are described in literature (e.g. Z. Rafinski et al., J. Org. Chem. 2015, 80, 7468; S. Dugar et al., Synthesis 2015, 47(5), 712; WO2005/066187).
  • Removal of the protective group in intermediates 6, applying methods known in the art (e.g., a Boc group using TFA in DCM at temperatures between 0° C. and room temperature, a Cbz group using hydrogen in the presence of a suitable catalyst such as Pd or Pd(OH)2 on charcoal in a suitable solvent such as MeOH, EtOH, EtOAc or mixtures thereof and as described for example in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wuts, 4th Ed., 2006, Wiley N.Y.), furnishes intermediates 1 (step c).
  • Intermediates 1 can be obtained as mixtures of diastereomers and enantiomers, respectively, or as single stereoisomers depending on whether racemic mixtures or enantiomerically pure forms of cis- or trans-3-aminopiperidin-4-ol derivatives 3 or intermediates 4 are employed in their syntheses. Intermediates 3 are commercially available and their synthesis has also been described in literature (e.g. WO2005/066187; WO2011/0059118; WO2016/185279). Optically pure cis-configured intermediates 1B and 1C can be obtained for example according to Scheme 3 by chiral separation of commercially available rac-(4aR,8aS)-4a,5,6,7,8,8a-hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one (1A) (optionally in form of a salt such as, e.g. a hydrochloride salt) using methods known in the art, e.g. by diastereomeric salt crystallization or by chiral chromatography (step a).
  • Figure US20200308190A1-20201001-C00012
  • In some embodiments, intermediates 2 are intermediates of type B. Intermediates of type B in which A, m, n and R2 are as described herein can be prepared by methods well known by a person skilled in the art and as exemplified by the general synthetic procedures outlined in Scheme 4.
  • Figure US20200308190A1-20201001-C00013
  • Ketones 7, either commercially available or prepared by methods known in the art, can be subjected for example to a Wittig reaction with alkylidene triphenylphosphoranes of type 8a in a suitable solvent such as, e.g. THF. Methyl-THF or DMSO to give intermediates 9 (step a). Phosphoranes 8a can be formed by treating the corresponding phosphonium salts with a suitable base such as BuLi. NaH, or KOtBu in a suitable solvent such as THF, dioxane or Methyl-THF and may be isolated or used in situ. Phosphonium salts in turn are readily available from an aryl/heteroaryl/heterocyclic-substituted alkylhalide (with halide being Cl, Br and iodo) and triphenylphosphine in a suitable solvent such as toluene. Heating may be applied to accelerate the reaction or drive the reaction to completion (e.g. H. J. Cristau, F. Plénat in PATAI'S Chemistry of Functional Groups, Editor(s): Frank R. Hartley, 07th August 2006, Series Editor(s): Prof Saul Patai).
  • Alternatively, intermediates 9 can be obtained using a Homer-Wadsworth-Emmons (HWE) reaction using ketones 7 and phosphonates 8b, wherein Ra is alkyl, for example methyl or ethyl. Phosphonates 8b are in situ α-metalated using a suitable base and solvent such as NaH, nBuLi or KOtBu in THF (step a). Phosphonates 8b are readily prepared using for example the Arbuzov reaction by alkylation of an aryl/heteroaryl/heterocyclic halide (with halide being Cl, Br and iodo) with commercially available trialkyl phosphite (e.g. Chem. Rev. 1984, 84, 577).
  • Olefination reactions of both types are broadly described in literature (e.g. Current Org. Chem. 2015, 19(9), page 744; Chem. Rev. 1989, 89(4), 863; Org. React. 1977, 25, 73; Liebigs Ann./Recueil 1997, 1283; Acc. Chem. Res. 1983, 16, 411).
  • Reduction of the double bond in intermediates 9 using, e.g. hydrogen in the presence of a suitable catalyst such as palladium on charcoal in an appropriate solvent or solvent mixture such as EtOAc, MeOH or AcOH yields compounds 10 (step b).
  • Removal of the protective group from intermediates 10 applying methods known in the art (e.g., a Boc group using TFA in DCM or 4M HCl in dioxane at temperatures between 0° C. and room temperature, a Cbz group using hydrogen in the presence of a suitable catalyst such as Pd or Pd(OH)2 on charcoal in a suitable solvent such as MeOH, EtOH, EtOAc or mixtures thereof and as described for example in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wuts, 4th Ed., 2006. Wiley N.Y.), furnishes intermediates B (step c).
  • In some embodiments, intermediates 2 are intermediates of type C. Intermediates of type C in which A, R2 and p are as described herein, r=0, 1 or 2 and (m+n)=2 or 3 can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 5.
  • Figure US20200308190A1-20201001-C00014
  • Alkylation of optionally mono-protected piperazine or 1,4-diazepane derivatives 11a,b (commercially available or synthesized in analogy to literature methods) with aryl/heteroaryl/heterocyclyl-substituted alkyl derivatives 12, either commercially available or synthesized according to literature procedures and in which LG signifies a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. mesylate (methanesulfonate), OSO2fluoroalkyl (e.g. triflate (trifluoromethanesulfonate) or OSO2aryl (e.g. tosylate (p-toluenesulfonate using a suitable base in an appropriate solvent (e.g. sodium hydride in DMF) at temperatures between 0° C. and the boiling temperature of the solvent, gives intermediates 14a,b (step a).
  • Alternatively, compounds 11a,b can be subjected to a reductive amination reaction with aldehydes of type 13 using a suitable reducing agent and solvent such as NaBH3CN in MeOH, AcOH or mixtures thereof to give intermediates 14a,b (step a).
  • Removal of the protective group from intermediates 14b applying methods known in the art (e.g., a Boc group using TFA in DCM or 4M HCl in dioxane at temperatures between 0° C. and room temperature, a Cbz group using hydrogen in the presence of a suitable catalyst such as Pd or Pd(OH)2 on charcoal in a suitable solvent such as MeOH, EtOH, EtOAc or mixtures thereof and as described for example in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wuts, 4th Ed., 2006. Wiley N.Y.), furnishes intermediates C (step c).
  • In some embodiments, compounds of formula I are compounds of type Ie. Compounds Ie in which A, p, R1 and R2 are defined as herein and (m+n)=2 or 3 can be prepared in analogy to literature procedures or the methods described under Scheme 6 below.
  • Figure US20200308190A1-20201001-C00015
  • Compounds 1 can be coupled with piperazine or 1,4-diazepane derivatives 11a applying for example the conditions outlined under Scheme 1, step a, to give intermediates 15 (step a).
  • Intermediates 15 can be converted to compounds IC in analogy to the procedure described under Scheme 5, step a (step b).
  • Alternatively, compounds 1 can be coupled with mono-protected piperazine or 1,4-diazepane derivatives lib in which PG signifies a suitable protective group such as a Cbz or Boc protective group applying for example the conditions outlined under Scheme 1, step a, to give intermediates 16 (step c).
  • Removal of the protective group by published methods or as described under Scheme 5, step c, furnishes intermediates 15 (step d).
  • In some embodiments, intermediates 2 are intermediates of type D. Intermediates of type D in which A, m, n and R2 are as described herein and R3 is selected from hydrogen, halogen, C1-6-alkoxy, C1-6-alkyl and halo-C1-6-alkyl, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 7.
  • Figure US20200308190A1-20201001-C00016
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 18 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 19 (step a). Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739; Chem. Rev. 2009, 109 (6), 2551).
  • Removal of the protective group from intermediates 19 applying literature methods and as described for example under Scheme 3, step c, furnishes intermediates D (step b).
  • Alternatively, intermediates 19 may be prepared from alcohols 17 that can be alkylated with compounds 20 in which LG is a suitable leaving group such as chlorine, bromine, iodine. OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate using a suitable base in an appropriate solvent (e.g. sodium hydride in DMF) at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • Furthermore, intermediates 19 may be synthesized via alkylation of alcohols of type 18 with compounds 21 under the conditions described under step c (step d).
  • In another embodiment, intermediates 2 are intermediates of type E. Intermediates of type E in which A, m, n, R2 and R3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 8. In case R3 is a hydroxy group a suitable protective group strategy known to those skilled in the art may be applied.
  • Figure US20200308190A1-20201001-C00017
  • The carboxylic acid functionality in derivatives 22 in which PG signifies a suitable protecting group such as, e.g. a Boc, Cbz or Bn protecting group, either commercially available or prepared by methods known in the art, can be converted into an acid chloride (LG=Cl) or Weinreb amide (LG=NMeOMe) by applying methods broadly described in literature to give intermediates 23 (step a).
  • Intermediates 23 can be reacted with compounds of type 24, either commercially available or synthesized by methods known in the art and as described below to yield intermediates 25 (step b).
  • If compounds 24 are commercially not available they can be prepared in analogy to literature methods. For example, deprotonation of a reactive methyl group in optionally substituted heterocycles 27 using an appropriate base such nBuLi or LiHMDS in a suitable solvent, e.g. THF, hexane or mixtures thereof, at temperatures ranging from −78° C. to room temperature, gives intermediates 24 in which MX=Li (step d).
  • Compounds 24 in which MX=MgHal with Hal being Cl, Br or I (Grignard reagents) may be prepared by reaction of the corresponding substituted benzyl halides 28 with magnesium in a suitable solvent such as THF, optionally in the presence of catalytic amounts of iodine at temperatures ranging from 0° C. to the boiling point of the solvent (step d).
  • Compounds 25 can be further converted into compounds 26 by a deoxyfluorination reaction using a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine trihydrofluoride and TEA or DBU) in a suitable solvent such as DCM or ACN (step d).
  • Removal of the protective group from intermediates 26 applying literature methods and as described for example under Scheme 3, step c, furnishes intermediates E (step e).
  • In a further embodiment, intermediates 2 are intermediates of type F. Intermediates F in which A, m, n, R2 and R3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 9. In case R3 is a hydroxy group a suitable protective group strategy known to those skilled in the art may be applied.
  • Figure US20200308190A1-20201001-C00018
  • The carboxylic acid functionality in intermediates 29, either commercially available or prepared by methods known in the art, in which PG signifies a suitable protecting group such as, e.g. a Boc, Cbz or Bn protecting group, can be converted for example into an acid chloride (LG=Cl) or Weinreb amide (LG=NMeOMe) by applying methods broadly described in literature to give intermediates 30 (step a).
  • Intermediates 30 can be reacted with compounds of type 31, either commercially available or synthesized by methods known in the art and as described below to yield intermediates 32 (step b).
  • In case compounds 31 are commercially not available they can be prepared in analogy to literature methods. For example, deprotonation of optionally substituted aryl or heteroaryl rings 33 using an appropriate base such n-BuLi, sec-BuLi, tert-BuLi, LiHMDS, NaH, KH in a suitable solvent, such as THF, n-hexane or mixtures thereof, at temperatures ranging from −78° C. to room temperature, gives intermediates 31 in which, depending on the base used, MX=Li, Na or K (step c).
  • Compounds 31 in which MX=MgHal with Hal being Cl, Br or I (Grignard reagents) may be prepared by reaction of the corresponding optionally substituted aryl or heteroaryl halides 34 via direct insertion of magnesium (e.g. magnesium turnings optionally in the presence of catalytic amounts of iodine, powder in the presence of LiCl or Rieke magnesium, organic halides) or by halogen-magnesium exchange by treating 34 in which Hal is preferably bromine or iodine, with an alkylmagnesium halide such as iPrMgCl (optionally in the presence of LiCl) in suitable solvents such as diethyl ether or THF at temperatures ranging from 0° C. to the boiling point of the solvent (step d).
  • Compounds 32 can be further converted into compounds 35 by a deoxyfluorination reaction using a suitable fluorinating agent such as DAST, Deoxo-Fluor (bis(2-methoxyethyl)aminosulfur trifluoride) or aminodifluorosulfinium tetrafluoroborates (XtalFluor-E®, XtalFluor-M® in the presence of, e.g. triethylamine trihydrofluoride and TEA or DBU) in a suitable solvent such as DCM or ACN (step e).
  • Removal of the protective group from intermediates 35 applying literature methods and as described for example under Scheme 3, step c, furnishes intermediates F (step f).
  • In some embodiments, intermediates 2 are intermediates of type G. Intermediates of type G in which A, m, n, R2 are as described herein and R3 is hydrogen, C1-6-alkoxy, C1-6-alkyl and halo-C1-6-alkyl, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 10.
  • Figure US20200308190A1-20201001-C00019
  • Intermediates 38 may be prepared from alcohols 36 in which PG is a suitable protective group such as a Cbz, Boc or Bn, that can be alkylated with compounds 37 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base, such as sodium hydride, potassium tert-butoxide, in an appropriate solvent (e.g. in DMF or THF) at temperatures between 0° C. and the boiling temperature of the solvent (step a).
  • Removal of the protective group from intermediates 38 applying literature methods and as described for example under Scheme 4, step c, furnishes intermediates G (step b).
  • In some embodiments, intermediates 2 are intermediates of type H. Intermediates of type H in which A, m, n, R2 and R3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 11.
  • Figure US20200308190A1-20201001-C00020
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 39 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 41 (step a). Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739; Chem. Rev. 2009, 109 (6), 2551).
  • Removal of the protective group from intermediates 41 applying literature methods and as described for example under Scheme 4, step c, furnishes intermediates H (step b).
  • Alternatively, intermediates 41 may be prepared from alcohols 17 that can be alkylated with compounds 40 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs2CO3, NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • Reacting intermediates H with intermediates 1, for example using the conditions described under scheme 1, step a, affords compounds of type If, wherein A, R1, R2, R3, m and n are as defined herein.
  • Figure US20200308190A1-20201001-C00021
  • Alternatively, compounds of type If may be prepared according to Scheme 12.
  • Figure US20200308190A1-20201001-C00022
  • Alcohols of type 17 can be subjected to a Mitsunobu reaction with intermediates 42, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give compounds ID (step a). Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739; Chem. Rev. 2009, 109 (6), 2551).
  • Alternatively, compounds ID may be directly prepared from alcohols 17 that can be alkylated with compounds 43 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs2CO3, NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step b).
  • In some embodiments, intermediates 2 are intermediates of type J. Intermediates of type J in which A, m, n, R2 and R3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 13.
  • Figure US20200308190A1-20201001-C00023
  • Intermediates 46 may be prepared from heterocycloalkyls or heteroaryls 45 that can be alkylated with compounds 44 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs2CO3, K2CO3, NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step a).
  • Removal of the protective group from intermediates 46 applying literature methods and as described for example under Scheme 4, step c, furnishes intermediates J (step b).
  • Reacting intermediates J with intermediates 1 affords compounds of type Ig, wherein A, R1, R2, R3, m and n are as defined herein.
  • Figure US20200308190A1-20201001-C00024
  • Alternatively, compounds of type Ig may be directly prepared from heterocycloalkyls or heteroaryls 45 that can be alkylated with compounds 47 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs2CO3, K2CO3, NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (Scheme 14).
  • Figure US20200308190A1-20201001-C00025
  • In some embodiments, intermediates 2 are intermediates of type K. Intermediates of type K in which A, m, n, R2 and R3 are as described herein, can be prepared by methods well known in the art and as exemplified by the general synthetic procedures outlined in Scheme 15.
  • Figure US20200308190A1-20201001-C00026
  • Alcohols of type 48 can be subjected to a Mitsunobu reaction with intermediates 39 in which PG is a suitable protective group such as a Cbz, Boc or Bn, using an appropriate phosphine such as triphenylphosphine and a dialkyl azodicarboxylate such as DEAD or DIAD in a suitable solvent such as THF to give intermediates 49 (step a). Mitsunobu reactions of that type are broadly described in literature (e.g. Org. Chem. Front. 2015, 2, 739; Chem. Rev. 2009, 109 (6), 2551).
  • Removal of the protective group from intermediates 49 applying literature methods and as described for example under Scheme 4, step c, furnishes intermediates K (step b).
  • Alternatively, intermediates 49 may be prepared from alcohols 48 that can be alkylated with compounds 40 in which LG is a suitable leaving group such as chlorine, bromine, iodine, OSO2alkyl (e.g. methanesulfonate), OSO2fluoroalkyl (e.g. trifluoromethanesulfonate) or OSO2aryl (e.g. p-toluenesulfonate) using a suitable base such as Cs2CO3, NaH, in an appropriate solvent, such as DMF at temperatures between 0° C. and the boiling temperature of the solvent (step c).
  • In some embodiments, intermediates 2 are intermediates of type L. Intermediates of type L in which A, m, n, R2 and R3 are as described herein can be prepared by methods well known by a person skilled in the art and as exemplified by the general synthetic procedures outlined in Scheme 16.
  • Figure US20200308190A1-20201001-C00027
  • Intermediates 51 can be prepared for example from aldehydes 50, either commercially available or prepared by methods known in the art, using a Wittig reaction or Homer-Wadsworth-Emmons (HWE) reaction using alkylidene triphenylphosphoranes of type 8a and phosphonates 8b, respectively, as described under step a in Scheme 4 (step a).
  • Reduction of the double bond in intermediates 51 applying the conditions described under step b in Scheme 4 yields compounds 52 (step b).
  • Removal of the protective group from intermediates 52 applying methods known in the art and as outlined under step c in Scheme 4 furnishes intermediates L (step c).
  • Removal of the protective group from intermediates 51 applying methods known in the art and as outlined under step c in Scheme 4 furnishes intermediates M (step d).
  • In one aspect, the present invention provides a process of manufacturing the urea compounds of formula (I) described herein, comprising:
      • reacting a first amine of formula 1, wherein R1 is as described herein, preferably wherein R1 is hydrogen,
  • Figure US20200308190A1-20201001-C00028
      • with a second amine 2, wherein A, L, m, n, X and R2 are as described herein
  • Figure US20200308190A1-20201001-C00029
      • in the presence of a base and a urea forming reagent,
  • to form said compound of formula (I).
  • In one embodiment, there is provided a process according to the invention, wherein said base is sodium bicarbonate.
  • In one embodiment, there is provided a process according to the invention, wherein said urea forming reagent is selected from bis(trichloromethyl) carbonate, phosgene, trichloromethyl chloroformate, (4-nitrophenyl)carbonate and 1,1′-carbonyldiimidazole, preferably wherein said urea forming reagent is bis(trichloromethyl) carbonate.
  • In one aspect, the present invention provides a compound of formula (I) as described herein, when manufactured according to any one of the processes described herein.
  • MAGL Inhibitory Activity
  • Compounds of the present invention are MAGL inhibitors. Thus, in one aspect, the present invention provides the use of compounds of formula (I) as described herein for inhibiting MAGL in a mammal.
  • In a further aspect, the present invention provides compounds of formula (I) as described herein for use in a method of inhibiting MAGL in a mammal.
  • In a further aspect, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for inhibiting MAGL in a mammal.
  • In a further aspect, the present invention provides a method for inhibiting MAGL in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • Compounds were profiled for MAGL inhibitory activity by measuring the enzymatic activity of MAGL by following the hydrolysis of 4-nitrophenylacetate resulting in 4-nitrophenol, which absorbs at 405-412 nm (G. G. Muccioli, G. Labar, D. M. Lambert, Chem. Bio. Chem. 2008, 9, 2704-2710). This assay is hereinafter abbreviated “4-NPA assay”.
  • The 4-NPA assay was carried out in 384 well assay plates (black with clear bottom, non-binding surface treated, Coming Ref 3655) in a total volume of 40 μL. Compound dilutions were made in 100% DMSO (VWR Chemicals 23500.297) in a polypropylene plate in 3-fold dilution steps to give a final concentration range in the assay from 25 μM to 1.7 nM. 1 μL compound dilutions (100% DMSO) were added to 19 μL MAGL (recombinant wild-type) in assay buffer (50 mM TRIS (GIBCO, 15567-027), 1 mM EDTA (Fluka, 03690-100 ml)). The plate was shaked for 1 min at 2000 rpm (Variomag Teleshake) and then incubated for 15 min at RT. To start the reaction, 20 μL 4-Nitrophenlyacetate (Sigma N-8130) in assay buffer with 6% EtOH was added. The final concentrations in the assay were 1 nM MAGL and 300 μM 4-Nitrophenylacetate. After shaking (1 min, 2000 rpm) and 5 min incubation at RT, the absorbance at 405 nm was measured for a fist time (Molecular Devices, SpectraMax Paradigm). A second measurement was then done after incubation for 80 min at RT. From the two measurements, the slope was calculated by subtracting the first from the second measurement.
  • Alternatively, compounds were profiled for MAGL inhibitory activity by determining the enzymatic activity by following the hydrolysis of the natural substrate 2-arachidonoylglycerol resulting in arachidonic acid, which can be followed by mass spectrometry. This assay is hereinafter abbreviated “2-AG assay”.
  • The 2-AG assay was carried out in 384 well assay plates (PP. Greiner Cat #784201) in a total volume of 20 μL. Compound dilutions were made in 100% DMSO (VWR Chemicals 23500.297) in a polypropylene plate in 3-fold dilution steps to give a final concentration range in the assay from 12.5 μM to 0.8 μM. 0.25 μL compound dilutions (100% DMSO) were added to 9 μL MAGL in assay buffer (50 mM TRIS (GIBCO, 15567-027), 1 mM EDTA (Fluka, 03690-100 ml), 0.01% (v/v) Tween. After shaking, the plate was incubated for 15 min at RT. To start the reaction, 10 μL 2-arachidonoylglycerol in assay buffer was added. The final concentrations in the assay was 50 pM MAGL and 8 μM 2-arachidonoylglycerol. After shaking and 30 min incubation at RT, the reaction was quenched by the addition of 40 μL of acetonitrile containing 4 μM of d8-arachidonic acid. The amount of arachidonic acid was traced by an online SPE system (Agilent Rapidfire) coupled to a triple quadrupole mass spectrometer (Agilent 6460). A C18 SPE cartridge (G9205A) was used in an acetonitrile/water liquid setup. The mass spectrometer was operated in negative electrospray mode following the mass transitions 303.1→259.1 for arachidonic acid and 311.1→267.0 for d8-arachidonic acid. The activity of the compounds was calculated based on the ratio of intensities [arachidonic acid/d8-arachidonic acid].
  • TABLE 1
    IC50 MAGL IC50 MAGL
    Ex. [μM][a] Ex. [μM][a]
    1 0.040 8 0.013
    2 0.046 9 0.035
    3 0.066 10 0.039
    4 0.092 11 0.042
    5 1.3 12 0.077
    6 5.7 13 0.087
    7 0.011 14 0.191
    15 0.190 35 0.019
    16 0.208 36 0.119
    17 0.268 37 1.3
    18 0.892 38 0.160
    19 1.5 39 0.116
    20 1.8 40 0.012
    21 0.909 42 1.2
    22 2.1 43 0.529
    23 0.856 44 0.038
    24 0.004 45 0.850
    25 0.452 46 0.005
    26 0.684 47 0.005
    27 0.007 48 0.288
    28 0.018 49 0.363
    29 0.005 50 0.008
    30 0.273 51 0.010
    31 0.0 52 0.058
    32 0.020 53 0.006
    33 0.004 54 0.001
    34 0.438 55 0.079
    56 0.042 89 0.034[b]
    57 0.049 90 0.168[b]
    58 0.003 91 0.099[b]
    59 0.012 92 0.014[b]
    60 0.680 93 0.124
    61 0.011 94 0.150
    62 0.009 95 0.008
    64 0.040 96 0.293
    66 0.351 97 0.146
    67 0.120 98 0.010
    72 0.108 99 0.012
    73 0.008 100 0.082
    74 0.003 101 0.011
    75 0.002 102 0.022
    76 0.008 103 0.005
    77 0.016 104 0.451
    79 0.001 105 0.285
    81 0.006 108 0.001
    87 0.006[b] 109 0.223
    88 0.005[b] 110 2.179
    111 0.020 203 0.871[b]
    112 0.012 204 0.100[b]
    113 0.005 205 0.086[b]
    114 0.005 206 0.023[b]
    115 0.003 207 0.050[b]
    116 0.004 208 0.003[b]
    117 0.010 209 0.074[b]
    118 0.132[b] 210 0.080[b]
    119 0.014[b] 211 0.017[b]
    120 0.007[b] 212 0.003[b]
    121 0.151[b] 213 0.098[b]
    122 0.008[b] 214 0.036[b]
    123 0.082[b] 215 0.005[b]
    124 1.035[b] 216 0.006[b]
    139 0.002[b] 217 0.010[b]
    140 0.059[b] 218 0.003[b]
    141 0.040[b] 219 0.002[b]
    142 0.075[b] 220 0.003[b]
    201 0.239[b] 221 0.0007[b]
    202 0.054[b] 222 0.143[b]
    223 0.075[b] 243 0.010[b]
    224 0.003[b] 244 0.030[b]
    225 0.007[b] 245 1.7[b]
    226 0.045[b] 246 1.0[b]
    227 0.067[b] 247 1.1[b]
    228 0.126[b] 248 0.607[b]
    229 0.0009[b] 249 0.005[b]
    230 0.073[b] 250 0.008[b]
    231 0.005[b] 251 0.005[b]
    232 0.061[b] 252 0.005[b]
    233 0.022[b] 253 0.010[b]
    234 0.002[b] 254 0.064[b]
    235 0.033[b] 255 0.010[b]
    236 0.175[b] 256 0.011[b]
    237 0.254[b] 257 0.014[b]
    238 0.002[b] 258 0.048[b]
    239 0.0004[b] 259 0.015[b]
    240 0.016[b] 260 0.018[b]
    241 0.015[b] 261 0.048[b]
    242 0.017[b] 262 0.181[b]
    263 1.1[b] 279 0.001[b]
    264 0.084[b] 280 0.041[b]
    265 0.006[b] 281 0.035[b]
    266 0.018[b] 282 0.136[b]
    267 0.042[b] 283 0.179[b]
    268 0.028[b] 284 0.072[b]
    269 0.024[b] 285 2.6[b]
    270 0.061[b] 286 0.128[b]
    271 0.020[b] 287 0.006[b]
    272 0.094[b] 288 0.053[b]
    273 0.089[b] 289 0.003[b]
    274 0.126[b] 290 0.044[b]
    275 0.032[b] 291 0.078[b]
    276 0.019[b] 292 0.005[b]
    277 0.455[b] 293 0.030[b]
    278 0.211[b] 294 3.0[b]
    [a]if not indicated otherwise (see [b]), the activity was measured in 4-NPA assay;
    [b]measured in 2-AG assay.
  • In one aspect, the present invention provides compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein, wherein said compounds of formula (I) and their pharmaceutically acceptable salts or esters have IC50's for MAGL inhibition below 25 μM, preferably below 10 μM, more preferably below 5 μM as measured in the MAGL assay described herein.
  • In one embodiment, compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein have IC50 (MAGL inhibition) values between 0.0000001 μM and 25 μM, particular compounds have IC50 values between 0.000005 μM and 10 μM, further particular compounds have IC50 values between 0.00005 μM and 5 μM, as measured in the MAGL assay described herein.
  • In one embodiment, the present invention provides compounds of formula (I) and their pharmaceutically acceptable salts or esters as described herein, wherein said compounds of formula (I) and their pharmaceutically acceptable salts or esters have an IC50 for MAGL below 25 μM, preferably below 10 μM, more preferably below 5 μM as measured in an assay comprising the steps of:
      • a) providing a solution of a compound formula (I), or a pharmaceutically acceptable salt or ester thereof, in DMSO:
      • b) providing a solution of MAGL (recombinant wild-type) in assay buffer (50 mM tris(hydroxymethyl)aminomethane; 1 mM ethylenediaminetetraacetic acid);
      • c) adding 1 μL of compound solution from step a) to 19 μL of MAGL solution from step b);
      • d) shaking the mixture for 1 min at 2000 rpm;
      • e) incubating for 15 min at RT;
      • f) adding 20 μL of a solution of 4-nitrophenlyacetate in assay buffer (50 mM tris(hydroxymethyl)aminomethane; 1 mM ethylenediaminetetraacetic acid, 6% EtOH);
      • g) shaking the mixture for 1 min at 2000 rpm;
      • h) incubating for 5 min at RT;
      • i) measuring the absorbance of the mixture at 405 nm a fist time;
      • j) incubating a further 80 min at RT;
      • k) measuring the absorbance of the mixture at 405 nm a second time;
      • l) subtracting the absorbance measured under i) from the absorbance measured under k) and calculating the slope of absorbance;
      • wherein:
      • i) the concentration of the compound of formula (I), or the pharmaceutically acceptable salt or ester thereof in the assay after step f) is in the range of 25 μM to 1.7 nM;
      • ii) the concentration of MAGL in the assay after step f) is 1 nM;
      • iii) the concentration of 4-nitrophenylacetate in the assay after step f) is 300 μM; and
      • iv) steps a) to l) are repeated for at least 3 times, each time with a different concentration of the compound of formula (I), or the pharmaceutically acceptable salt or ester thereof.
  • Using the Compounds of the Invention
  • In one aspect, the present invention provides compounds of formula (I) as described herein for use as therapeutically active substance.
  • In a further aspect, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of cancer in a mammal.
  • In one aspect, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis. Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a preferred embodiment, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • In a particularly preferred embodiment, the present invention provides the use of compounds of formula (I) as described herein for the treatment or prophylaxis of multiple sclerosis in a mammal.
  • In one aspect, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In one embodiment, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • In one embodiment, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of cancer in a mammal.
  • In one embodiment, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • In one aspect, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a preferred embodiment, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • In a particularly preferred embodiment, the present invention provides compounds of formula (I) as described herein for use in the treatment or prophylaxis of multiple sclerosis in a mammal.
  • In one aspect, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of neurodegenerative diseases in a mammal.
  • In one embodiment, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of cancer in a mammal.
  • In a further aspect, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain and/or spasticity associated with pain in a mammal.
  • In a preferred embodiment, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal.
  • In a particularly preferred embodiment, the present invention provides the use of compounds of formula (I) as described herein for the preparation of a medicament for the treatment or prophylaxis of multiple sclerosis in a mammal.
  • In one aspect, the present invention provides a method for the treatment or prophylaxis of neuroinflammation, neurodegenerative diseases, pain, cancer and/or mental disorders in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • In one embodiment, the present invention provides a method for the treatment or prophylaxis of neuroinflammation and/or neurodegenerative diseases in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • In one embodiment, the present invention provides a method for the treatment or prophylaxis of neurodegenerative diseases in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • In one aspect, the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression and/or pain in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • In a preferred embodiment, the present invention provides a method for the treatment or prophylaxis of multiple sclerosis, Alzheimer's disease and/or Parkinson's disease in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • In a particularly preferred embodiment, the present invention provides a method for the treatment or prophylaxis of multiple sclerosis in a mammal, which method comprises administering an effective amount of a compound of formula (I) as described herein to the mammal.
  • Pharmaceutical Compositions and Administration
  • In one aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) as described herein and a therapeutically inert carrier.
  • The compounds of formula (I) and their pharmaceutically acceptable salts and esters can be used as medicaments (e.g. in the form of pharmaceutical preparations). The pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragées, hard and soft gelatin capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays) or rectally (e.g. in the form of suppositories). However, the administration can also be effected parentally, such as intramuscularly or intravenously (e.g. in the form of injection solutions).
  • The compounds of formula (I) and their pharmaceutically acceptable salts and esters can be processed with pharmaceutically inert, inorganic or organic adjuvants for the production of tablets, coated tablets, dragées and hard gelatin capsules. Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts etc. can be used, for example, as such adjuvants for tablets, dragées and hard gelatin capsules.
  • Suitable adjuvants for soft gelatin capsules are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols, etc.
  • Suitable adjuvants for the production of solutions and syrups are, for example, water, polyols, saccharose, invert sugar, glucose, etc.
  • Suitable adjuvants for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc.
  • Suitable adjuvants for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols, etc.
  • Moreover, the pharmaceutical preparations can contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances.
  • The dosage can vary in wide limits and will, of course, be fitted to the individual requirements in each particular case. In general, in the case of oral administration a daily dosage of about 0.1 mg to 20 mg per kg body weight, preferably about 0.5 mg to 4 mg per kg body weight (e.g. about 300 mg per person), divided into preferably 1-3 individual doses, which can consist, for example, of the same amounts, should be appropriate. It will, however, be clear that the upper limit given herein can be exceeded when this is shown to be indicated.
  • In accordance with the invention, the compounds of formula (I) or their pharmaceutically acceptable salts and esters can be used for the treatment or prophylaxis of type 2 diabetes related microvascular complications (such as, but not limited to diabetic retinopathy, diabetic neuropathy and diabetic nephropathy), coronary artery disease, obesity and underlying inflammatory diseases, chronic inflammatory and autoimmune/inflammatory diseases.
  • EXAMPLES
  • The invention will be more fully understood by reference to the following examples. The claims should not, however, be construed as limited to the scope of the examples.
  • In case the preparative examples are obtained as a mixture of enantiomers, the pure enantiomers can be separated by methods described herein or by methods known to the man skilled in the art, such as e.g., chiral chromatography (e.g., chiral SFC) or crystallization.
  • All reaction examples and intermediates were prepared under an argon atmosphere if not specified otherwise.
  • Method A1 Example 11 rac-(4aR,aS)-6-[4-[[4-(Trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00030
  • To a solution of 4-nitrophenyl 4-(4-(trifluoromethyl)benzyl)piperidine-1-carboxylate (100 mg, 245 μmol, BB2) in DMF (1.5 mL), rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one dihydrochloride (45.9 mg, 294 μmol, ChemBridge Corporation, BB1) and TEA (49.6 mg, 68.3 μL, 490 μmol) were added. The resultant reaction mixture was heated at 80° C. for 18 h. The reaction mixture was diluted with EtOAc and washed three times with H2O and NaHCO3. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (silica gel, eluting with a gradient of MeOH/EtOAc 0-10%) to afford the title compound as an off-white oil (0.045 g; 43.2%). MS (ESI): m/z=426.4 [M+H]+.
  • Method A2 Example 3 rac-(4aR,8aS)-6 [4-[(4-tert-Butylthiazol-2-yl)methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00031
  • To an ice-cold suspension of bis(trichloromethyl) carbonate (45.3 mg, 153 μmol, CAS RN 32315-10-9) and NaHCO3(73.3 mg, 873 μmol) in DCM (2 mL) was added in one portion 4-tert-butyl-2-(4-piperidylmethyl)thiazole hydrochloride (60 mg, 218 μmol, Enamine Ltd) and the mixture was stirred at RT overnight. The suspension was filtered and the filtrate was evaporated. The residue was diluted in DCM (1 mL) and added dropwise to an ice-cold solution of rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one dihydrochloride (50 mg, 218 μmol, ChemBridge Corporation, BB1) and DIPEA (152 μL, 870 μmol) in DCM (1 mL). The suspension was stirred at RT for 19 h to become a solution. The reaction mixture was poured on H2O and DCM and the layers were separated. The aqueous layer was extracted three times with DCM. The organic layers were washed twice with water, dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 4 g column using an MPLC system eluting with a gradient of DCM:MeOH (100:0 to 90:10) to provide the desired compound as a colorless foam (0.039 g; 42.5%). MS (ESI); m/z=421.2 [M+H]+.
  • Method A3 Example 34 (+)- or (−)-4-[[1-[(4aR,8aS)-3-Oxo-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazine-6-carbonyl]-4-piperidyl]methyl]benzonitrile
  • Figure US20200308190A1-20201001-C00032
  • To an ice-cold solution of bis(trichloromethyl) carbonate (39.9 mg, 134 μmol, CAS RN 32315-10-9) in DCM were added NaHCO3(64.5 mg, 768 μmol) and (+)-cis-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (30 mg, 192 μmol, BB a) and the mixture was stirred at RT overnight. To the suspension was added 4-(piperidin-4-ylmethyl)benzonitrile (38.5 mg, 192 μmol, CAS RN 333987-57-8) and DIPEA (99.3 mg, 134 μL, 768 μmol). The suspension was stirred at RT for 4.5 h. The reaction mixture was poured on H2O and DCM and the layers were separated. The aqueous layer was extracted three times with DCM. The organic layers were washed twice with H2O, dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 4 g column using an MPLC system eluting with a gradient of DCM:MeOH (100:0 to 90:10) to furnish the desired compound as a colorless gum (0.023 g; 31.3%). MS (ESI): m/z=383.2 [M+H]+.
  • Method A4 Example 79 (4aR,8aS)-6-(4-((2-chloro-4-fluorophenoxy)methyl)-4-methylpiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00033
  • To a solution of 4-nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate (25 mg, 77.8 μmol, BB7a) in NMP (1 mL) was added DIPEA (25.1 mg, 34 μL, 195 μmol) and 4-((2-chloro-4-fluorophenoxy)methyl)-4-methylpiperidine; hydrochloride salt (19.5 mg, 66.1 μmol, BB12). The reaction vial was stirred at 140° C. for 45 min. The crude material was purified by reversed-phase HPLC to yield 23.2 mg of the desired product. MS (ESI): m/z=440.2 [M+H]+.
  • Method A5 Example 64 (4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00034
  • A microwave vial was heat gun-dried and charged with bis(trichloromethyl) carbonate (26.6 mg, 89.6 μmol) and sodium bicarbonate (32.3 mg, 384 mol). The flask was placed under argon and DCM (1 mL) was added to give a suspension. The suspension was cooled by an ice-bath and 4-((2-chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine; hydrochloride salt (36.1 mg, 121 mol, BB15) was added. The mixture was stirred at 0° C. for 15 min and at RT overnight. The reaction mixture was cooled down in an-ice bath and DCM (500 μL) and DIPEA (49.7 mg, 67.1 μL, 384 μmol) followed by (4aR,8aS)-6-(4-((2-chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (21.1 mg, 47.5 μmol, BB1a) were added. The resulting off-white suspension was stirred at room temperature for 7 h. The reaction mixture was poured on water, DCM was added and the layers were separated. The aqueous layer was extracted twice with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and evaporated to afford a yellow oil (58 mg). The crude product was purified by reverse-phase HPLC and lyophilized to provide the title compound as a white solid (21.1 mg, 37.1% yield). MS (ESI): m/z=444.2 [M+H]+.
  • Method A6 Example 39 (4aR,8aS)-6-[4-[(2-Fluoro-4-methoxyphenoxy)methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00035
  • To a solution of 2-fluoro-4-methoxyphenol (16.5 mg, 13 μL, 116 μmol), (4aR,8aS)-6-[4-(hydroxymethyl)piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (34.5 mg, 116 μmol, BB16) and triphenylphosphine (33.5 mg, 128 μmol) in DCM (580 μL) was added DIAD (25.8 mg, 24.8 μL, 128 μmol) dropwise and the reaction was stirred at room temperature for 22 h. The reaction mixture was diluted with DCM and washed with 1M aq. NaOH. The phases were separated and the aq. phase was extracted with DCM twice. The combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness to give a red oil (99 mg). The crude product was purified by reverse-phase HPLC and lyophilized to afford the desired compound (20 mg, 40.9% yield) as a white solid. MS (ESI): m/z=422.3[M+H]+.
  • Method A7 Example 42 and 43 (4aS,8aR)-6-(4-(((6-(Trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 42) and (4aR,8aS)-6-(4-(((6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 43)
  • Figure US20200308190A1-20201001-C00036
  • Step a) rac-(4aR,8aS)-6-(4-(Chloromethyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00037
  • To a solution of rac-(4aR,8aS)-6-(4-(hydroxymethyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (80 mg, 269 μmol, BB16) in dry DMF (2 mL) was added to DIPEA (52.2 mg, 70.5 μL, 404 μmol). DMAP (1.64 mg, 13.5 μmol) and methanesulfonyl chloride (46.2 mg, 404 μmol) and the reaction mixture was stirred at room temperature for 2 h. Addition of 4,4-difluoropiperidine; hydrochloride salt (84.8 mg, 538 μmol). DIPEA (139 mg, 188 μL, 1.08 mmol) and the reaction mixture was stirred at room temperature for 2 h. The reaction was then stirred at 70° C. for 14 h. The crude reaction was submitted for reversed-phase HPLC purification to yield the title compound as a side product (35 mg). MS (ESI): m/z=315.1 [M+H]+.
  • Step b) (4aS,8aR)-6-(4-(((6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 42) and (4aR 8aS)-6-(4-(((6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 43)
  • Figure US20200308190A1-20201001-C00038
  • To a solution of rac-(4aR,8aS)-6-(4-(chloromethyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (70 mg, 222 μmol) in dry DMF (1 mL) was added 6-(trifluoromethyl)pyridin-3-ol (54.2 mg, 332 μmol) and Cs2CO3 (108 mg, 332 μmol). The reaction mixture was stirred at 95° C. for 18 h. Insolubles were removed by filtration over Celite, the filtrate was concentrated down to dryness and the crude residue was purified and the enantiomers separated by chiral SFC to yield Example 42 (33.8 mg) and Example 43 (32.5 mg). MS (ESI): m/z=443.2 [M+H]+ for both examples.
  • Method A8 Example 26 (4aS,8aR)-6-(4-((4-(Trifluoromethyl)-1H-pyrazol-1-yl)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00039
  • To a solution of rac-(4aR,8aS)-6-(4-(hydroxymethyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (75 mg, 252 μmol, BB16) in dry DMF (2 mL) was added DIPEA (39.1 mg, 52.9 μL, 303 μmol), DMAP (3.08 mg, 25.2 μmol) and methanesulfonyl chloride (30.3 mg, 265 μmol) and the reaction mixture was stirred at room temperature for 2 h. 4-(Trifluoromethyl)-1H-pyrazole (68.6 mg, 504 μmol) and K2CO3 (87.1 mg, 631 μmol) were added and the reaction mixture was stirred at 90° C. for 18 h. Insolubles were removed by filtration over celite, the filtrate was concentrated to dryness in vacuo and the crude residue was directly purified by flash chromatography with an eluent mixture of DCM and MeOH (0% to 10%), to yield 90 mg of the desired product as a racemate. This was submitted for SFC chiral separation to yield Example 26 (25 mg) as a colorless oil and the enantiomer (31 mg) as a colorless oil. MS (ESI): m/z=416.2 [M+H]+.
  • Method A9 Example 37 (4aR,8aS)-6-(4-((4,4-Difluoropiperidin-1-yl)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00040
  • To a solution of (4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (40 mg, 256 μmol, BB1a) in dry DMF (2 mL) cooled down to 0° C. was added DIPEA (39.7 mg, 53.7 μL, 307 μmol) and 4-nitrophenyl carbonochloridate (61.9 mg, 307 μmol). The reaction mixture was stirred at 0° C. for 20 min. LCMS control showed formation of the intermediate carbamate. DIPEA (116 mg, 157 μL, 896 μmol) and 4,4-difluoro-1-(piperidin-4-ylmethyl)piperidine; dihydrochloride salt (89.5 mg, 307 μmol, BB17) were added and the reaction mixture was then stirred at room temperature for 30 min, then stirred at 100° C. for 14 h. Volatiles were removed in vacuo and the crude residue was directly submitted for SFC purification to yield the desired compound (9.5 mg) as a light orange oil. MS (ESI): m/z=401.3 [M+H]+.
  • Method A10 Example 125 (+)-(4aR,8aS)-6-[4-[2-(2-Chlorophenyl)ethynyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00041
  • In a sealed tube, 4-[2-(2-chlorophenyl)ethynyl]piperidine (BB 18, 0.02 g, 0.078 mmol) and 4-nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate (BB7a, 0.025 g, 0.078 mmol) were mixed in ACN (0.6 mL). Then, Huenig's base (0.041 mL, 0.234 mmol) was added, followed by DMAP (0.005 g, 0.039 mmol) and the reaction mixture was heated to 90° C. overnight. The mixture was evaporated to dryness and the crude residue purified by reverse phase HPLC to give the title compound (0.013 g, 41%) as a colorless solid. MS (ESI): m/z=402.2 [M+H]+.
  • Method B1 Example 1 (+)-(4aR,8aS)-6-(4-((4-(tert-Butyl)thiazol-2-yl)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00042
  • The enantiomers of example 3 were separated by preparative chiral HPLC (Chiralcel OD column) using an isocratic mixture of EtOH (containing 0.05% of NH4OAc):n-heptane (20:80). The fractions were evaporated to provide the desired compound as a colorless solid (0.012 g; 34.3%). MS (ESI): m/z=421.2 [M+H]+.
  • Method B2 Example 12 (+)- or (−)-(4aR,8aS)-6-(4-(4-(Trifluoromethyl)phenoxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00043
  • The enantiomers of example 13 were separated using preparative chiral HPLC (Chiralpak AD column) using an isocratic mixture of EtOH (containing 0.05% of NH4OAc):n-heptane (40:60). The fractions were evaporated to yield the desired compound as a light brown oil (0.013 g: 28.4%). MS (ESI): m/z=428.2 [M+H]+.
  • Method B3 Examples 103, 104 and 105 (4aR,aS)-6-[2-Methyl-3-[[4-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (Isomer A+B, Isomer C, Isomer D)
  • Figure US20200308190A1-20201001-C00044
  • The stereoisomers of example 117 were separated by preparative chiral HPLC (Reprosil Chiral NR column) using an isocratic mixture of EtOH (containing 0.05% of NH4OAc): n-heptane (40:60) to provide examples 103 and 104 as single isomers and example 105 as mixture of two stereoisomers. The fractions were evaporated to provide the desired compounds as colorless solids.
  • Method C Example 21 rac-(4aR,8aS)-6(4-(4-(Trifluoromethyl)benzyl)piperazine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00045
  • A mixture of rac-cis-6-(piperazine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (35 mg, 130 μmol, BB3), 4-(trifluoromethyl)benzaldehyde (22.7 mg, 17.4 μL, 130 μmol) and sodium triacetoxyborohydride (27.6 mg, 130 μmol) in DCM (1 mL) was stirred at RT for 15 h. The reaction mixture was concentrated and the residue was purified by preparative HPLC to give the desired compound as a white solid (8 mg, 14.4%). MS (ESI): m/z=427.4 [M+H]+.
  • If not indicated otherwise the following examples were synthesized from rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one dihydrochloride (ChemBridge Corporation) and the suitable building blocks in analogy to the reaction methods described herein.
  • Building
    Ex. Systematic Name/Structure block(s) MS, m/z Method
    2
    Figure US20200308190A1-20201001-C00046
    Example  4 405.3 [M + H]+ B1
    4
    Figure US20200308190A1-20201001-C00047
    Supplier of building block: BCH Research (UK) and BB1 405.4 [M + H]+ A2
    5
    Figure US20200308190A1-20201001-C00048
    Example  3 421.2 [M + H]+ B1
    6
    Figure US20200308190A1-20201001-C00049
    Example   4 405.3 [M + H]+ B1
    7
    Figure US20200308190A1-20201001-C00050
    Supplier of building block: UkrOrg- Syntez Ltd. and BB1 426.2 [M + H]+ A2
    8
    Figure US20200308190A1-20201001-C00051
    Example  11 426.4 [M + H]+ B1
    9
    Figure US20200308190A1-20201001-C00052
    4-[(4- Chloro- phenoxy) methyl] piperi- dine (CAS RN 63608- 33-3) and BB1 408.3 [M + H]+ A2
    10
    Figure US20200308190A1-20201001-C00053
    4-(4- Chloro- benzyl)- piperi- dine hydro- chloride (CAS RN 36938- 76-8) and BB1 392.2 [M + H]+ A2
    13
    Figure US20200308190A1-20201001-C00054
    4-(4- Trifluoro- methyl- phenoxy) piperi- dine hydro- chloride (CAS RN 28033- 37-6) and BB1 428.2 [M + H]+ A2
    14
    Figure US20200308190A1-20201001-C00055
    Example  17 426.3 [M + H]+ B1
    15
    Figure US20200308190A1-20201001-C00056
    BB4 and BB1 374.4 [M + H]+ A1
    16
    Figure US20200308190A1-20201001-C00057
    FCH Group and BB1 405.2 [M + H]+ A2
    17
    Figure US20200308190A1-20201001-C00058
    4-(4- Trifluoro- methyl benzyl) piperi- dine HCl (CAS RN 192990- 03-7) and BB6 426.3 [M + H]+ A2
    18
    Figure US20200308190A1-20201001-C00059
    3-Phenyl- 5-(piperi- din-4- ylmethyl)- 1,2,4- oxa- diazole (CAS RN 1239730- 22-3) and BB1 426.3 [M + H]+ A2
    19
    Figure US20200308190A1-20201001-C00060
    Example  17 426.4 [M + H]+ B1
    20
    Figure US20200308190A1-20201001-C00061
    Example  11 426.4 [M + H]+ B1
    22
    Figure US20200308190A1-20201001-C00062
    1-(4- Chlor- benzyl)- pipera- zine (CAS RN 23145- 88-2) and BB1 393.2 [M + H]+ A2
    23
    Figure US20200308190A1-20201001-C00063
    Example   7 426.2 [M + H]+ B1
    24
    Figure US20200308190A1-20201001-C00064
    Example   7 426.2 [M + H]+ B1
    25
    Figure US20200308190A1-20201001-C00065
    BB5 and BB1 427.2 [M + H]+ A2
    27
    Figure US20200308190A1-20201001-C00066
    Supplier of building block: HDH Pharma, Inc. and BB1 432.2 [M + H]+ A2
    28
    Figure US20200308190A1-20201001-C00067
    Supplier of building block: ZereneX Mole- cular Limi- ted and BB1a 398.1 [M + H]+ A2
    29
    Figure US20200308190A1-20201001-C00068
    Example  27 432.2 [M + H]+ B1
    30
    Figure US20200308190A1-20201001-C00069
    Example  27 432.2 [M + H]+ B1
    31
    Figure US20200308190A1-20201001-C00070
    4-[4- (Trifluoro- methoxy) benzyl] piperi- dine (CAS RN 681482- 50-8) and BB1a 442.2 [M + H]+ A2
    32
    Figure US20200308190A1-20201001-C00071
    4-((2,4- Difluoro- phenoxy) methyl) piperi- dine HCl CAS RN 614731- 39-4 and BB1a 410.2 [M + H]+ A5
    33
    Figure US20200308190A1-20201001-C00072
    Building block prepared as des- cribed in WO2013/ 179024 and BB1a 410.2 [M + H]+ A2
    35
    Figure US20200308190A1-20201001-C00073
    4-(4- Chloro- benzyl)- piperi- dine hydro- chloride (CAS RN 36938- 76-8) and BB1a 392.2 [M + H]+ A2
    36
    Figure US20200308190A1-20201001-C00074
    3-[[4- (Trifluoro- methyl) phenyl] methyl] azetidine (CAS RN 937614- 88-5) and BB1a 398.3 [M + H]+ A1
    38
    Figure US20200308190A1-20201001-C00075
    BB8 and BB1a 405.3 [M + H]+ A2
    40
    Figure US20200308190A1-20201001-C00076
    BB9 and BB1a 462.1 [M + H]+ A2
    41
    Figure US20200308190A1-20201001-C00077
    BB19 and BB1a 495.18 [M + H]+ A3
    44
    Figure US20200308190A1-20201001-C00078
    4-(3- (Trifluoro- methyl) phenoxy) piperi- dine (CAS RN 337912- 66-0) and BB1a 428.2 [M + H]+ A3
    45
    Figure US20200308190A1-20201001-C00079
    2-Chloro- 4-(tri- fluoro- methoxy) phenol (CAS: 35852- 58-5) and BB1b 492.2 [M + H]+ A6
    46
    Figure US20200308190A1-20201001-C00080
    2-Chloro- 4-(tri- fluoro- methoxy) phenol (CAS RN: 35852- 58-5) and BB1a 492.2 [M + H]+ A6
    47
    Figure US20200308190A1-20201001-C00081
    BB20 and BB1a 432.2 [M + H]+ A5
    48
    Figure US20200308190A1-20201001-C00082
    BB12 and BB7b 440.2 [M + H]+ A4 (1:1 ACN: iPrOH)
    49
    Figure US20200308190A1-20201001-C00083
    4-[(2,4- Difluoro- phenoxy) methyl] piperi- dine hydro- chloride (CAS RN: 614731- 39-4) and BB7b 410.2 [M + H]+ A4
    50
    Figure US20200308190A1-20201001-C00084
    4-Chloro- 2-fluoro- phenyl 4- piperi- dinyl- methyl ether; hydro- chloride salt (CAS: 946680- 87-1) and BB7A 426.2 [M + H]+ A4 (micro- wave heat- ing)
    51
    Figure US20200308190A1-20201001-C00085
    BB21 and BB7a 460.2 [M + H]+ A4
    52
    Figure US20200308190A1-20201001-C00086
    BB22 and BB7a 460.2 [M + H]+ A4
    53
    Figure US20200308190A1-20201001-C00087
    BB91 and BB1a 495.3 [M + H]+ A3
    54
    Figure US20200308190A1-20201001-C00088
    BB23 and BB1a 476.3 [M + H]+ A5
    55
    Figure US20200308190A1-20201001-C00089
    BB24 and BB1a 417.2 [M + H]+ A5
    56
    Figure US20200308190A1-20201001-C00090
    1-[[2- (Pyrro- lidin- 1-yl)-4- (trifluoro- methyl) phenyl] methyl] pipera- zine (synthe- sized accord- ing to WO2015/ 179559) and BB1a 496.26 [M + H]+ A3
    57
    Figure US20200308190A1-20201001-C00091
    BB25 and BB1a 398.2 [M + H]+ A5
    58
    Figure US20200308190A1-20201001-C00092
    BB10 and BB1a 494.3 [M + H]+ A3
    59
    Figure US20200308190A1-20201001-C00093
    BB26 and BB1a 433.2 [M + H]+ A5
    60
    Figure US20200308190A1-20201001-C00094
    BB27 and BB1a 416.3 [M + H]+ A9
    61
    Figure US20200308190A1-20201001-C00095
    BB28 and BB1a 406.3 [M + H]+ A5
    62
    Figure US20200308190A1-20201001-C00096
    BB29 and BB1a 448.2 [M + H]+ A5
    63
    Figure US20200308190A1-20201001-C00097
    BB30 and BB1a 445.24 [M + H]+ A3
    65
    Figure US20200308190A1-20201001-C00098
    BB31 and BB1a 401.22 [M + H]+ A3
    66
    Figure US20200308190A1-20201001-C00099
    BB32 and BB1a 404.3 [M + H]+ A9 (puri- fied by RP- HPLC)
    67
    Figure US20200308190A1-20201001-C00100
    4-(4- Trifluoro- methyl benzyl) piperi- dine HCl (CAS RN 192990- 03-7) and BB33 440.3 [M + H]+ A3
    68
    Figure US20200308190A1-20201001-C00101
    BB34 and BB1a 495.18 [M + H]+ A3
    69
    Figure US20200308190A1-20201001-C00102
    BB35 and BB1a 495.11 [M + H]+ A3 fol- lowed by RP- HPLC
    70
    Figure US20200308190A1-20201001-C00103
    BB35 and BB1a 461.16 [M + H]+ A3 fol- lowed by RP- HPLC
    71
    Figure US20200308190A1-20201001-C00104
    BB35 and BB1a 427.2 [M + H]+ A3 fol- lowed by RP- HPLC
    72
    Figure US20200308190A1-20201001-C00105
    BB11 and BB7a 399.2 [M + H]+ A1
    73
    Figure US20200308190A1-20201001-C00106
    BB13 and BB7a 504.1 [M + H]+ A1
    74
    Figure US20200308190A1-20201001-C00107
    BB36 and BB1a 440.1 [M + H]+ A3
    75
    Figure US20200308190A1-20201001-C00108
    BB7a and BB37 448.2 [M + H]+ A4 (sol- vent ACN instead of NMP)
    76
    Figure US20200308190A1-20201001-C00109
    BB7a and BB38 414.2 [M + H]+ A4 (sol- vent ACN instead of NMP)
    77
    Figure US20200308190A1-20201001-C00110
    BB7a and BB39 448.2 [M + H]+ A4 (sol- vent ACN instead of NMP)
    78
    Figure US20200308190A1-20201001-C00111
    BB7a and BB40 411.2 [M + H]+ A4 (sol- vent ACN not NMP)
    80
    Figure US20200308190A1-20201001-C00112
    BB41 and BB1a 509.2 [M + H]+ A3
    81
    Figure US20200308190A1-20201001-C00113
    BB14 and BB7a 492.2 [M + H]+ A1
    82
    Figure US20200308190A1-20201001-C00114
    BB42 and BB1a 509.2 [M + H]+ A3
    83
    Figure US20200308190A1-20201001-C00115
    BB43 and BB1a 517.18 [M + H]+ A3
    84
    Figure US20200308190A1-20201001-C00116
    BB44 and BB1a 517.18 [M + H]+ A3
    85
    Figure US20200308190A1-20201001-C00117
    N- methyl- N- (piperi- din-4-yl)- 4-(tri- fluoro- methyl) benza- mide hydro- chloride (CAS RN 1580795- 67-0) and BB1a 469.20 [M + H]+ A3
    86
    Figure US20200308190A1-20201001-C00118
    BB94 and BB1a 483.22 [M + H]+ A3
    87
    Figure US20200308190A1-20201001-C00119
    BB88 and BB1a 414.3 [M + H]+ A5
    88
    Figure US20200308190A1-20201001-C00120
    BB45 and BB1a 444.3 [M + H]+ A4 (ACN as sol- vent)
    89
    Figure US20200308190A1-20201001-C00121
    BB46 and BB1a 457.2 [M + H]+ A3
    90
    Figure US20200308190A1-20201001-C00122
    3-[(3- Chloro- phenoxy) methyl] pyrroli- dine (CAS RN 914299- 54-0) and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    91
    Figure US20200308190A1-20201001-C00123
    BB47 and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    92
    Figure US20200308190A1-20201001-C00124
    BB48 and BB1a 444.2 [M + H]+ A4 (ACN as sol- vent)
    93
    Figure US20200308190A1-20201001-C00125
    BB49 and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    94
    Figure US20200308190A1-20201001-C00126
    BB50 and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    95
    Figure US20200308190A1-20201001-C00127
    BB51 and BB1a 466.23 [M + H]+ A4 (ACN as sol- vent)
    96
    Figure US20200308190A1-20201001-C00128
    BB52 and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    97
    Figure US20200308190A1-20201001-C00129
    BB53 and BB1a 394.15 [M + H]+ A4 (ACN as sol- vent)
    98
    Figure US20200308190A1-20201001-C00130
    BB54 and BB1a 440.4 [M + H]+ A3
    99
    Figure US20200308190A1-20201001-C00131
    BB55 and BB1a 460.16 [M + H]+ A3
    100
    Figure US20200308190A1-20201001-C00132
    3-(4- Trifluoro- methyl- benzyl) pyrroli- dine hydro- chloride salt (CAS RN: 957988- 84-4) and BB1a 412.19 [M + H]+ A3
    101
    Figure US20200308190A1-20201001-C00133
    BB56 and BB1a 432.2 [M + H]+ A4 (ACN as sol- vent)
    102
    Figure US20200308190A1-20201001-C00134
    BB1a and BB57 446.3 [M + H]+ A4 (ACN as sol- vent)
    103
    Figure US20200308190A1-20201001-C00135
    Example 117 428.19 [M + H]+ B3
    104
    Figure US20200308190A1-20201001-C00136
    Example 117 428.19 [M + H]+ B3
    105
    Figure US20200308190A1-20201001-C00137
    Example 117 428.19 [M + H]+ B3
    106
    Figure US20200308190A1-20201001-C00138
    BB58 and BB1a 445.19 [M + H]+ A3
    107
    Figure US20200308190A1-20201001-C00139
    BB59 and BB1a 523.22 [M + H]+ A3
    108
    Figure US20200308190A1-20201001-C00140
    BB60 and BB1a 448.2 [M + H]+ A4 (1:1 iPrOH: ACN as sol- vent)
    109
    Figure US20200308190A1-20201001-C00141
    BB61 and BB1a 416.2 [M + H]+ A4 (1:1 iPrOH: ACN as sol- vent)
    110
    Figure US20200308190A1-20201001-C00142
    BB62 and BB1a 500.2 [M + H]+ A4 (ACN as sol- vent)
    111
    Figure US20200308190A1-20201001-C00143
    BB63 and BB1a 446.2 [M + H]+ A4 (ACN as sol- vent)
    112
    Figure US20200308190A1-20201001-C00144
    BB64 and BB1a 450.2 [M + H]+ A4 (ACN as sol- vent)
    113
    Figure US20200308190A1-20201001-C00145
    BB65 and BB1a 432.2 [M + H]+ A4 (ACN as sol- vent)
    114
    Figure US20200308190A1-20201001-C00146
    BB66 and BB1a 432.2 [M + H]+ A4 (ACN as sol- vent)
    115
    Figure US20200308190A1-20201001-C00147
    BB67 and BB1a 444.3 [M + H]+ A4 (ACN as sol- vent)
    116
    Figure US20200308190A1-20201001-C00148
    BB68 and BB1a 448.2 [M + H]+ A4 (1:1 iPrOH: ACN as sol- vent)
    117
    Figure US20200308190A1-20201001-C00149
    BB69 and BB1a 428.18 [M + H]+ A3
    118
    Figure US20200308190A1-20201001-C00150
    BB70 and BB1a 400.2 [M + H]+ A4 (ACN as sol- vent)
    119
    Figure US20200308190A1-20201001-C00151
    BB71 and BB1a 462.2 [M + H]+ A4 (ACN as sol- vent)
    120
    Figure US20200308190A1-20201001-C00152
    BB72 and BB1a 434.1 [M + H]+ A4 (ACN as sol- vent)
    121
    Figure US20200308190A1-20201001-C00153
    BB73 and BB1a 479.2 [M + H]+ A4 (ACN as sol- vent)
    122
    Figure US20200308190A1-20201001-C00154
    BB74 and BB1a 442.1 [M + H]+ Hans A4 (ACN as sol- vent)
    123
    Figure US20200308190A1-20201001-C00155
    BB75 and BB1a 453.0 [M + H]+ Hans A4 (ACN as sol- vent)
    124
    Figure US20200308190A1-20201001-C00156
    BB76 and BB1a 400.2 [M + H]+ A4 (ACN as sol- vent)
    126
    Figure US20200308190A1-20201001-C00157
    BB77 403.3 [M + H]+ A10
    127
    Figure US20200308190A1-20201001-C00158
    BB78 403.2 [M + H]+ A10
    128
    Figure US20200308190A1-20201001-C00159
    BB79 420.3 [M + H]+ A10
    129
    Figure US20200308190A1-20201001-C00160
    BB80 402.3 [M + H]+ A10
    130
    Figure US20200308190A1-20201001-C00161
    BB81 402.3 [M + H]+ A10
    131
    Figure US20200308190A1-20201001-C00162
    BB82 436.3 [M + H]+ A10
    132
    Figure US20200308190A1-20201001-C00163
    BB83 418.4 [M + H]+ A10
    133
    Figure US20200308190A1-20201001-C00164
    BB84 374.2 [M + H]+ A10
    134
    Figure US20200308190A1-20201001-C00165
    BB85 408.3 [M + H]+ A10
    135
    Figure US20200308190A1-20201001-C00166
    BB86 392.2 [M + H]+ A10
    136
    Figure US20200308190A1-20201001-C00167
    BB92 413.2 [M + H]+ A1 (ACN as sol- vent DIPEA as base)
    137
    Figure US20200308190A1-20201001-C00168
    BB93 441.2 [M + H]+ A1 (ACN as sol- vent DIPEA as base)
    139
    Figure US20200308190A1-20201001-C00169
    BB95 430.4 [M + H]+ A1 (ACN as sol- vent DIPEA as base)
    140
    Figure US20200308190A1-20201001-C00170
    BB96 440.18 [M + H]+ A3 fol- lowed by chiral SFC
    141
    Figure US20200308190A1-20201001-C00171
    BB96 440.18 [M + H]+ A3 fol- lowed by chiral SFC
    142
    Figure US20200308190A1-20201001-C00172
    BB97 440.2 [M + H]+ A3
    143
    Figure US20200308190A1-20201001-C00173
    BB98 and BB1a 450.1 [M + H]+ A3
    144
    Figure US20200308190A1-20201001-C00174
    BB99 and BB1a 482.2 [M + H]+ A3
    145
    Figure US20200308190A1-20201001-C00175
    BB100 and BB1a 466.2 [M + H]+ A3
    146
    Figure US20200308190A1-20201001-C00176
    Example 145 466.2 [M + H]+ SFC, Chiral- pak AD, 40% MeOH
    147
    Figure US20200308190A1-20201001-C00177
    Example 145 466.2 [M + H]+ SFC, Chiral- pak AD, 40% MeOH
    148
    Figure US20200308190A1-20201001-C00178
    BB101 and BB1a 464.1 [M + H]+ A3
    152
    Figure US20200308190A1-20201001-C00179
    BB104 and BB1a 496.1 [M + H]+ A3
    153
    Figure US20200308190A1-20201001-C00180
    BB105 and BB1a 480.1 [M + H]+ A3
    154
    Figure US20200308190A1-20201001-C00181
    Example 153 480.1 [M + H]+ A3, then HPLC Repro- sil Chiral NR, 60% hep- tane, 40% EtOH + NH4Ac
    155
    Figure US20200308190A1-20201001-C00182
    Example 153 480.1 [M + H]+ A3, then HPLC Repro- sil Chiral NR, 60% hep- tane, 40% EtOH + NH4Ac
    156
    Figure US20200308190A1-20201001-C00183
    BB106 and BB1a 448.1 [M + H]+ A3
    157
    Figure US20200308190A1-20201001-C00184
    BB107 408.2 [M + H]+ A10
    158
    Figure US20200308190A1-20201001-C00185
    BB108 426.3 [M + H]+ A10
    159
    Figure US20200308190A1-20201001-C00186
    BB109 376.3 [M + H]+ A10
    160
    Figure US20200308190A1-20201001-C00187
    BB110 442.3 [M + H]+ A10
    161
    Figure US20200308190A1-20201001-C00188
    BB111 392.3 [M + H]+ A10
    162
    Figure US20200308190A1-20201001-C00189
    BB112 414.4 [M + H]+ A10
    163
    Figure US20200308190A1-20201001-C00190
    BB113 370.4 [M + H]+ A10
    164
    Figure US20200308190A1-20201001-C00191
    BB114 442.3 [M + H]+ A10
    165
    Figure US20200308190A1-20201001-C00192
    BB115 374.3 [M + H]+ A10
    166
    Figure US20200308190A1-20201001-C00193
    BB116 424.3 [M + H]+ A10
    167
    Figure US20200308190A1-20201001-C00194
    BB117 408.4 [M + H]+ A10
    168
    Figure US20200308190A1-20201001-C00195
    BB118 372.2 [M + H]+ A10
    169
    Figure US20200308190A1-20201001-C00196
    BB119 368.4 [M + H]+ A10
    170
    Figure US20200308190A1-20201001-C00197
    BB120 424.3 [M + H]+ A10
    171
    Figure US20200308190A1-20201001-C00198
    BB121 420.3 [M + H]+ A10
    172
    Figure US20200308190A1-20201001-C00199
    BB122 392.3 [M + H]+ A10
    173
    Figure US20200308190A1-20201001-C00200
    BB123 354.3 [M + H]+ A10
    174
    Figure US20200308190A1-20201001-C00201
    BB124 392.3 [M + H]+ A10
    175
    Figure US20200308190A1-20201001-C00202
    BB125 406.3 [M + H]+ A10
    176
    Figure US20200308190A1-20201001-C00203
    BB126 399.3 [M + H]+ A10
    177
    Figure US20200308190A1-20201001-C00204
    BB127 406.3 [M + H]+ A10
    178
    Figure US20200308190A1-20201001-C00205
    BB128 405.4 [M + H]+ A10
    179
    Figure US20200308190A1-20201001-C00206
    BB129 380.4 [M + H]+ A10
    180
    Figure US20200308190A1-20201001-C00207
    BB130 396.4 [M + H]+ A10
    181
    Figure US20200308190A1-20201001-C00208
    BB131 370.3 [M + H]+ A10
    182
    Figure US20200308190A1-20201001-C00209
    BB132 390.3 [M + H]+ A10
    183
    Figure US20200308190A1-20201001-C00210
    BB133 384.3 [M + H]+ A10
    184
    Figure US20200308190A1-20201001-C00211
    BB134 388.3 [M + H]+ A10
    185
    Figure US20200308190A1-20201001-C00212
    BB135 392.2 [M + H]+ A10
    186
    Figure US20200308190A1-20201001-C00213
    BB136 418.3 [M + H]+ A10
    187
    Figure US20200308190A1-20201001-C00214
    BB137 380.2 [M + H]+ A10
    188
    Figure US20200308190A1-20201001-C00215
    BB138 380.2 [M + H]+ A10
    189
    Figure US20200308190A1-20201001-C00216
    BB139 460.3 [M + H]+ A10
    190
    Figure US20200308190A1-20201001-C00217
    BB140 390.3 [M + H]+ A10
    191
    Figure US20200308190A1-20201001-C00218
    BB141 384.3 [M + H]+ A10
    192
    Figure US20200308190A1-20201001-C00219
    BB143 436.4 [M + H]+ A10
    193
    Figure US20200308190A1-20201001-C00220
    BB144 398.4 [M + H]+ A10
    194
    Figure US20200308190A1-20201001-C00221
    BB145 382.4 [M + H]+ A10
    195
    Figure US20200308190A1-20201001-C00222
    BB146 396.4 [M + H]+ A10
    196
    Figure US20200308190A1-20201001-C00223
    BB147 450.3 [M + H]+ A10
    197
    Figure US20200308190A1-20201001-C00224
    BB148 434.4 [M + H]+ A10
    198
    Figure US20200308190A1-20201001-C00225
    BB149 330.3 [M − H2O + H]+ A10
    199
    Figure US20200308190A1-20201001-C00226
    BB149 (Elimi- nation product iso- lated during synthe- sis of example 198) 330.3 [M + H]+ A10
    200
    Figure US20200308190A1-20201001-C00227
    BB142 442.3 [M + H]+ A10
    201
    Figure US20200308190A1-20201001-C00228
    BB7a and BB98 494.2 [M + H]+ A4
    202
    Figure US20200308190A1-20201001-C00229
    BB7a and BB99 524.2 [M + H]+ A4
    203
    Figure US20200308190A1-20201001-C00230
    BB7a and BB100 495.2 [M + H]+ A4
    204
    Figure US20200308190A1-20201001-C00231
    BB7a and BB101 434.1 [M + H]+ A4
    205
    Figure US20200308190A1-20201001-C00232
    BB7a and BB102 460.2 [M + H]+ A4
    206
    Figure US20200308190A1-20201001-C00233
    BB7a and BB103 513.3 [M + H]+ A4
    207
    Figure US20200308190A1-20201001-C00234
    BB7a and BB104 461.2 [M + H]+ A4
    208
    Figure US20200308190A1-20201001-C00235
    BB7a and BB105 468.2 [M + H]+ A4
    209
    Figure US20200308190A1-20201001-C00236
    BB7a and BB106 494.3 [M + H]+ A4
    210
    Figure US20200308190A1-20201001-C00237
    BB7a and 4-(4- Chloro- phen- noxy) piperi- dine hydro- chloride (CAS RN 63843- 53-8) 394.1 [M + H]+ A4
    211
    Figure US20200308190A1-20201001-C00238
    BB7a and BB107 462.2 [M + H]+ A4
    212
    Figure US20200308190A1-20201001-C00239
    BB7a and BB108 522.2 [M + H]+ A4
    213
    Figure US20200308190A1-20201001-C00240
    BB7a and BB109 434.1 [M + H]+ A4
    214
    Figure US20200308190A1-20201001-C00241
    BB7a and BB110 432.1 [M + H]+ A4
    215
    Figure US20200308190A1-20201001-C00242
    BB7a and BB111 376.0 [M + H]+ A4
    216
    Figure US20200308190A1-20201001-C00243
    BB7a and BB112 442.3 [M + H]+ A4
    217
    Figure US20200308190A1-20201001-C00244
    BB7a and BB113 428.3 [M + H]+ A4
    218
    Figure US20200308190A1-20201001-C00245
    BB7a and BB114 442.1 [M + H]+ A4
    219
    Figure US20200308190A1-20201001-C00246
    BB7a and BB115 426.1 [M + H]+ A4
    220
    Figure US20200308190A1-20201001-C00247
    BB7a and BB116 454.3 [M + H]+ A4
    221
    Figure US20200308190A1-20201001-C00248
    BB7a and BB117 491.0 [M + H]+ A4
    224
    Figure US20200308190A1-20201001-C00249
    BB170 and BB1a 414.1 [M + H]+ A3
    225
    Figure US20200308190A1-20201001-C00250
    BB171 and BB1a 414.1 [M + H]+ A3
    226
    Figure US20200308190A1-20201001-C00251
    BB173 and BB1a 429.4 [M + H]+ A3
    227
    Figure US20200308190A1-20201001-C00252
    BB173 and BB1a 442.2 [M + H]+ A3 Chiral HPLC (Repro- sil Chiral NR, 60% n-hep- tane, 40% EtOH + NH4Ac
    228
    Figure US20200308190A1-20201001-C00253
    BB173 and BB1a 442.2 [M + H]+ A3 Chiral HPLC (Repro- sil Chiral NR, 60% n-hep- tane, 40% EtOH + NH4Ac
    229
    Figure US20200308190A1-20201001-C00254
    BB173 and BB1a 442.2 [M + H]+ A3 Chiral HPLC (Repro- sil Chiral NR, 60% n-hep- tane, 40% EtOH + NH4Ac
    230
    Figure US20200308190A1-20201001-C00255
    BB174 and BB1a 415.2 [M + H]+ A3
    231
    Figure US20200308190A1-20201001-C00256
    BB175 and BB1a 471.2 [M + H]+ A3 SFC: OD-H co- lumn, 20% EtOH
    232
    Figure US20200308190A1-20201001-C00257
    BB175 and BB1a 471.2 [M + H]+ A3 SFC: OD-H co- lumn, 20% EtOH
    233
    Figure US20200308190A1-20201001-C00258
    BB176 and BB1a 464.2 [M + H]+ A3
    234
    Figure US20200308190A1-20201001-C00259
    BB177 and BB1a 483.2 [M + H]+ A3
    235
    Figure US20200308190A1-20201001-C00260
    BB178 and BB1a 464.1 [M + H]+ A3 HPLC: YMC- Triart C18, 25- 45- 60- 100% ACN in water
    236
    Figure US20200308190A1-20201001-C00261
    Example 233 464.4 [M + H]+ Repro- sil Chiral NR, 70% Hep- tan, 30% EtOH + NH4Ac
    237
    Figure US20200308190A1-20201001-C00262
    Example 233 464.3 [M + H]+ Repro- sil Chiral NR, 70% Hep- tan, 30% EtOH + NH4Ac
    238
    Figure US20200308190A1-20201001-C00263
    Example 233 464.4 [M + H]+ Repro- sil Chiral NR, 70% n- hep- tane, 30% EtOH + NH4Ac
    239
    Figure US20200308190A1-20201001-C00264
    BB179 and BB1a 472.2 [M + H]+ A3
    240
    Figure US20200308190A1-20201001-C00265
    BB180 and BB1a 490.1 [M + H]+ A3
    241
    Figure US20200308190A1-20201001-C00266
    BB181 and BB1a 454.3 [M + H]+ A3
    242
    Figure US20200308190A1-20201001-C00267
    BB182 and BB1a 525.3 [M + H]+ A3
    243
    Figure US20200308190A1-20201001-C00268
    BB183 and BB1a 525.3 [M + H]+ A3
    244
    Figure US20200308190A1-20201001-C00269
    BB184 and BB1a 447.2 [M + H]+ A3
    245
    Figure US20200308190A1-20201001-C00270
    BB185 and BB1a 514.2 [M + H]+ A3
    246
    Figure US20200308190A1-20201001-C00271
    BB186 and BB1a 508.1 [M + H]+ A3
    247
    Figure US20200308190A1-20201001-C00272
    Example 246 508.0 [M + H]+ HPLC: Repro- sil Chiral NR, 60% n- hep- tane, 40% EtOH + NH4Ac
    248
    Figure US20200308190A1-20201001-C00273
    Example 246 508.0 [M + H]+ HPLC: Repro- sil Chiral NR, 60% n- hep- tane, 40% EtOH + NH4Ac
    249
    Figure US20200308190A1-20201001-C00274
    BB7a and BB187 482.1 [M + H]+ A10
    250
    Figure US20200308190A1-20201001-C00275
    BB7a and BB188 428.3 [M + H]+ A10
    251
    Figure US20200308190A1-20201001-C00276
    BB7a and BB189 444.2 [M + H]+ A10
    252
    Figure US20200308190A1-20201001-C00277
    BB7a and BB190 458.2 [M + H]+ A10
    253
    Figure US20200308190A1-20201001-C00278
    BB7a and BB191 442.3 [M + H]+ A10
    254
    Figure US20200308190A1-20201001-C00279
    BB7a and BB192 446.3 [M + H]+ A10
    255
    Figure US20200308190A1-20201001-C00280
    BB7a and BB193 462.3 [M + H]+ A10
    256
    Figure US20200308190A1-20201001-C00281
    BB194 406.4 [M + H]+ A10
    257
    Figure US20200308190A1-20201001-C00282
    BB7a and BB195 462.3 [M + H]+ A10
    258
    Figure US20200308190A1-20201001-C00283
    BB7a and BB196 446.0 [M + H]+ A10
    259
    Figure US20200308190A1-20201001-C00284
    BB7a and BB197 406.4 [M + H]+ A10
    260
    Figure US20200308190A1-20201001-C00285
    BB7a and BB199 440.1 [M + H]+ A10
    261
    Figure US20200308190A1-20201001-C00286
    BB7a and BB200 434.1 [M + H]+ A10
    262
    Figure US20200308190A1-20201001-C00287
    BB7a and BB202 453.4 [M + H]+ A10
    264
    Figure US20200308190A1-20201001-C00288
    BB7a and BB205 444.2 [M + H]+ A10
    279
    Figure US20200308190A1-20201001-C00289
    BB7a and BB206 430.2 [M + H]+ A4 ACN as sol- vent fol- lowed by prep- HPLC
    280
    Figure US20200308190A1-20201001-C00290
    BB7a and BB207 432.2 [M + H]+ A4 ACN as sol- vent fol- lowed by prep- HPLC
    281
    Figure US20200308190A1-20201001-C00291
    BB7a and BB208 448.3 [M + H]+ A4 ACN as sol- vent fol- lowed by prep- HPLC
    282
    Figure US20200308190A1-20201001-C00292
    BB7a and 3-(3- (Tri- fluoro- methoxy) benzyl) aze- tidine hydro- chloride (CAS RN 1354963- 49-7) 414.3 [M + H]+ A4 ACN as sol- vent fol- lowed by prep- HPLC
    283
    Figure US20200308190A1-20201001-C00293
    BB7a and BB209 432.2 [M + H]+ A4 ACN as sol- vent fol- lowed by MPLC (n-hep- tane: EtOAc/ EtOH/ 3/1 (70:30 to 10:90)
    284
    Figure US20200308190A1-20201001-C00294
    BB7a and BB210 448.2 [M + H]+ A4 ACN as sol- vent fol- lowed by MPLC (n-hep- tane: EtOAc/ EtOH/ 3/1 (70:30 to 10:90)
    285
    Figure US20200308190A1-20201001-C00295
    Example 283 432.2 [M + H]+ B3
    286
    Figure US20200308190A1-20201001-C00296
    Example 283 432.2 [M + H]+ B3
    287
    Figure US20200308190A1-20201001-C00297
    BB1a and BB211 374.2 [M + H]+ A3
    288
    Figure US20200308190A1-20201001-C00298
    BB7a and BB212 448.3 [M + H]+ A4
    289
    Figure US20200308190A1-20201001-C00299
    BB1a and BB213 428.2 [M + H]+ A3
    290
    Figure US20200308190A1-20201001-C00300
    BB7a and BB214 374.2 [M + H]+ A4 (ACN as sol- vent)
    291
    Figure US20200308190A1-20201001-C00301
    BB7a and BB215 374.2 [M + H]+ A4 (ACN as sol- vent)
    292
    Figure US20200308190A1-20201001-C00302
    BB7a and BB216 428.2 [M + H]+ A4 (ACN as sol- vent)
    293
    Figure US20200308190A1-20201001-C00303
    Example 216 427.2 [M + H]+ G
    294
    Figure US20200308190A1-20201001-C00304
    BB217 428.2 [M + H]+ A3
    295
    Figure US20200308190A1-20201001-C00305
    BB1a and BB218 448.1 [M + H]+ A3
  • Example 222 (4aR,8aS)-6-[3-[[6-Fluoro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00306
  • Step a) tert-Butyl 3-[[6-chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (CAS Nr. 142253-56-3) (2.60 g, 13.9 mmol) and 2,6-dichloro-4-(trifluoromethyl)pyridine (CAS Nr. 39890-98-7) (3.00 g, 13.9 mmol) in THF (60 mL) was added NaH (60%, 1.11 g, 27.8 mmol) and the mixture was stirred 3 h at 25° C. The solution was poured into sat. aq. NH4Cl (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layers were concentrated under vacuum to give crude tert-butyl 3-[[6-chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate (3.00 g, 59%) as colorless oil, which was used directly in the next step. LC-MS (ESI): m/z=367.1 [M+H]+.
  • Step b) 2-(Azetidin-3-ylmethoxy)-6-chloro-4-(trifluoromethyl)pyridine
  • A solution of trifluoroacetic acid (6.3 mL, 81.8 mmol, 10 eq) and tert-butyl 3-[[6-chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate (3.00 g, 8.18 mmol) in DCM (30 mL) was stirred at 25° C. for 4 h. The solution was concentrated under vacuum to give a residue, which was purified by Prep-HPLC (HCl condition) to give 2-(azetidin-3-ylmethoxy)-6-chloro-4-(trifluoromethyl)pyridine (1.00 g, 46%) as white solid. LC-MS (ESI): m/z=267.0 [M+H]+.
  • Step c) (4aR,8aS)-6-[3-[[6-Chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • A solution of 2-(azetidin-3-ylmethoxy)-6-chloro-4-(trifluoromethyl)pyridine (150 mg, 0.560 mmol), N,N-diisopropylethylamine (0.29 mL, 1.69 mmol) and 4-nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate (BB7a) (199 mg, 0.620 mmol) in ACN (5 mL) was stirred at 25° C. for 16 h. The solution was concentrated under vacuum to give a residue, which was purified by prep-HPLC (TFA conditions) to give (4aR,8aS)-6-[3-[[6-chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (10×) mg, 40%) as colorless oil. LC-MS (ESI): m/z=449.2 [M+H]+.
  • Step d) (4aR,8aS)-6-[3-[[6-Fluoro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • A solution of (4aR,8aS)-6-[3-[[6-chloro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (75 mg, 0.17 mmol) and cesium fluoride (101 mg, 0.670 mmol) in DMSO (3 mL) was stirred at 80° C. for 16 h. The solution was filtered and purified by prep-HPLC (TFA conditions) to give (4aR,8aS)-6-[3-[[6-fluoro-4-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (22 mg, 28%) as white solid. LC-MS (ESI): m/z=433.0 [M+H]+.
  • Example 223 (4aR,8aS)-6-[3-[[6-Fluoro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00307
  • Step a) tert-Butyl 3-[[6-chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (CAS Nr. 142253-56-3) (1.56 g, 8.33 mmol) in THF (50 mL) was added NaH (60%. 741 mg, 18.5 mmol) followed by 2,6-dichloro-3-(trifluoromethyl)pyridine (CAS Nr. 55304-75-1) (2.00 g, 9.26 mmol). The resulting mixture was stirred at 25° C. for 3 h. The solution was poured into sat.aq. NH4Cl (50 mL) and extracted with EtOAc (2×30 mL). The combined organic layers were concentrated under vacuum to give a residue, which was purified by flash column chromatography (petroleum ether:EtOAc=5:1) to give tert-butyl 3-[[6-chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate (1.10 g, 32%) as colorless oil. LC-MS (ESI): m/z=311.0 [M−56+H]+.
  • Step b) 6-(Azetidin-3-ylmethoxy)-2-chloro-3-(trifluoromethyl)pyridine
  • A solution of trifluoroacetic acid (0.37 mL, 4.8 mmol) and tert-butyl 3-[[6-chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carboxylate (1.1 g, 3.0 mmol) in DCM (30 mL) was stirred at 25° C. for 4 h. The solution was concentrated under vacuum to give a residue, which was purified by Prep-HPLC (HCl condition) to give 6-(azetidin-3-ylmethoxy)-2-chloro-3-(trifluoromethyl)pyridine (600) mg, 75%) as white solid. LC-MS (ESI): m/z=267.0 [M+H]+.
  • Step c) (4aR,8aS)-6-[3-[[6-Chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • To a solution of 6-(azetidin-3-ylmethoxy)-2-chloro-3-(trifluoromethyl)pyridine (100 mg, 0.380 mmol) and 4-nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxane-6(5H)-carboxylate (BB7a) (120 mg, 0.380 mmol) in ACN (5 mL) was added N,N-diisopropylethylamine (0.13 mL, 0.75 mmol) with stirring at 25° C. The solution was stirred at 25° C. for 16 h. The solution was concentrated under vacuum to give a residue, which was purified by Prep-HPLC (TFA conditions) to give (4aR,8aS)-6-[3-[[6-chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (76 mg, 45%) as white solid. LC-MS (ESI): m/z=449.1 [M+H]+.
  • Step d) (4aR,8aS)-6-[3-[[6-Fluoro-5-(trifluoromethyl)-2-pyridyl]ox methyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • A solution of (4aR,8aS)-6-[3-[[6-chloro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (70 mg, 0.16 mmol) and cesium fluoride (95 mg, 0.62 mmol) in DMSO (3 mL) was stirred at 60° C. for 24 h. The solution was filtered and then purified by Prep-HPLC (TFA conditions) to give (4aR,8aS)-6-[3-[[6-fluoro-5-(trifluoromethyl)-2-pyridyl]oxymethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (38 mg, 49%) as white solid. LC-MS (ESI): m/z=433.3 [M+H]+.
  • Synthesis of Building Blocks BB1a & BB1b (+)-cis-4a,5,6,7,8,8a-Hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one and (−)-cis-4a,5,6,7,8,8a-Hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one
  • The enantiomers of rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one dihydrochloride (BB 1, 500 mg, 2.18 mmol, ChemBridge Corporation) were separated by preparative chiral HPLC (ReprosilChiral NR column) using an isocratic mixture of EtOH (containing 0.05% of NH4OAc):n-heptane (30:70).
  • First eluting enantiomer: (+)-cis-4a,5,6,7,8,8a-Hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one (BB1a). Yellow solid (0.150 g; 44.0%). MS (ESI): m/z=157.1 [M+H]+.
  • Second eluting enantiomer: (−)-cis-4a,5,6,7,8,8a-Hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one. (BB1b). Yellow solid (0.152 g; 44.6%). MS (ESI): m/z=157.1 [M+H]+.
  • BB2 (4-Nitrophenyl) 4-[[4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • To a solution of 4-(4-(trifluoromethyl)benzyl)piperidine (10×) mg, 411 μmol, CAS RN 192990-03-7) in DCM (1 mL), TEA (83.2 mg, 115 μL, 822 μmol) was added. On cooling to 0° C. 4-nitrophenyl carbonochloridate (91.1 mg, 452 μmol, CAS RN 7693-46-1) was added, the reaction mixture was allowed to warm to RT and stirred for 18 hours. The reaction mixture was diluted with DCM and subsequently washed with H2O and sat. aqueous NaHCO3 solution. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The crude material was purified by flash chromatography (silica 10 g, eluting with EtOAc/Heptane 0-50%), to afford title compound as a light yellow solid. (0.165 g; 98.3%). MS (ESI): m/z=409.3 [M+H]+.
  • BB3 rac-(4aR,8aS)-6-(Piperazine-1-carbonyl)-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • To a mixture of rac-tert-butyl 4-((4aR,8aS)-3-oxooctahydro-2H-pyrido[4,3-b][1,4]oxazine-6-carbonyl)piperazine-1-carboxylate (100 mg, 271 μmol) in DCM (3 mL) was added TFA (155 mg, 105 μL, 1.36 mmol) and the mixture was stirred at RT for 15 h under an argon atmosphere.
  • The reaction mixture was washed with a saturated aqueous NaHCO3 solution. The H2O layer was concentrated in vacuo to give a white solid which was triturated with DCM for 30 min. before it was filtered. The filtrate was concentrated to give a light yellow gum (70 mg, 96.1%). MS (ESI): nm/z=269.3 [M+H]+.
  • Step a) rac-tert-Butyl 4-((4aR,8aS)-3-oxooctahydro-2H-pyrido[4,3-b][1,4]oxazine-6-carbonyl)piperazine-1-carboxylate
  • To a mixture of triphosgene (1.29 g, 4.36 mmol) and Na2CO3 (1.98 g, 18.7 mmol) in THF (3 mL) at 0° C. were added dropwise a solution of tert-butyl piperazine-1-carboxylate (1.16 g, 6.23 mmol, CAS RN 57260-71-6) in THF (90 mL). The reaction mixture was stirred for 10 min. at 0° C., then allowed to warm up to RT and stirring was continued at RT for 5 h. The suspension was filtered off and the filtrate was concentrated in vacuo. The residue was dissolved in THF (40 mL) and added dropwise to a stirred suspension of rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one hydrochloride (1200 mg, 6.23 mmol, Chembridge Corporation) and DIPEA (4.83 g, 6.53 mL, 37.4 mmol) in THF (40 mL) at 0° C. After 30 min. at 0° C., the reaction mixture was allowed to warm up to RT, and stirred at RT for 15 h. The mixture was filtered and the filtrate concentrated in vacuo. The residue was diluted with DCM and washed with water, aq. NaHCO3 solution and brine. The organic layer was dried over Na2SO4, filtered and concentrated to give a white solid (1.13 g, 58.6%). MS (ESI): m/z=313.3 [M+H]+.
  • BB4 (4-Nitrophenyl) 4-(phenoxymethyl)piperidine-1-carboxylate
  • The compound was prepared in analogy to BB2 using 4-(phenoxymethyl)piperidine (CAS N63614-86-8) to afford title compound as a white solid which was used in the next step without further purification.
  • BB5 2-(4-Piperidylmethyl)-5-(trifluoromethyl)pyridine; hydrochloride salt
  • tert-Butyl 4-[[5-(trifluoromethyl)-2-pyridyl]methyl]piperidine-1-carboxylate (320 mg, 0.930 mmol) was dissolved in 4 M HCl in EtOAc (10.0 mL, 40 mmol) and the solution stirred at 20° C. for 2 h. The mixture was concentrated to yield the desired compound as light yellow solid (0.259, 94.8%). MS (ESI): m/z=245.0 [M−HCl+H]+.
  • Step a) tert-Butyl 4-[[5-(trifluoromethyl)-2-pyridyl]methyl]piperidine-1-carboxylate
  • 2-Bromo-5-(trifluoromethyl)pyridine (500.0 mg, 2.21 mmol, CAS RN 1000773-62-5) was degassed before 9-BBN solution 0.5 M in THF (4.87 mL, 2.43 mmol, CAS RN 280-64-8) was added. The resulting solution was refluxed for 1 h. After cooling to RT, the solution was added to a solution of tert-butyl 4-methylenepiperidine-1-carboxylate (480.1 mg, 2.43 mmol, CAS RN 159635-49-1), [1,1′-bis(diphenylphosphino)ferrocene]palladium (II) chloride (161.89 mg, 0.220 mmol, CAS RN 72287-26-4) and K2CO3 (611.56 mg, 4.42 mmol) in DMF (5 mL) and water (0.5 mL). The resulting mixture was heated at 80° C. for 4 h. The mixture was cooled to RT and poured into water, the pH was adjusted to 11 with 10% aqueous NaOH and the mixture was extracted with EtOAc. The combined organic extracts were washed with brine, dried over Na2SO4, filtered, and evaporated to give a crude oil, which was purified by column chromatography (silica adsorbent; gradient of PE:EtOAc 10:1 then 5:1) to yield the desired compound as a light yellow oil (320 mg, 0.930 mmol, 42%). MS (ESI): m/z=289.0 [M−C4H8+H]+.
  • BB6 rac-(4aS,8aS)-Hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • rac-Benzyl (4aS,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate (125 mg, 431 μmol) was dissolved in MeOH (5 mL). The reaction solution was degassed in vacuo and backfilled with argon. Pd—C(20 mg, 188 μmol) was added under an argon atmosphere. Argon was evacuated from the reaction mixture and backfilled with hydrogen. The reaction mixture was stirred at RT for 15 h under a hydrogen atmosphere. The reaction mixture was filtered through a syringe filter and concentrated in vacuo to afford the desired product as a white solid (62 mg, 92.2%). MS (ESI): m/z=157.098 [M+H]+.
  • Step a) rac-Benzyl (3S,4S)-3-(2-chloroacetamido)-4-hydroxypiperidine-1-carboxylate
  • To a stirred suspension of rac-benzyl (3S,4S)-3-amino-4-hydroxypiperidine-1-carboxylate (317 mg, 1.27 mmol, synthesized according to patent US 2011/59118 A1) and sodium acetate (208 mg, 2.53 mmol, CAS RN 127-09-3) in a mixture of acetone (4 mL)/H2O (0.5 mL) was added dropwise a solution of chloroacetyl chloride (150 mg, 107 μL, 1.33 mmol, CAS RN 79-04-9) in acetone (3 mL) between 0-5° C. After the addition the reaction mixture was stirred at RT for 1 h and subsequently evaporated to dryness giving a yellow gum. The crude product was purified by silica gel chromatography to afford the desired product as a yellow solid (385 mg, 93%). MS (ESI): m/z=325.2 [M−H]+.
  • Step b) rac-Benzyl (4aS,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate
  • To a stirred solution of rac-Benzyl (3S,4S)-3-(2-chloroacetamido)-4-hydroxypiperidine-1-carboxylate (385 mg, 1.18 mmol) in dry THF (4 mL) was added NaH (67.9 mg, 1.7 mmol) at 0° C. The mixture was allowed to reach RT and then stirred for 90 min under an argon atmosphere. H2O (5 mL) was added and stirring was continued for 10 min at RT. THF was removed in vacuo from the reaction mixture. The residue was treated with DCM and the organic phase was washed with H2O and brine, dried over Na2SO4, filtered and then concentrated in vacuo. The residue was purified by flash chromatography (12 g reversed phase column, gradient 0-100% ACN (0.1% FA) in water (0.1% FA) to afford the desired product as a white solid (133 mg, 38.9%). MS (ESI): m/z=291.3 [M+H]+.
  • BB7a and BB7b 4-Nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate BB7a and 4-nitrophenyl (4aS,8aR)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate BB7b
  • To a suspension of rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one; dihydrochloride salt (4.5 g, 19.6 mmol, BB1) in dry DCM (125 mL) at 0° C. was added DIPEA (6.35 g, 8.58 mL, 49.1 mmol) followed by 4-nitrophenyl carbonochloridate (4.35 g, 21.6 mmol). The reaction mixture was stirred at 0° C. for 10 min and at RT for 2 h. The crude reaction was diluted with DCM and transferred into a separating funnel for extraction with sat. aq. Na2CO3 solution. The organic phase was collected and the aqueous phase was back-extracted with DCM. The combined organic phases were dried over Na2SO4 and evaporated down to dryness to yield 6.62 g of a crude racemic product (BB7) as a yellow solid. The crude material was directly submitted for a chiral SFC separation to yield enantiomer BB7b (2.72 g, second eluting to enantiomer) as a yellow solid and enantiomer BB7a (3.25 g, first eluting enantiomer) as a light beige solid but contaminated with BB7b. A further SFC chiral separation was carried out to yield 2.71 g of BB7a. MS (ESI): m/z=322.2 [M+H]+ for both enantiomers.
  • BB8 5-tert-Butyl-2-(4-piperidylmethyl)oxazole; hydrochloride salt
  • A solution of tert-butyl 4-[(5-tert-butyloxazol-2-yl)methyl]piperidine-1-carboxylate (167 mg, 518 μmol) in HCl 2M in diethyl ether (2.59 mL, 5.18 mmol) was stirred at RT for 5 h before another 1.29 mL (2.59 mmol) of HCl 2M in diethyl ether was added. The white suspension was stirred at RT overnight. The mixture was cooled down in an ice-bath, then filtered and washed with diethyl ether to get the desired compound as a colorless solid (0.126 g, 94.0%). MS (ESI): m/z=223.2 [M+H]+.
  • Step a) (5-tert-Butyloxazol-2-yl)methyl-triphenyl-phosphonium bromide
  • To a solution of 2-(bromomethyl)-5-(tert-butyl)oxazole (600 mg, 2.75 mmol, CAS RN 1334492-54-4) in diethyl ether (5 mL) was added triphenylphosphine (722 mg, 2.75 mmol, CAS RN 603-35-0) and the mixture was stirred at RT for 64 h. The suspension was cooled down in an ice-bath and then filtered. The filter cake was washed a small volume of cold diethyl ether to give the desired compound as a light yellow solid (0.864 g, 65.4%). MS (ESI): m/z=400.2 [M−Br+H]+.
  • Step b) tert-Butyl 4-[(5-tert-butyloxazol-2-yl)methylene]piperidine-1-carboxylate
  • To an ice-cold suspension of (5-tert-butyloxazol-2-yl)methyl-triphenyl-phosphonium bromide (355 mg, 739 μmol) in THF (7 mL) was added potassium tert-butylate 1M solution in THF (738 μL, 738 μmol) and the reaction stirred at this temperature for 15 min. Then, tert-butyl 4-oxopiperidine-1-carboxylate (162 mg, 813 μmol, CAS RN 79099-07-3) was added to the turbid, orange solution and stirring was continued at 0° C. for another 15 min., then at RT for 42 h. The reaction mixture was poured on half-saturated aqueous NH4Cl solution and EtOAc and the layers were separated. The aqueous layer was extracted twice with EtOAc. The combined organic layers were dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 12 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (100:0 to 50:50) to provide the desired compound as a colorless solid (0.180 mg; 76.0%). MS (ESI): m/z=321.3 [M+H]+.
  • Step c) tert-Butyl 4-[(5-tert-butyloxazol-2-yl)methyl]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[(5-tert-butyloxazol-2-yl)methylene]piperidine-1-carboxylate (180 mg, 562 μmol) in MeOH (1 mL) and EtOAc (1 mL) was added Pd/C 10% (17.9 mg, 16.9 μmol) and the suspension was stirred under a hydrogen atmosphere at 1.3 bar for 2 h. The suspension was filtered over a microfilter and the filtrate was evaporated to get the desired compound as a colorless oil (0.167 g; 92.2%). MS (ESI): m/z=323.3 [M+H]+.
  • BB12 4-[(2-Chloro-4-fluoro-phenoxy)methyl]-4-methyl-piperidine; hydrochloride salt
  • To a solution of tert-butyl 4-[(2-chloro-4-fluoro-phenoxy)methyl]-4-methyl-piperidine-1-carboxylate (186 mg, 0.520 mmol) in EtOAc (1.5 mL) was added HCl in EtOAc (4 M, 1.5 mL) at 0° C. The solution was stirred at 15° C. for 3 h. The solution was concentrated under vacuum, then dried by lyophilization to give desired product as a white solid (64.0 mg, 0.220 mmol, 40.3% yield). MS (ESI): m/z=258 [M+H]+.
  • Step a) tert-Butyl 4-methyl-4-(methylsulfonyloxyethyl)piperidine-1-carboxylate
  • To a solution of tert-butyl 4-(hydroxymethyl)-4-methyl-piperidine-1-carboxylate (500 mg, 2.14 mmol, CAS RN: 614730-97-1) in DCM (5 mL) was added NEt3 (0.45 mL, 3.22 mmol) and methanesulfonyl chloride (0.23 mL, 3.0 mmol) at 0° C. The mixture was stirred at 0° C. for 2 h. The mixture was washed twice with water (3 mL each) at 0° C., and dried over Na2SO4. The organic layer was concentrated in vacuum to yield the desired compound as colorless oil (766 mg, 2.46 mmol, 98.5%) which was used in the next step without further purification. MS (ESI): m/z=256 [M−56+H]+.
  • Step b) tert-Butyl 4-[(2-chloro-4-fluoro-phenoxy)methyl]-4-methyl-piperidine-1-carboxylate
  • To a solution of tert-butyl 4-methyl-4-(methylsulfonyloxymethyl)piperidine-1-carboxylate (450 mg, 1.46 mmol) in DMF (5 mL) was added Cs2CO3 (620 mg, 1.9 mmol) and 2-chloro-4-fluorophenol (0.14 mL, 1.46 mmol) at 15° C. The mixture was heated to 90° C. and stirred for 16 h. The reaction solution was diluted by EtOAc (10 mL), washed twice with brine (10 mL each), and dried over Na2SO4. The organic layer was concentrated under vacuum to give the crude product (0.7 g) as light yellow oil. The crude product was purified by prep-HPLC and dried by lyophilization to give the desired compound as colorless solid (186 mg, 0.520 mmol, 35.5% yield). MS (ESI): m/z=302 [M−56+H]+.
  • BB15 4-[(2-Chloro-4-fluoro-phenoxy)methyl]-4-fluoro-piperidine; hydrochloride salt
  • To a solution of tert-butyl 4-1 (2-chloro-4-fluoro-phenoxy)methyl-4-fluoro-piperidine-1-carboxylate (220 mg, 0.610 mmol) in EtOAc (2 mL) was added HCl/EtOAc (0.4 mL, 3.6 mmol) at 0° C. The solution was stirred at 15° C. for 2.5 h. The solution was concentrated in vacuo, then dried by lyophilization to give desired product as a white solid (136.7 mg, 75.4%).
  • Step a) tert-Butyl 4-fluoro-4-(methylsulfonyloxymethyl)piperidine-1-carboxylate
  • To a solution of tert-butyl 4-fluoro-4-(hydroxymethyl)piperidine-1-carboxylate (500 mg, 2.14 mmol) in DCM (5 mL) was added NEt3 (0.45 mL, 3.22 mmol) and methanesulfonyl chloride (0.23 mL, 3 mmol) at 0° C. The mixture was stirred at 0° C. for 2 h. The mixture was washed twice with H2O (3 mL each) at 0° C., and dried over Na2SO4. The organic layer was concentrated to provide the compound as a colorless oil (766 mg, 98.5%) which was used in next step without further purification. MS (ESI): m/z=256 [M−56+H]+.
  • Step b) tert-Butyl 4-[(2-chloro-4-fluoro-phenoxy)methyl]-4-fluoro-piperidine-1-carboxylate
  • To a solution of tert-butyl 4-fluoro-4-(methylsulfonyloxymethyl)piperidine-1-carboxylate (383 mg, 1.23 mmol) in DMF (4 mL) was added Cs2CO3 (601 mg, 1.85 mmol), 2-chloro-4-fluorophenol (0.13 mL, 1.35 mmol) and 2-chloro-4-fluorophenol (0.13 mL, 1.35 mmol) at 15° C. The mixture was heated to 85° C. and stirred for 16 h. The mixture was extracted three times with EtOAc (5 mL each) at 15° C., the combined organic layers washed three times with brine (5 mL each), dried over Na2SO4, filtered and evaporated. The crude product was purified by preparative HPLC and dried by lyophilization to give the desired compound as light yellow oil (275 mg, 0.760 mmol, 61.5%). MS (ESI): m/z=306 [M−56+H]+.
  • BB16 rac-(4aR,8aS)-6-(4-(Hydroxymethyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • To a suspension of rac-(4aR,8aS)-hexahydro-2H-pyrido[4,3-b][14]oxazin-3(4H)-one; dihydrochloride salt (450 mg, 1.96 mmol, BB1) in dry DMF (9 mL) cooled down to 0° C. under an inert atmosphere was added DIPEA (787 mg, 1.06 mL, 6.09 mmol) and 4-nitrophenyl carbonochloridate (475 mg, 2.36 mmol). The reaction mixture was stirred at 0° C. for 30 min. Piperidin-4-ylmethanol (271 mg, 2.36 mmol, CAS RN 6457-49-4) and DIPEA (381 mg, 515 μL, 2.95 mmol) were added, and the reaction mixture was stirred at 100° C. for 14 h. Volatiles were removed in vacuo and the crude residue was purified by flash chromatography with a 24 g SiO2 column using an eluent mixture of DCM and MeOH (5% to 25%). The crude product was submitted for SFC purification to yield the desired compound as a light yellow oil (338 mg). MS (ESI): m/z=298.3 [M+H]+.
  • BB17 4,4-Difluoro-1-(piperidin-4-ylmethyl)piperidine; dihydrochloride salt
  • To a solution of tert-butyl 4-((4,4-difluoropiperidin-1-yl)methyl)piperidine-1-carboxylate (453 mg, 1.07 mmol) in dioxane (2.5 mL) was added HCl (4.0M solution in dioxane) (2.67 mL, 10.7 mmol) and the reaction mixture was stirred at room temperature for 14 h. Volatiles were removed in vacuo to yield the desired compound as a white solid (286 mg) which was used in the next step without further purification. MS (ESI): m/z=219.3 [M+H]+.
  • Step a) tert-Butyl 4-((4,4-difluoropiperidin-1-yl)methyl)piperidine-1-carboxylate
  • To a solution of a tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (0.5 g, 1.8 mmol, CAS RN: 158407-04-6) in dry DMF (4 mL) was added 4,4-difluoropiperidine; dihydrochloride salt (425 mg, 2.7 mmol) and Cs2CO3 (1.17 g, 3.59 mmol). The reaction mixture was then stirred at 80° C. under microwave radiation for 60 min. Insolubles were removed by filtration, the filtrate was then concentrated in vacuo, and the obtained crude residue was suspended in DCM and filtered through a pad of Celite to give a crude yellow oil, which was purified by flash chromatography on a SiO2 column, using an eluent mixture of n-heptane and EtOAc (10% to 60%) to yield the desired product as a colorless oil (453 mg). The compound was carried forwards to the next step without further purification. MS (ESI): m/z=319.3 [M+H]+.
  • BB19 N-(azetidin-3-ylmethyl)-22,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethan-1-amine; bis(trifluoroacetate) salt
  • To a solution of tert-butyl 3-(((2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate (1 g, 2.42 mmol) in DCM (10 mL) was added TFA (5.53 g, 3.74 mL, 48.5 mmol). The resulting reaction mixture was stirred at RT for 1 h. The reaction mixture was concentrated in vacuo to yield the desired compound as colorless oil (1.29 g). MS (ESI): m/z=313.5 [M+H]+.
  • Step a) tert-Butyl 3-(((2,2,2-trifluoro-1-(4-trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate
  • To a dry flask with septum was added under nitrogen tert-butyl 3-(aminomethyl)azetidine-1-carboxylate (0.852 g, 4.57 mmol), triethylamine (1.39 g, 1.91 mL, 13.7 mmol), 2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethan-1-one (1.11 g, 780 μL, 4.57 mmol), and dry DCM (28 mL). Titanium tetrachloride 1 M in DCM (2.29 mL, 2.29 mmol) was added via a syringe to the ice-cooled flask (exothermic). The reaction was stirred overnight at RT, then carefully quenched with a solution of NaCNBH3 (862 mg, 13.7 mmol) in MeOH (8.79 g, 11.1 mL, 274 mmol) and stirred overnight. The reaction was basified with sat. NaHCO3 solution. The obtained insoluble material was filtered off over celite. Extraction of the filtrate with DCM, the organic layers were combined, washed with brine, dried over Na2SO4 and concentrated. The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in n-heptane to yield tert-butyl 3-(((2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate which was used in the next step without further purification.
  • BB26 3-Chloro-4-(4-piperidylmethoxy)benzonitrile; hydrochloride salt
  • To a solution of tert-butyl 4-[(2-chloro-4-cyano-phenoxy)methyl]piperidine-1-carboxylate (300 mg, 0.860 mmol) in EtOAc (3 mL) was added HCl in EtOAc (4M, 2.0 mL) at 0° C. The solution was stirred at 15° C. for 3 h. The solution was concentrated in vacuo, then dried by lyophilization to give desired product as a white solid (238 mg, 0.830 mmol, 96% yield). MS (ESI): m/z=251 [M+H]+.
  • Step a) tert-Butyl 4-(((methylsulfonyl)oxy)methyl)piperidine-1-carboxylate
  • To a solution of N-Boc-4-piperidinemethanol (10.0 g, 46.5 mmol, 1 eq) in DCM (200 mL) was added NEt3 (7.04 g, 69.7 mmol), then methanesulfonyl chloride (3.95 mL, 51.1 mmol) was added at 0° C. and the mixture was stirred at 0° C. for 1 h. The mixture was poured into ice-water, the aqueous phase was extracted twice with DCM (50 mL each). The combined organic layers were washed with brine (50 mL), and concentrated under vacuum. The residue was directly used without any purification. MS (ESI): m/z=238.1 [M+H].
  • Step b) tert-Butyl 4-[(2-chloro-4-cyano-phenoxy)methyl]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-(methylsulfonyloxymethyl)piperidine-1-carboxylate (700 mg, 2.39 mmol) in DMF (7 mL) was added Cs2CO3 (855 mg, 2.62 mmol) and 3-chloro-4-hydroxybenzonitrile (0.25 mL, 2.39 mmol) at 15° C. The mixture was heated to 85° C. and stirred for 16 h. The reaction mixture was diluted with EtOAc (8 mL) at 15° C., washed three times with brine (8 mL each), the combined organic layers were dried over Na2SO4 and evaporated. The colorless residue (0.75 g) was purified by prep-HPLC and dried by lyophilization to give the desired product as a white solid (531 mg, 1.51 mmol, 53.4%). MS (ESI): m/z=295 [M−56+H]+.
  • BB27 4-((4-(Trifluoromethyl)-1H-imidazol-1-yl)methyl)piperidine; hydrochloride salt
  • To a solution of tert-butyl 4-((4-(trifluoromethyl)-1H-imidazol-1-yl)methyl)piperidine-1-carboxylate (430 mg, 1.29 mmol) in dioxane (3 mL) was added HCl (4 M solution in dioxane: 3.22 mL, 12.9 mmol) and the reaction mixture was stirred at RT for 14 h. Volatiles were removed in vacuo to give the crude product (362 mg) which was used in the next step without further purification. MS (ESI): m/z=234.2 [M+H]+.
  • Step a) tert-Butyl 4-((4-(trifluoromethyl)-1H-imidazol-1-yl)methyl)piperidine-1-carboxylate
  • To a solution of a tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (0.5 g, 1.8 mmol, CAS RN: 158407-04-6) in dry DMF (4 mL) was added 4-(trifluoromethyl)-1H-imidazole (293 mg, 2.16 mmol) and Cs2CO3 (1.17 g, 3.59 mmol). The reaction mixture was then stirred at 80° C. for 14 h. Insolubles were removed by filtration, and the filtrate was concentrated in vacuo. The crude residue was suspended in DCM and filtered through a pad of Celite to give a yellow oil, which was purified by flash chromatography with a SiO2 column, using an eluent mixture of n-heptane and EtOAc (10% to 90° 6). This yielded the first fraction (301 mg) of the desired product as a colorless oil, and a second fraction (261 mg) of a mixture of the desired product with impurities. The second fraction was submitted for SFC purification, and the purified product was combined with the first fraction to yield 430 mg of the desired product as a colorless oil. MS (ESI): m/z=334.2 [M+H]+.
  • BB29 3-((2-Chloro-4-(trifluoromethyl)phenoxy)methyl)azetidine
  • Trifluoroacetic acid (2 g, 1.35 mL, 17.5 mmol) was added to a solution of tert-butyl 3-((2-chloro-4-(trifluoromethyl)phenoxy)methyl)azetidine-1-carboxylate (320 mg, 875 μmol) in DCM (4.37 mL) and the solution was stirred at RT for 2 h. The solvent was removed under reduced pressure and the resulting pale oil (470 mg) was diluted with EtOAc and washed with aq. Na2CO3 solution. The aqueous phase was extracted three times with EtOAc, and the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure to afford the compound as a yellow oil (259 mg, 877 μmol). MS (ESI): m/z=266.1 to [M+H]+.
  • Step a) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenoxy)methyl)azetidine-1-carboxylate
  • To a solution of 2-chloro-4-(trifluoromethyl)phenol (525 mg, 357 μL, 2.67 mmol), tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (500 mg, 2.67 mmol, CAS RN: 142253-56-3) and triphenylphosphine (770 mg, 2.94 mmol) in DCM (13.4 mL) was added DIAD (594 mg, 571 μL, 2.94 mmol) dropwise and the reaction was stirred at RT for 17 h. The reaction mixture was quenched by addition of sat. aq. NaHCO3 solution (20 mL). The phases were separated and the aq. phase was extracted with DCM twice. The combined organic layers were dried over Na2SO4 and concentrated to dryness. The residue was dissolved in EtOH (7 mL) and a homogeneous solution of zinc chloride (218 mg, 1.6 mmol) in EtOH (2 mL, 0.5 M) was added. The mixture was stirred for 30 min during which a white solid precipitated. The white solid was filtered off and washed with EtOH The filtrate was concentrated to give a yellow oil with a white precipitate. The crude was immobilized on Isolute and purified by column chromatography (40 g, 0 to 30% EtOAc in heptanes) to afford the title compound as a white solid (764.6 mg, 1.99 mmol, 74.4%). MS (ESI): m/z=310.1 [M−56+H]+.
  • BB30 N-benzyl-N-(2-hydroxyethyl)piperidine-4-carboxamide hydrochloride
  • To a solution of tert-butyl 4-(benzyl(2-hydroxyethyl)carbamoyl)piperidine-1-carboxylate (0.080 g, 221 μmol) in DCM (1 mL) was added HCl 2 M in diethyl ether (1.1 mL, 2.21 mmol). The resultant reaction mixture was stirred at RT for 1 h and then concentrated under vacuum at 22° C. to yield the desired compound as a colorless oil (63 mg) (BB30). MS (ESI): m/z=263.18 [M+H]+.
  • Step a) tert-Butyl 4-(benzyl(2-hydroxyethyl)carbamoyl)piperidine-1-carboxylate
  • In a 10 mL glastube, to 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (0.1 g, 436 μmol) in DMF (2 mL) was added 2-(benzylamino)ethan-1-ol (72.5 mg, 480 μmol), DIPEA (169 mg, 229 μL, 1.31 mmol) and HATU (182 mg, 480 μmol), stirred at RT for 1 h and extracted with H2O/DCM. The crude material was purified by flash chromatography (silica gel, 20 g, 50% to 100% EtOAc in n-heptane) to yield the compound as a light yellow oil (156 mg).
  • BB31 N-benzylpiperidine-4-carboxamide hydrochloride
  • tert-Butyl 4-(benzylcarbamoyl)piperidine-1-carboxylate (0.138 g, 433 μmol) was dissolved in DCM (1 mL) and HCl 2M in diethyl ether (2.17 mL, 4.33 mmol) was added. The reaction mixture was stirred for 2 h. The residue was concentrated in vacuo to yield the compound (108 mg) as a colorless oil. MS (ESI): m/z=219.15 [M+H]+.
  • Step a) tert-Butyl 4-(benzylcarbamoyl)piperidine-1-carboxylate
  • In a 10 mL glastube, to 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (0.1 g, 436 μmol) in DMF (2 mL) was added phenylmethanamine (51.4 mg, 52.4 μL, 480 μmol), DIPEA (169 mg, 229 ML, 1.31 mmol) and HATU (182 mg, 480 μmol), stirred at RT for 2 h and extracted with H2O/DCM. The crude material was purified by flash chromatography (silica gel, 20 g, 50% to 100% EtOAc in n-heptane) to yield the compound as a colorless oil (0.138 g).
  • BB32 4-((4-(tert-Butyl)-1H-pyrazol-1-yl)methyl)piperidine; hydrochloride salt
  • To a solution of tert-butyl 4-((4-(tert-butyl)-1H-pyrazol-1-yl)methyl)piperidine-1-carboxylate (100 mg, 311 μmol) in dioxane (I mL) was added HCl (4.0M solution in dioxane; 1.17 mL, 4.67 mmol) and the reaction mixture was stirred at RT for 14 h. Volatiles were removed in vacuo to give 84 mg of a crude product which was used in the next step without further purification. MS (ESI): m/z=222.3 [M+H]+.
  • Step a) tert-Butyl 4-4-((4-tert-butyl)-1H-pyrazol-1-yl)methyl)piperidine-1-carboxylate
  • To a solution of a tert-butyl 4-(bromomethyl)piperidine-1-carboxylate (0.5 g, 1.8 mmol, CAS RN 158407-04-6) in dry DMF (4 mL) was added 4-(tert-butyl)-1H-pyrazole (268 mg, 2.16 mmol) and NaH (86.3 mg, 2.16 mmol). The reaction mixture was stirred at 80° C. for 14 h. The reaction was quenched by addition of few drops of sat. aq. NH4Cl solution, and transferred into a separating funnel for partitioning between DCM and sat. aq. NaHCO3 solution. The organic phase was collected and the aqueous phase was back-extracted with DCM. The combined organic phases were dried over Na2SO4 and evaporated down to dryness. The crude material was purified by flash chromatography with a SiO2 column, eluting with a mixture of n-heptane and EtOAc (5% to 60%) to yield the desired compound as a colorless oil (102 mg). MS (ESI): m/z=322.3 [M+H]+.
  • BB33 (2R,4aR,8aS)-2-methyl-4a,5,6,7,8,8a-hexahydro-4H-pyrido[4,3-b][1,4]oxazin-3-one
  • To a solution of 6-benzyl-2-methyl-5,6,7,8-tetrahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Isomer A, 1.10 g, 4.26 mmol) in EtOAc (16 mL) and MeOH (16 mL) was added under argon Pd—C (227 mg, 213 μmol) and the suspension was stirred under a hydrogen atmosphere (balloon) at 1 bar for 24 h. The suspension was filtered over a microglass filter and washed with 20 mL EtOAc under inert gas. The filtrate was evaporated to give BB33 as a colorless solid (715 mg). MS (ESI): m/z=170.8 [M+H]+. Note: Only the single enantiomer formed during the reduction.
  • Step a) 2-Methyl-4H-pyrido[4,3-b][1,4]oxazin-3-one
  • To a solution of 3-aminopyridin-4-ol (2.5 g, 22.7 mmol) in DMF (100 mL) was added dropwise 2-chloropropanoyl chloride (3.03 g, 2.31 mL, 23.8 mmol) and the mixture was stirred at RT for 30 min. After addition of K2CO3 (7.84 g, 56.8 mmol), the suspension was heated to 100° C. (oil bath) for 20 h. The DMF was removed in vacuo, then 100 mL EtOAc were added and stirred at RT for 10 min, and it was washed with 50 mL H2O, extracted 3 times with EtOAc. The organic phases were combined, dried with MgSO4 and concentrated under vacuo to yield 3.72 g of 2-methyl-4H-pyrido[4,3-b][1,4]oxazin-3-one which was used in the next step without further purification.
  • Step b) 6-Benzyl-2-methyl-3-oxo-3,4-dihydro-2H-pyrido[4,3-b][1,4]oxazin-6-ium bromide
  • A suspension of 2-methyl-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (3.72 g, 22.7 mmol) in DCM (32 mL) and MeOH (8 mL) was treated with (bromomethyl)benzene (4.65 g, 3.23 mL, 27.2 mmol) and the mixture was stirred at RT for 60 h. A suspension formed, which was cooled down to 0° C., 20 mL n-hexane were added and then the precipitate was filtered. The residue was washed with 15 mL of cold DCM/n-hexan to yield the compound as an off-white solid (5.2 g). MS (ESI): m/z=255 [M+H]+.
  • Step c) 6-Benzyl-2-methyl-5,6,7,8-tetrahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • To a suspension of 6-benzyl-2-methyl-3-oxo-3,4-dihydro-2H-pyrido[4,3-b][1,4]oxazin-6-ium bromide (5.2 g, 15.5 mmol) in EtOH (38 mL) was added in portions NaBH4 (763 mg, 20.2 mmol) (exothermic, 22° C. to 30° C., yellow suspension). After the exothermic reaction faded out the mixture was stirred at room temperature for 3 h, then at 60° C. for 1 h and at 22° C. for 1 h. The reaction mixture was evaporated, partitioned between H2O and EtOAc and the layers were separated. The aqueous layer was extracted once with EtOAc. The organic layers were washed twice with H2O, dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 120 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (50 to 100 in 30 min.) to provide the compound as a light yellow solid (2.48 g) which could be used in the following step without further purification.
  • Step d) 6-Benzyl-2-methyl-5,6,7,8-tetrahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • The enantiomers were separated by preparative chiral HPLC (Chiralcel OD column) using an isocratic mixture of EtOH (containing 0.05% of NH4OAc):n-heptane (10:90). The fractions were evaporated to provide the desired compounds as light yellow solids (Isomer A 1.17 g, Isomer B 1.10 g).
  • BB34 N-(azetidin-3-ylmethyl)-2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethan-1-amine
  • In a 100 mL two-necked flask, benzyl 3-(((2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate (0.913 g, 2.05 mmol) was dissolved in a mixture of THF (5 mL) and MeOH (5 mL). Pd/C 10% (109 mg, 102 μmol) was added under argon. The flask was purged and backfilled with H2 gas (3 times). The reaction mixture was then stirred at 25° C. for 4 h. The suspension was filtered over decalite, concentrated and the resulting title compound (611 mg, colorless oil) used directly for the next step. MS (ESI): m/z=313.4 [M+H]+.
  • Step a) Benzyl 3-(((2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate
  • To a dry flask with septum was added benzyl 3-(aminomethyl)azetidine-1-carboxylate (0.5 g, 2.27 mmol), NEt3 (689 mg, 949 μL, 6.81 mmol), 2,2,2-trifluoro-1-(3-(trifluoromethyl)phenyl)ethan-1-one (554 mg, 391 μL, 2.27 mmol), and dry DCM (15 mL). Titanium tetrachloride 1M in DCM (1.13 mL, 1.13 mmol) was added via a syringe and the flask was cooled in an ice bath (exothermic). The reaction was stirred at RT overnight, carefully quenched with a solution of NaCNBH3 (428 mg, 6.81 mmol) in MeOH (4.36 g, 5.51 mL, 136 mmol) and acetic acid (0.1 mL) and stirred at RT overnight. The reaction was basified with sat. aq. NaHCO3 solution and the insoluble material obtained was filtered away over celite. The filtrate was extracted with DCM. The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in n-heptane) to yield the desired compound as a colorless oil (913 mg). MS (ESI): m/z=447.2 [M+H]+.
  • BB35 N-(azetidin-3-ylmethyl)-1-(2,4-dichlorophenyl)-2,2,2-trifluoroethan-1-amine
  • In a 100 mL two-necked flask, benzyl 3-(((1-(2,4-dichlorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate (0.660 g, 1.48 mmol) was dissolved in EtOAc (20 mL) to give a colorless solution. Pd/C 10% (78.5 mg, 73.8 μmol) was added under argon. The flask was purged and backfilled with H2 gas (3 times). The reaction mixture was stirred at 25° C. for 4 h. LC-MS showed a mixture of the title product N-(azetidin-3-ylmethyl)-1-(2,4-dichlorophenyl)-2,2,2-trifluoroethan-1-amine together with the dehalogenated side-products N-(azetidin-3-ylmethyl)-1-(2-chlorophenyl)-2,2,2-trifluoroethan-1-amine and N-(azetidin-3-ylmethyl)-1-phenyl-2,2,2-trifluoroethan-1-amine. The reaction mixture was filtered over decalite, concentrated in vacuo and used directly for the next step.
  • Step a) Benzyl 3-[[[1-(2,4-dichlorophenyl)-2,2,2-trifluoro-ethylidene]amino]methyl]azetidine-1-carboxylate
  • To a dry flask with septum was added under nitrogen benzyl 3-(aminomethyl)azetidine-1-carboxylate (0.500 g, 2.27 mmol, CAS RN 1016731-24-0), NEt3 (689 mg, 949 μL, 6.81 mmol), 1-(2,4-dichlorophenyl)-2,2,2-trifluoroethan-1-one (556 mg, 2.27 mmol, and dry DCM (16.4 mL). Titanium tetrachloride (1 M solution in DCM; 1.13 mL, 1.13 mmol) was added via a syringe to the ice-cooled flask (exothermic). The reaction was stirred at RT overnight, carefully quenched with a solution of NaCNBH3 (428 mg, 6.81 mmol) in MeOH (4.36 g, 5.51 mL, 136 mmol) and stirred for 6 h. LCMS indicated the reaction stopped at the imine.
  • The reaction was basified with sat. NaHCO3. The obtained insoluble material was filtered over celite and the filtrate was extracted with DCM. The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated. The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in n-heptane) to give the desired compound as a colorless oil (1 g).
  • Step b) Benzyl 3-(((1-(2,4-dichlorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate
  • In a 25 mL two-necked flask, benzyl 3-[[[1-(2,4-dichlorophenyl)-2,2,2-trifluoro-ethylidene]amino]methyl]azetidine-1-carboxylate (1 g, 2.25 mmol) was dissolved in THF (10 mL) and MeOH (1 mL) to give a colorless solution. Acetic acid (135 mg, 129 μL, 2.25 mmol) and NaCNBH3 (423 mg, 6.74 mmol) were added. The reaction mixture was stirred at 25° C. for 6 h. The reaction was basified with sat. NaHCO3. The obtained insoluble material was filtered over celite and the filtrate was extracted with DCM. The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated. The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in heptane) to afford the title compound as a colorless oil (660 mg) which used in the next step without further purification.
  • BB36 cis-4-((2-Chloro-4-fluorophenoxy)methyl)-3-methylpiperidine; hydrochloride salt
  • tert-Butyl cis-4-((2-chloro-4-fluorophenoxy)methyl)-3-methylpiperidine-1-carboxylate (115 mg, 321 μmol) was dissolved in DCM (2 mL) and 2M HCl in ether (161 μL, 321 μmol) was added. The reaction was stirred at RT for 6 h, then the solvent was removed in vacuo. The crude product (94 mg, colorless foam) was used in the next step without purification. MS (ESI): m/z=258.2 [M+H]+.
  • Step a) tert-Butyl cis-4-((2-chloro-4-fluorophenoxy)methyl)-3-methylpiperidine-1-carboxylate
  • Mitsunobu reaction: In a 50 mL four-necked sulphonation flask under argon, tert-butyl cis-4-(hydroxymethyl)-3-methylpiperidine-1-carboxylate (840 mg, 3.66 mmol) was dissolved in THF (15 mL), 2-chloro-4-fluorophenol (590 mg, 439 μL, 4.03 mmol) and triphenylphosphine (1.06 g, 4.03 mmol) were added. The clear solution was stirred 5 min at RT, then cooled to 0-2° C. and DEAD (702 mg, 638 μL, 4.03 mmol) was added over 10 min. The reaction mixture was stirred at 2-4° C. for 1 h, then stirred over night at RT. 50 mL diethylether were added, the mixture was washed with 2×25 mL water, 3×20 mL 1 N NaOH, 1×20 mL brine, the organic phase was dried with Mg2SO4, after removing the solvent in vacuo 2.7 g yellow oil were obtained. To remove the triphenylphosphinoxide, the residue was stirred in n-Heptane/diethylether for 30 min, the solids was filtered away, the filtrate was concentrated in vacuo, to obtain 1.8 g crude product that was purified by flash chromatography (silica gel, 50 g, 0% to 30% EtOAc in heptane, 40 min): tert-butyl cis-4-((2-chloro-4-fluorophenoxy)methyl)-3-methylpiperidine-1-carboxylate, 1.21 g white solid.
  • BB39 3-((2-Fluoro-4-(trifluoromethoxy)benzyl)oxy)azetidine; trifluoroacetate salt
  • To a solution of tert-butyl 3-((2-fluoro-4-(trifluoromethoxy)benzyl)oxy)azetidine-1-carboxylate (415 mg, 1.14 mmol) in DCM (5 mL) was added TFA (1.3 g, 875 μL, 11.4 mmol) and the reaction mixture was stirred at RT for 3 h. Volatiles were removed in vacuo to yield 455 mg of a light yellow oil that was used in the next step without further purification. MS (ESI): m/z=266.1 [M+H]+.
  • Step a) tert-Butyl 3-((2-fluoro-4-(trifluoromethoxy)benzyl)ox)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (200 mg, 1.15 mmol) in dry THF (5 mL) was added potassium tert-butoxide (1.65 M solution in THF, 735 μL, 1.21 mmol) and the reaction mixture was stirred at RT for 15 min followed by addition of l-(bromomethyl)-2-fluoro-4-(trifluoromethoxy)benzene (315 mg, 1.15 mmol). The reaction mixture was then stirred at room temperature for 14 h. The crude reaction was diluted with EtOAc and extracted with aq. 1 M NaHCO3 solution, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over NaSO4 and evaporated down to dryness to yield 418 mg of the crude product which was used in the next step without further purification. MS (ESI): m/z=310.1 [M−56+H]+.
  • BB40 N-(azetidin-3-yl)-2-chloro-4-fluoro-benzamide; trifluoroacetate salt
  • To a solution of tert-butyl 3-[(2-chloro-4-fluoro-benzoyl)amino]azetidine-1-carboxylate (346 mg, 1.05 mmol) in DCM (3.5 mL) was added TFA (0.7 mL) at 0° C. The solution was stirred at 0° C. for 2 h. The reaction was concentrated in vacuum to give the crude product (600 mg) as light yellow oil. The crude product was purified by prep-HPLC (0.1% TFA in H2O and MeCN) and dried by lyophilization to give desired compound as colorless solid (223 mg, 0.650 mmol, 59.2% yield). MS (ESI): m/z=229 [M+H]+.
  • Step a) tert-Butyl 3-[(2-chloro-4-fluoro-benzoyl)amino]azetidine-1-carboxylate
  • To a solution of 2-chloro-4-fluorobenzoic acid (500 mg, 2.86 mmol), 1-Boc-3-(amino)azetidine (493 mg, 2.86 mmol) and DMAP (35.0 mg, 0.290 mmol) in THF (10 mL) was added 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (714 mg, 3.72 mmol) at 0° C. The mixture was heated to 30° C. and stirred for 16 h. The reaction was diluted with EtOAc (5 mL), washed three times with brine (10 mL each) and dried over Na2SO4. The organic layer was concentrated in vacuum to give the crude product (0.72 g) as yellow oil. The crude product was purified by prep-HPLC and dried by lyophilization to give desired compound as a colorless solid (546 mg, 1.66 mmol, 57.9% yield). MS (ESI): m/z=273 [M−56+H]+.
  • BB41 N-(azetidin-3-ylmethyl)-2,2,2-trifluoro-N-methyl-1-[4-(trifluoromethyl)pentyl]ethanamine; trifluoroacetate salt
  • To a solution of tert-butyl 3-((methyl(2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate (0.256 g, 600 μmol) in DCM (5 mL) was added TFA (1.37 g, 925 μL, 12 mmol). The resulting reaction mixture was stirred at RT for 1 h. The reaction mixture was concentrated in vacuo to provide the desired compound as a colorless oil (268 mg). MS (ESI): m/z=327.4 [M+H]+.
  • Step a) tert-Butyl 3-((methyl(2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethyl)amino)methyl)azetidine-1-carboxylate
  • To a dry flask with septum and 3 A molecular sieves was added under nitrogen tert-butyl 3-((methylamino)methyl)azetidine-1-carboxylate (0.300 g, 293 μL, 1.5 mmol), TEA (455 mg, 626 μL, 4.49 mmol), 2,2,2-trifluoro-1-(4-(trifluoromethyl)phenyl)ethan-1-one (363 mg, 255 μL, 1.5 mmol), and dry DCM (9.86 mL). Titanium tetrachloride 1 M in DCM (749 μL, 749 μmol) was added via a syringe to the ice-cooled flask (exothermic). The reaction was stirred at RT overnight, carefully quenched with a solution of NaCNBH3 (282 mg, 4.49 mmol) in MeOH (3.64 mL, 89.9 mmol) and stirred at RT for 2 h. The reaction was basified with sat. NaHCO3 solution. The obtained insoluble material was filtered over celite and the filtrate was extracted with DCM. The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated. The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in n-heptane) and was used directly for the next step.
  • BB42 N-methyl-N-(piperidin-4-yl)-1-(3-(trifluoromethyl)phenyl)cyclopropane-1-carboxamide hydrochloride
  • To a solution of tert-butyl 4-(N-methyl-1-(3-(trifluoromethyl)phenyl)cyclopropane-1-carboxamido)piperidine-1-carboxylate (0.301 g, 706 μmol) in DCM (2 mL) was added HCl 2M in diethyl ether (3.53 mL, 7.06 mmol). The resulting reaction mixture was stirred at RT overnight and then concentrated under vacuum at 22° C. to give 256 mg of BB42 as off white solid, MS (ESI): m/z=327.2 [M+H]+
  • Step a) tert-butyl 4-(N-methyl-1-(3-(trifluoromethyl)phenyl)cyclopropane-1-carboxamido)piperidine-1-carboxylate
  • In a 20 mL glastube, to 1-(3-(trifluoromethyl)phenyl)cyclopropane-1-carboxylic acid (177 mg, 770 μmol) in DMF (5 mL) was added HATU (293 mg, 770 μmol) and DIPEA (271 mg, 367 μL, 2.1 mmol). The reaction mixture was stirred for 15 min and then tert-butyl 4-(methylamino)piperidine-1-carboxylate (0.15 g, 700 μmol) was added. The reaction mixture was stirred at RT for 2 hours. The reaction mixture was extracted with Water/DCM. The crude material was purified by flash chromatography (silica gel, 20 g, 0% to 100% EtOAc in heptane) to yield the desired compound as a light yellow oil (301 mg). MS (ESI): m/z=371.2 [M−56+H]+
  • BB43 2-(2-chloro-3-(trifluoromethyl)phenyl)-N-methyl-N-(piperidin-4-yl)acetamide; hydrochloride salt
  • To a solution of tert-butyl 4-(2-(2-chloro-3-(trifluoromethyl)phenyl)-N-methylacetamido)piperidine-1-carboxylate (0.301 g, 692 μmol) in DCM (2 mL) was added HCl (3.46 mL, 6.92 mmol). The resulting reaction mixture was stirred at RT for 2 days and then concentrated under vacuum at 22° C. to yield 252 mg of BB43 as off white solid. MS (ESI): m/z=335.1 [M+H]+.
  • Step a) tert-butyl 4-(2-(2-chloro-3-(trifluoromethyl)phenyl)-N-methylacetamido)piperidine-1-carboxylate
  • In a 20 mL glass tube, to 2-(2-chloro-3-(trifluoromethyl)phenyl)acetic acid (184 mg, 770 μmol) in DMF (5 mL) was added HATU (293 mg, 770 μmol), DIPEA (271 mg, 367 μL, 2.1 mmol). The reaction mixture was stirred for 15 min and then tert-butyl 4-(methylamino)piperidine-1-carboxylate (0.150 g, 700 μmol) was added. The reaction mixture was stirred at RT for 2 hours, and then extracted with Water/DCM. The crude material was purified by flash chromatography (silica gel, 20 g, 0% to 100% EtOAc in heptane) to yield tert-butyl 4-(2-(2-chloro-3-(trifluoromethyl)phenyl)-N-methylacetamido)piperidine-1-carboxylate as light yellow oil, 301 mg, MS (ESI): m/z=379.1 [M−56+H]+
  • BB44 2-(2-Chloro-5-(trifluoromethyl)phenyl)-N-methyl-N-(piperidin-4-yl)acetamide; hydrochloride salt
  • Synthesized from 2-(2-chloro-5-(trifluoromethyl)phenyl)acetic and tert-butyl 4-(methylamino)piperidine-1-carboxylate. See synthesis of BB43 for details. MS (ESI): m/z=335.1 [M+H]+.
  • BB46 3-Methyl-5-(piperidin-4-ylmethoxy)-2-(trifluoromethyl)pyridine; dihydrochloride salt
  • In a 25 mL tube tert-butyl 4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carboxylate (87 mg, 232 μmol) was dissolved in DCM (2 mL) and then HCl in ether 2M (697 μL, 1.39 mmol) was added, the reaction mixture was stirred 12 h at RT. The mixture was concentrated in vacuum, yielding 80 mg of BB46 as a white solid. MS (ESI): m/z=275.2 [M+H]+.
  • Step a) tert-Butyl 4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carboxylate
  • In a 5 mL tube, tert-butyl 4-(hydroxymethyl)piperidine-1-carboxylate (80.7 mg, 375 μmol) was dissolved in DMF (1.5 mL), then NaH in Oil 60% (18 mg, 450 μmol) was added at room temperature, the mixture was stirred for 20 min, then 5-bromo-3-methyl-2-(trifluoromethyl)pyridine (90 mg, 60 μL, 375 μmol) was added, and it was stirred for 2 hr at RT, yielding a brown solution. 10 mL sat. NH4C were added, it was extracted with water/ethyl acetate, dried over MgSO4, the solvent was removed at 40° C./150 mbar. The crude product was purified by flash chromatography (silica gel, 20 g, 0 to 40% EtOAc in n-heptane, in 35 min) to yield 87 mg of tert-butyl 4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carboxylate. MS (ESI): m/z=319.2 [M−56+H]+
  • BB58 N-(azetidin-3-ylmethyl)-2,2,2-trifluoro-1-(4-fluorophenyl)ethan-1-amine
  • In a 100 mL two-necked flask, benzyl 3-(((1-(2-chloro-4-fluorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate (707 mg, 1.64 mmol) was combined with THF (5 mL) and MeOH (5 mL) to give a colorless solution. Pd/C 10% (87.3 mg, 82.1 μmol) was added under argon. The flask was purged and backfilled with H2 (3 times). The reaction mixture was stirred at 25° C. for 1 h. The reaction mixture was filtered over decalite, concentrated and used directly for the next step. Colorless oil (472 mg). MS (ESI): m/z=263.2 [M+H]+ (the ortho-chlorine was lost during the hydrogenation).
  • Step a:) Benzyl 3-(((1-(2-chloro-4-fluorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate
  • To a dry flask under a stream of nitrogen was added benzyl 3-(aminomethyl)azetidine-1-carboxylate (0.5 g, 2.27 mmol), triethylamine (689 mg, 949 μL, 6.81 mmol), 1-(2-chloro-4-fluoro-phenyl)-2,2,2-trifluoro-ethanone (519 mg, 2.27 mmol), and dry DCM (15 mL). Titanium tetrachloride 1M in DCM (1.13 mL, 1.13 mmol) was added via a syringe to the ice-cooled flask (exothermic). The reaction was stirred overnight at RT, carefully quenched with a methanolic solution of sodium cyanoborohydride (428 mg, 6.81 mmol) in methanol (4.36 g, 5.51 mL, 136 mmol)+Acetic Acid (0.1 mL) and stirred overnight at RT. The reaction was basified with sat. NaHCO3. The insoluble material obtained was filtered away over celite, the filtrate was extracted with DCM, the organic layers were combined, washed with brine, dried over Na2SO4 and concentrated. Purification: The crude material was purified by flash chromatography (silica gel, 50 g, 0% to 50% EtOAc in heptane) to yield 707 mg of Benzyl 3-(((1-(2-chloro-4-fluorophenyl)-2,2,2-trifluoroethyl)amino)methyl)azetidine-1-carboxylate as a colorless oil. MS (ESI): m/z=431.2 [M+H]+.
  • BB59 2,2,2-Trifluoro-1-(piperidin-4-yl)-N-(3-(trifluoromethyl)benzyl)ethan-1-amine; hydrochloride salt
  • To a solution of tert-butyl 4-(2,2,2-trifluoro-1-((3-(trifluoromethyl)benzyl)amino)ethyl)piperidine-1-carboxylate (0.140 g, 318 μmol) in DCM (2 mL) was added HCl 2M in diethyl ether (1.59 mL, 3.18 mmol). The resulting reaction mixture was stirred at RT overnight and then concentrated under vacuum at 22° C. to yield 119 mg of the title compound as off-white solid. MS (ESI): m/z=340.8 [M+H]+.
  • Step a) tert-Butyl 4-(2,2,2-trifluoro-1-((3-(trifluoromethyl)benzyl)amino)ethyl)piperidine-1-carboxylate
  • A solution of tert-butyl 4-(1-amino-2,2,2-trifluoroethyl)piperidine-1-carboxylate (0.150 g, 531 μmol) and 3-(trifluoromethyl)benzaldehyde (92.5 mg, 71.1 μL, 531 μmol) in 1,2-DCE (1 mL) was stirred for 1 hour at RT. Sodium triacetoxyborohydride (225 mg, 1.06 mmol) was then added at 0° C., and the reaction mixture was stirred at RT overnight. The reaction mixture was poured onto sat. NaHCO3 and extracted with DCM. The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude material was purified by flash chromatography (silica gel, 20 g, 0% to 100% EtOAc in heptane) to yield 145 mg of the desired compound as a colorless oil. MS (ESI): m/z=383.1 [M−56+H]+
  • BB69 2-methyl-3-((4-(trifluoromethyl)benzyl)oxy)azetidine; trifluoroacetate salt
  • To a solution of tert-butyl 2-methyl-3-((4-(trifluoromethyl)benzyl)oxy)azetidine-1-carboxylate (0.36 g, 1.04 mmol) in DCM (4 mL) was added trifluoroacetic acid (1.19 g, 10.4 mmol). The resulting reaction mixture was stirred at RT for 1 hour. The reaction mixture was concentrated on high vacuum to yield BB69 as a light yellow oil, 399 mg, mixture of all four stereoisomers. MS (ESI): m/z=246.1 [M+H]+.
  • Step a) tert-Butyl 2-methyl-3-((4-(trifluoromethyl)benzyl)oxy)azetidine-1-carboxylate
  • In a 25 mL two-necked flask, tert-butyl-3-hydroxy-2-methylazetidine-1-carboxylate (215 mg, 1.15 mmol) was dissolved in DMF (5 mL) to give a colorless solution. At 0° C., sodium hydride (60% dispersion in mineral oil) (41.8 mg, 1.05 mmol) was added. The reaction mixture was stirred at 0° C. for 15 min. Then 1-(bromomethyl)-4-(trifluoromethyl)benzene (0.250 g, 1.05 mmol) was added at 0° C. The reaction mixture was stirred at RT overnight. The reaction mixture was poured onto 20 mL sat. NH4Cl and extracted with EtOAc (2×50 mL). The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude material was purified by flash chromatography (silica gel, 20 g, 0% to 70% EtOAc in heptane) to yield 360 mg of tert-butyl 2-methyl-3-((4-(trifluoromethyl)benzyl)oxy)azetidine-1-carboxylate as a colorless oil. MS (ESI): m/z=290.1 [M−56+H]+
  • BB87 3-Fluoro-5-(trifluoromethyl)benzyl 4-methylbenzenesulfonate
  • To a solution of (3-fluoro-5-(trifluoromethyl)phenyl)methanol (100 mg, 72.5 μL, 515 μmol, Eq: 1) in DCM (2.58 mL) was added p-toluenesulfonic anhydride (185 mg, 567 μmol), DIPEA (79.9 mg, 108 μL, 618 μmol) and DMAP (6.29 mg, 51.5 μmol). The reaction mixture was stirred for 4 h at 0° C. and for 2 days at room temperature. The reaction mixture was taken up in EtOAc and washed with water and brine. The organic layers were dried over MgSO4 and concentrated in vacuo to give a yellow oil (178 mg) which was used without further purification.
  • In analogy to BB39, and if not specified otherwise, the intermediates shown in the following table were prepared from commercially available benzyl bromides or the prepared tosylate intermediates and the corresponding tert-butyl 3-hydroxyazetidine-1-carboxylate building blocks.
  • BB MS,
    No. Systematic Name Starting material m/z
    BB37 3-((2-Chloro-4- 1-(Bromomethyl)-2-chloro-4- 266.1
    (trifluoroinethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB38 3-((4- 1-(Bromomethyl)-4- 232.1
    (Trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB45 3((3-Methoxy-4- 4-(Bromomethyl)-2-methoxy-1- 262.2
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB56 3-((3-Fluoro-5- 3-Fluoro-5- 250.1
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzyl 4- [M + H]+
    trifluoroacetate salt methylbenzenesulfonate (BB87)
    BB60 3-((3-Chloro-4- 4-(Bromomethyl)-2-chloro-1- 266.1
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB62 3-((2-Fluoro-4- 1-(Bromomethyl)-2-fluoro-4- 318.3
    (trifluoromethyl)benzyl)oxy)-3- (trifluoromethyl)benzene and [M + H]+
    (trifluoromethyl)azetidine; Tert-butyl 3-hydroxy-3-
    trifluoroacetate salt trifluoromethyl)azetidine-1-
    carboxylate (CAS: 398489-42-4)
    BB63 3-((2-Fluoro-4- 1-(Bromornethyl)-2-fluoro-4- 264.1
    (trifluoromethyl)benzyl)oxy)-3- (trifluoromethyl)benzene and [M + H]+
    methylazetidine; trifluoroacetate salt Tert-butyl 3-hydroxy-3-
    methylazetidine-1-carboxylate
    (CAS: 1104083-23-9)
    BB64 3-((2,4-Difluoro-5- 1-(Bromornethyl)-2,4-difluoro-5- 268.1
    (trifluoromethypbenzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB65 3-((2-Fluoro-5- 2-(Bromomethyl)-1-fluoro-4- 250.1
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB66 3-((2-Fluoro-5- 4-(Bromomethyl)-2-fluoro-1- 250.1
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB67 3-((2-Methoxy-4- 1-(Bromomethyl)-2-methoxy-4- 262.2
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB68 3-((4-Chloro-2- 1-(Bromomethyl)-4-chloro-2- 266.2
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene [M + H]+
    trifluoroacetate salt
    BB88 3[(2,4- 1-(Bromomethyl)-2,4-dichloro- 232.1
    dichlorophenyl)methoxy]azetidine benzene [M + H]+
     BB170 3((3,4- 4-(Bromomethyl)-1,2,dichloro- 232.1
    Dichlorobenzyl)oxy)azetidine; 2,2,2- benzene [M + H]+
    trifluoroacetate
     BB171 3((2,5- 2-(Bromomethyl)-1,4-dichloro- 232.1
    Dichlorobenzyl)oxy)azetidine; 2,2,2- benzene [M + H]+
    trifluoroacetate
     BB172 3-((3- 3-(Bromomethyl)- 248.1
    (Trifluoromethoxy)benzyl)oxy)azetidine; trifluoromethoxy-benzene [M + H]+
    2,2,2-trifluoroacetate
     BB173 2-Methyl-3-((4-methyl-3- tert-Butyl-3-hydroxy -2- 266.2
    (trifluoromethyl)benzyl)oxy)azetidine; methylazetidine-1-carboxylate [M + H]+
    2,2,2-trifluoroacetate and
    4-(bromomethyl)-1-methyl-2-
    trifluoromethyl)benzene
     BB178 3-(((2-Fluoro-4- tert-Butyl 3- 264.2
    (trifluoromethyl) benzyl)oxy)methyl) (hydroxymethyl)azetidine- 1- [M + H]+
    azetidine; 2,2,2-trifluoroacetate carboxylate and
    1-(Bromomethyl)-2-fluoro-4-
    (trifluoromethyl)benzene
     BB180 [4-(Azetidin-3-yloxymethyl)-3- (4-(Bromomethyl)-3- 308.2
    fluoro-phenyl]-pentafluoro-□6- fluorophenyl)pentafluoro-□6- [M + H]+
    sulfane; 2,2,2-trifluoroacetate sulfane
     BB185 3-((2-Fluoro-4- tert-Butyl 3-hydroxy-3- 332.2
    (trifluoromethyl)benzyl)oxy)-3- (trifluoromethyl)pyrrolidine-1- [M + H]+
    (trifluoromethyl)pyrrolidine; 2,2,2- carboxylate and
    trifluoroacetate 1-(Bromomethyl)-2-fluoro-4-
    (trifluoromethyl)benzene
     BB187 3-[[2,4- 1-(Bromomethyl)-2,4- 336.2
    bis(Trifluoromethyl)phenyl]methoxy] bis(trifluoromethy)benzene [M + H]+
    azetidine
     BB188 3-[[2-Methyl-3- 1-(Bromomethyl)-2-methyl-3- 246.1
    (trifluoromethyl)phenyl]methoxy] (trifluoromethyl)benzene [M + H]+
    azetidine
     BB189 3[[2-Methyl-4- 1-(Bromornethyl)-2-methyl-4- 262.1
    (trifluoromethoxy)phenyl]methoxy] (trifluoromethoxy)benzene [M + H]+
    azetidine
     BB190 2-Methyl-3-[[2-methyl-4- 1-(Bromomethyl)-2-methyl-4- 276.2
    (trifluoromethoxy)phenyl]methoxy] (trifluoromethoxy)benzene [M + H]+
    azetidine and
    tert-butyl 3-hydroxy-2-
    methylazetidine-1-carboxylate
     BB191 2-Methyl-3-[[2-methyl-3- 1-(Bromomethyl)-2-methyl-3 260.2
    (trifluoromethyl)phenyl]methoxy] (trifluoromethyl)benzene [M + H]+
    azetidine and
    tert-Butyl 3-hydroxy-2-
    methylazetidine-1-carboxylate
     BB207 3-[[4-Fluoro-2- 1-(Chloromethyl)-4-fluoro-2- 250.7
    (trifluoromethyl)phenyl]methoxy] trifluoromethyl)benzene (CAS [M + H]+
    azetidine; 4-methylbenzenesulfonate RN 248262-29-5)
     BB212 3-[[3-Fluoro-4- 4-(Bromornethyl)-2-fluoro-1- Used
    ('trifluoromethoxy)phenyl]methoxy] (trifluoromethoxy)benzene without
    azetidine; 2,2,2-trifluoroacetate purification
     BB217 3-((4-Methyl-3- 4-(Bromomethyl)-1-methyl-2 246.2
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene. [M + H]+
    2,2,2-trifluoroacetate tBuOK as base
  • In analogy to BB29, intermediates BB20, BB25 and BB61 of the following table were prepared from the commercially available phen9ols. Where trifluoroacetate salts are indicated, the crude product resulting from concentration of the reaction mixture was used directly without further neutralization or purification.
  • BB MS,
    No. Systematic Name Starting material m/z
    BB20 3-((2-Fluoro-4- 2-Fluoro-4- 250.1
    (trifluoromethyl)phenoxy)methyl)azetidine; (trifluoromethyl)phenol (CAS [M + H]+
    trifluoroacetate salt RN: 77227-78-2)
    BB25 3-[(2-Chloro-4- 2-Chloro-4-fluorophenol (CAS 216.1
    fluorophenoxy)methyl]azetidine RN: 1996-41-4) [M + H]+
    (purified
    by RP-
    HPLC)
    BB61 3-((2-Chloro-4- 2-Chloro-4-fluorophenol (CAS 234.1
    fluorophenoxy)methyl)-3- RN: 1996-41-4) [M + H]+
    fluoroazetidine; trifluoroacetate salt and teft-Butyl 3-fluoro-3-
    (hydroxymethyl)azetidine-1-
    carboxylate (CAS: 1126650-66-5)
  • In analogy to BB26, intermediates BB21-BB24 and BB28 of the following table were prepared from the commercially available phenols.
  • BB No. Systematic Name Starting material MS, m/z
    BB21  4-((4-Fluoro-2- 4-Fluoro-2- 278.1
    (trifluoromethyl)phenoxy)methyl)piperidine; (trifluoromethyl)phenol (CAS: [M + H]+
    hydrochloride salt 130047-19-7
    BB22  4[[2-Fluoro-4- 2-Fluoro-4- 278.0
    (trifluoromethyl)phenoxy]methyl]piperidine; (trifluoromethol)phenol (CAS: [M + H]+
    hydrochloride salt 77227-78-2)
    BB23  4-((2-Chloro-4- 2-Chloro-4- 294.1
    (trifluoromethyl)phenoxy)methyl)piperidine; (trilluoromethyl)phenol (CAS: [M + H]+
    hydrochloride salt 35852-58-5)
    BB24  5-Fluoro-2-(piperidin-4- 5-Fluoro-2-hydroxybenzonitrile 235.1
    ylmethoxy)benzonitrile; (CAS: 91407-41-9) [M + H]+
    hydrochloride salt
    BB28  4-[(4-Fluoro-2-methyl- 4-Fluoro-2-methylphenol (CAS: 224.0
    phenoxy)methyl]piperidine; 452-72-2) [M + H]+
    hydrochloride salt
    BB170 3((3,4- 4(Bromomethyl)-1,2-dichloro- 232.1
    Dichlorobenzyl)oxy)azetidine benzene [M + H]+
    2,2,2-trifluoroacetate
    BB171 3-((2,5- 2(Bromomethyl)-1,4-dichloro- 232.1
    Dichlorobenzyl)oxy)azetidine benzene [M + H]+
    2,2,2-trifluoroacetate
    BB172 3((3- 3(Bromomethyl)- 248.1
    (Trifluoromethoxy)benzyl)oxy)azetidine trifluoromethoxy-benzene [M + H]+
    2,2,2-trifluoroacetate
    BB173 2-Methyl-3((4-methyl-3- tert-Butyl-3-hydroxy-2- 266.2
    (triftuoromethyl)benzyl)oxy)azetidine methylazetidine-l-carboxylate [M + H]+
    2,2,2-trifluoroacetate and
    4(Bromomethyl)-1-methyl-2 -
    (trifluoromethyl)benzene
    BB178 3-(((2-Fluoro-4- tert-Butyl 3- 264.2
    (trifluoromethyl)benzyl)oxy)methyl)azetidine (hydroxymethyl)azetidine-1- [M + H]+
    2,2,2-trifluoroacetate carboxylate and
    1(Bromomethyl)-2-fluoro-4-
    (trifluoromethyl)benzene
    BB180 [4(Azetidin-3-yloxylmethyl)-3- (4-Bromomethyl)-3- 308.2
    fluoro-phenyl]-pentafluoro-□6- fluorophenyl)pentafluoro-□6- [M + H]+
    sulfane 2,2,2-trifluoroacetic acid sulfane
    BB185 3((2-Fluoro-4- tert-Butyl 3-hydroxy-3- 332.2
    (trifluoromethyl)benzyl)oxy)-3- (trifluoromethyl)pyrrolidine-1- [M + H]+
    (trifluoromethyl)pyrrolidine 2,2,2- carboxylate and
    trifluoroacetate 1(Bromomethyl)-2-fluoro-4-
    (trifluoromethyl)benzene
    BB187 3-[[2,4- 1-(Bromomethyl)-2,4- 336.2
    bis(Trifluoromethyl)phenyl]methoxy]azetidine bis(trifluoromethyl)benzene [M + H]+
    BB188 3-[[2-Methyl-3- 1-(Bromomethyl)-2-methyl-3 - 246.1
    (trifluoromethyl)phenyl]methoxy]azetidine (trifluoromethyl)benzene [M + H]+
    BB189 3[[2-Methyl-4- 1-(Bromomethyl)-2-methyl-4- 262.1
    (trifluoromethoxy)phenyl]methoxy]azetidine (trifluoromethyl)benzene [M + H]+
    BB190 2-Methyl-3-[[2-methyl-4- 1-(Bromomethyl)-2-methyl-4- 276.2
    (trifluoromethoxy)phenyl]methoxy]azetidine (trifluoromethyl)benzene [M + H]+
    and tert-butyl 3-hydroxy-2-
    methylazetidine-1-carboxylate
    BB191 2-Methyl-3-[[2-methyl-3- 1-(Bromomethyl)-2-methyl-3- 260.2
    (trifluoromethoxy)phenyl]methoxy]azetidine (trifluoromethyl)benzene [M + H]+
    and tert-Butyl 3-hydroxy-2-
    methylazetidine-1-carboxylate
    BB207 3-[[4-Fluoro-2- 1-(Chloromethyl)-4-fluoro-2- 250.2
    (trifluoromethoxy)phenyl]methoxy]azetidine; (trifluoromethyl)benzene (CAS [M + H]+
    4-methylbenzenesulfonate RN 248262-29-5)
    BB212 3-[[3-Fluoro-4- 4-(Bromomethyl)-2-fluoro-1- Used
    (trifluoromethoxy)phenyl]methoxy]azetidine (trifluoromethoxy)benzene without
    2,2,2-trifluoroacetate purification
    BB217 3-((-Methyl-3- 4-(Bromomethyl)-1-methyl-2- 246.2
    (trifluoromethyl)benzyl)oxy)azetidine; (trifluoromethyl)benzene. [M + H]+
    2,2,2-trifluoroacetate tBuOK as base
    BB218 3-((2-Fluoro-6- tert-Butyl 3-mercaptoazetidine- 266.2
    (trifluoromethyl)benzyl)thio)azetidine 1-carboxylate and [M + H]+
    2,2,2-trifluoroacetate 2-(Bromomethyl)-1-fluoro-3-
    (trifluoromethyl)benzene
  • Method D1 BB9 4-[2-Chloro-4-(trifluoromethyl)phenoxy]piperidine; trifluoroacetate salt
  • A mixture of tert-butyl 4-[2-chloro-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (750.0 mg, 1.97 mmol) in DCM (20 mL) and TFA (0.76 mL) was stirred at 20° C. for 12 h. The mixture was concentrated. The residue was dissolved in H2O (20 mL) and washed twice with PE:EA=10:1 (20 mL each). The aqueous layer was lyophilized to give the desired product as light yellow solid (716 mg, 1.82 mmol, 87.8%). MS (ESI): m/z=280.1 [M+H]+.
  • Step a) tert-Butyl 4-[2-chloro-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • A mixture of 2-chloro-4-(trifluoromethyl)phenol (500 mg, 2.54 mmol), 1-Boc-4-hydroxypiperidine (768 mg, 3.82 mmol) and triphenylphosphine (1334 mg, 5.09 mmol) in THF (10 mL) was stirred at 0° C. until completely dissolved. DIAD (1542 mg, 7.63 mmol) was slowly added dropwise at 0° C. The mixture was stirred at 20° C. for 3 h and then concentrated under vacuum. The residue was purified by prep-HPLC to give the desired compound as light yellow solid (760 mg, 2 mmol, 78.7% yield). MS (ESI): m/z=324.0 [M-56+H]+.
  • BB57 3-(((2-Fluoro-6-(trifluoromethyl)benzyl)oxy)methyl)azetidine; trifluoroacetate salt
  • To a solution of tert-butyl 3-(((2-fluoro-6-(trifluoromethyl)benzyl)oxy)methyl)azetidine-1-carboxylate (158 mg, 435 μmol) in DCM (1.74 mL) was added TFA (793 mg, 536 μL, 6.96 mmol) and the reaction was stirred at room temperature for 3 h. The reaction mixture was concentrated to give 3-(((2-fluoro-6-(trifluoromethyl)benzyl)oxy)methyl)azetidine; trifluoroacetate salt (202 mg, 434 μmol, 99.7% yield) as a colorless oil. The crude was used without further purification. MS (ESI): m/z=264.1 [M+H]+.
  • Step a) Tert-butyl 3-(((2-fluoro-4-(trifluoromethyl)benzyl)oxy)methyl)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate (100 mg, 534 μmol) in dry THF (2.67 mL) was added potassium tert-butoxide 1.65 M solution in THF (340 μL, 561 μmol) and the turbid reaction mixture was stirred at RT for 15 min followed by addition of 1-(bromomethyl)-2-fluoro-6-(trifluoro-6-(trifluoromethyl)benzene (137 mg, 534 μmol). The reaction mixture was then stirred at room temperature for 3 h. The crude reaction was diluted with ethyl acetate and extracted with sat. aq. NaHCO3 solution, the organic phase was collected and the aqueous phase was back-extracted with ethyl acetate. The combined organic phases were dried over sodium sulfate and evaporated down to dryness to yield a clear oil. The crude was immobilized on Isolute and purified by column chromatography eluting with 0 to 30% EtOAc in heptanes to afford tert-butyl 3-(((2-fluoro-6-(trifluoromethyl)benzyl)oxy)methyl)azetidine-1-carboxylate (158 mg, 413 μmol, 77.3% yield) as a colorless oil. MS (ESI): m/z=308.1 [M−56+H]+
  • Method D2 BB10 4-[[2-Cyclopentyl-4-(trifluoromethyl)phenyl]methyl]piperidine; formic acid salt
  • A mixture of tert-butyl 4-[[2-cyclopentyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate (440 mg, 0.610 mmol) and 5.0 mL of 4 M HCl in EtOAc in EtOAc (10 mL) was stirred at 20° C. for 12 h. The mixture was concentrated under vacuum. The residue was re-dissolved in H2O (5 mL), washed twice with PE:EA (3:1; 10 mL each) and the layers were separated. The aqueous layer was purified by prep-HPLC to give the desired compound as light yellow solid (124 mg, 0.350 mmol, 65.3% yield). MS (ESI): m/z=312.2 [M+H]+.
  • Step a) tert-Butyl 4-[[2-cyclopentyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • A solution of tert-butyl 4-[[2-bromo-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (500 mg, 1.19 mmol), cyclopentyl bromide (266 mg, 1.78 mmol), Ir(dF(CF3)ppy)2(dtbbpy)PF6 (13.4 mg, 0.010 mmol, CAS RN 870987-63-6), NiCl2.glyme (0.77 mg, 0.060 mmol), dtbbpy (19.2 mg, 0.070 mmol, CAS RN 72914-19-3), TTMSS (296 mg, 1.19 mmol, CAS RN 1873-77-4) and Na2CO3 (252 mg, 2.38 mmol) in DMF (20 mL) was degassed by bubbling argon stream for 20 min. The reaction mixture was irradiated with Blue LED (4×1) at 25° C. for 16 h. The mixture was diluted with H2O and then extracted three times with EtOAc (100 mL each). The combined organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by prep-HPLC to give the compound as a colorless oil (460 mg, 1.12 mmol, 53.8%). MS (ESI): m/z=354.1 [M−56+H]+.
  • Step b) tert-Butyl 4-[[2-cyclopentyl-4-(rifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • To a mixture of tert-butyl 4-[[2-cyclopentyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (460 mg, 0.640 mmol) in EtOAc (10 mL) was added wet Pd/C (40 mg), and then the mixture was stirred at 20° C. for 12 h under H2 (1520 mmHg). The mixture was filtered and the filtrate was concentrated to give the compound as colorless oil (460 mg, 1.12 mmol, 99.5%). MS (ESI): m/z=356.1 [M+H−56]+.
  • BB11 2-(4-Piperidylmethyl)-1,3-benzoxazole; formic acid salt
  • A solution of 2-aminophenol (1.0 g, 9.16 mmol) and 1-Boc-4-piperidylacetic acid (2.68 g, 11 mmol) in polyphosphoric acid (2.2 g) was stirred at 180° C. for 2 h. The mixture was diluted with a mixture of 12M aqueous NH4OH solution and ice to reach pH>7, and then extracted three times with EtOAc (10 mL each). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated, and the residue was purified by prep-HPLC to give the desired compound as a brown oil (251 mg, 0.960 mmol, 9.7%). MS (ESI): m/z=217.2 [M+H]+.
  • Method D3 BB13 4-[4-Chloro-3-(4-chlorophenyl)phenoxy]piperidine; hydrochloride salt
  • A solution of tert-butyl 4-[4-chloro-3-(4-chlorophenyl)phenoxy]piperidine-1-carboxylate (1000 mg, 2.37 mmol) in a 4 M solution of HCl in dioxane (50 mL) was stirred at 20° C. for 12 h. The mixture was concentrated to give the title compound as a white solid (845 mg, 2.35 mmol, 96.2%). MS (ESI): m!z=322.0 [M+H]+.
  • Step a) tert-Butyl 4-(3-bromo-4-chloro-phenoxy) piperidine-1-carboxylate
  • A mixture of 3-bromo-4-chlorophenol (1000 mg, 4.82 mmol), 1-Boc-4-hydroxypiperidine (1164 mg, 5.78 mmol) and triphenylphosphine (2529 mg, 9.64 mmol) was stirred in THF (10 mL) until completely dissolved. Then DIAD (1948 mg, 9.64 mmol) was slowly added drop wise at 0° C.
  • The mixture was stirred at 20° C. for 12 h, concentrated and the residue was purified by reversed flash chromatography to give the compound as yellow oil (1300 mg, 3.33 mmol, 69.0%). MS (ESI): m/z=336.0 [M−56+H]+.
  • Step b) tert-Butyl 4-[4-chloro-3-(4-chlorophenyl)phenoxy]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate (1150 mg, 2.94 mmol) and 4-chlorophenylboronic acid (506 mg, 3.24 mmol), Na2CO3 (1248 mg, 11.8 mmol) in 1,4-dioxane (20 mL) and H2O (5 mL) was added tetrakis(triphenylphosphine)palladium(0) (340 mg, 0.290 mmol, CAS RN 14221-01-3), and the mixture was stirred at 110° C. under N2 atmosphere for 12 h. The mixture was filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography, eluting with a 5-20% EtOAc-PE gradient to give the desired compound as light yellow oil (1100 mg, 2.6 mmol, 88.5%). MS (ESI): m/z=366.1 [M−56+H]+.
  • BB14 4-[[2-(1H-Pyrazol-4-yl)-4-(trifluoromethyl)phenyl]methyl)phenylmethyl]piperidine; trifluoroacetate salt
  • To a mixture of tert-butyl 4-[[2-(1-tert-butoxycarbonylpyrazol-4-yl)-4-(trifluoromethyl) phenyl]methyl]piperidine-1-carboxylate (150.0 mg, 0.290 mmol) in DCM (5 mL) was added TFA (1.0 mL). The mixture was stirred at 20° C. for 15 h. The mixture was concentrated under vacuum and then lyophilized to give the title compound as light yellow gum (149 mg, 0.280 mmol, 85.1% yield). MS (ESI): m/z=310.0 [M+H]+.
  • Step a) tert-Butyl 4-[[2-(1-tert-butoxycarbonylpyrazol-4-yl)-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[[2-bromo-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (600 mg, 1.43 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3-dioxolan-2-yl)pyrazole-1-carboxylate (846 mg, 2.86 mmol) and K2CO3 (592 mg, 4.28 mmol) in DMF (10 mL) and H2O (0.5 mL) was stirred at 80° C. for 12 h. The mixture was poured into H2O (30 mL) and extracted twice with EtOAc (50 mL each). The combined organic layers were washed with brine (30 mL), dried over Na2SO4 and filtered. The filtrated was concentrated in vacuum to give the compound as light yellow oil (520 mg, 1.02 mmol, 71.8% yield). MS (ESI): m/z=308.1 [M+H]+.
  • Step b) tert-Butyl 4-[[2-(1-tert-butoxycarbonylpyrazol-4-yl)-4-(trifluoromethyl) phenyl]methyl]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[[2-(1-tert-butoxycarbonylpyrazol-4-yl)-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (180 mg, 0.350 mmol) and wet Pd/C (18 mg) in EtOAc (10 mL) was stirred at 30° C. for 24 h under H2 atmosphere (˜1520 mm Hg). The mixture was filtered and concentrated under vacuum to give the compound as brown oil (150 mg, 0.290 mmol, 83%). MS (ESI): m/z=354.1 [M−56-100+H]+.
  • BB18 4-[2-(2-Chlorophenyl)ethynyl]piperidine
  • To a suspension of tert-butyl 4-((2-chlorophenyl)ethynyl)piperidine-1-carboxylate (0.05 g, 0.156 mmol) in MeOH (3 mL) was added 4 M HCl in dioxane (0.391 mL, 1.56 mmol) and the reaction mixture was stirred at room temperature for 2 h. The mixture was evaporated to dryness and the residue triturated in diisopropyl ether, filtered off and further dried under high vacuum to give the title compound as a white solid as the hydrochloride salt (0.02 g, 50%). MS (ESI): m/z=220.1 [M+H]+.
  • Step a) tert-Butyl 4-[2-(2-chlorophenyl)ethyl]piperidine-1-carboxylate
  • In a sealed tube, a mixture of tert-butyl 4-ethynylpiperidine-1-carboxylate (0.1 g, 0.478 mmol, CAS RN 287192-97-6), 1-bromo-2-chlorobenzene (0.084 mL, 0.717 mmol), copper (I) iodide (0.002 g, 0.009 mmol), TEA (0.666 mL, 4.78 mmol) and bis(triphenylphosphine)palladium(II) chloride (0.027 g, 0.038) in THF (2.8 mL) was degassed for 5 min under Argon. The reaction mixture was then heated to 70° C. and stirred for 4 h. The mixture was filtered off over a pad of Dicalite, washed with EtOAc and the mother liquors were evaporated to dryness. The residue was purified by silica gel flash chromatography, eluting with a gradient of 0-50% EtOAc/n-heptane to give the title compound as a white solid (0.05 g, 33%). MS (ESI): m/z=264.1 [M−56+H]+.
  • BB48a tert-Butyl 4-[[2-fluoro-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • A degassed solution of tert-butyl 4-methylenepiperidine-1-carboxylate (4465 mg, 22.6 mmol, CAS RN 159635-49-1) in 9-BBN (45.3 mL, 22.6 mmol) was refluxed for 1 h. After cooling to room temperature, the solution was added into a solution of 4-bromo-3-fluorobenzotrifluoride (5.0 g, 20.6 mmol, CAS RN 40161-54-4), Pd(dppf)Cl2 (1514 mg, 2.06 mmol) and K2CO3 (5687 mg, 41.1 mmol) in DMF (50 mL) and water (5 mL). The resulting mixture was heated at 80° C. for 5 h. After the mixture was cooled to room temperature and poured into water, the pH was adjusted to 11 with 10% aqueous NaOH solution, and the mixture was extracted with EtOAc. The combined organic extracts were dried with brine and Na2SO4, filtered, and evaporated to give a residue, which was further purified by column chromatography (silica gel, PE:EtOAc=10:1 to 5:1) to give the compound as light yellow solid (240 mg, 3.2%). MS (ESI): m/z=306 [M+H−56]+.
  • BB51a
  • A mixture of tert-butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (1000 mg, 2.62 mmol) and PtO2 (100 mg, 0.440 mmol) in EtOAc (20 mL) was stirred at 20° C. for 12 h under H2 atmosphere (1520 mmHg). The mixture was filtered and the filtrate concentrated to furnish the compound as light yellow solid (940 mg, 93.5%). MS (ESI): m/z=328.2 [M+H]+.
  • Step a) 2-Bromo-1-(bromomethyl)-4-(rifluoromethyl)benzene
  • A mixture of 2-bromo-1-methyl-4-(trifluoromethyl)benzene (5.5 g, 23.0 mmol, CAS RN 128-08-5), benzoyl peroxide (835 mg, 3.45 mmol) and NBS (4.07 g, 23.01 mmol) in CCl4 (50.0 mL, 23.0 mmol) was stirred at 70° C. for 5 h. The mixture was poured into water (20 mL) and extracted twice with DCM (20 mL each). The combined organic layer was washed with brine (20 mL), dried over Na2SO4, filtered and concentrated in vacuum to give the desired compound as light yellow oil which was used in the next step without further purification (7.1 g, 97%).
  • Step b) 2-Bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene
  • A mixture of 2-bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene (7.1 g, 22.3 mmol) and triethyl phosphite (30 mL) was stirred at 155° C. for 5 h. The mixture was concentrated in vacuum to remove triethyl phosphite, the residue was diluted with water (100 mL) and extracted three times with EtOAc (100 mL each). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography (PE:EtOAc=100:1 to 10:1) to give the compound as light yellow oil which was used without further purification in the next step (8 g, 95.5%).
  • Step c) tert-Butyl 4-(2-bromo-4-(trifluoromethyl)benzylidene)piperidine-1-carboxylate
  • To a mixture of 2-bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene (6.9 g, 18.4 mmol) in THF (100 mL) was added sodium hydride (2.21 g, 55.2 mmol) at 0° C. The mixture was stirred at 0° C. for 1 h, then 1-Boc-4-piperidone (7.33 g, 36.79 mmol, CAS RN 79099-07-3) was added and the mixture was stirred at 20° C. for 12 h. The mixture was poured into water (100 mL) and extracted three times with EtOAc (100 mL each). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (PE:EA=100:1 to 50:1) to yield the desired compound as off-white solid (4 g, 51.7%). MS (ESI): m/z=365.9 [M-56+H].
  • Step d) tert-butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[[2-bromo-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (2.0 g, 4.76 mmol), cyclopropylboronic acid (818 mg, 9.52 mmol, CAS RN 411235-57-9) and potassium carbonate (1973 mg, 14.3 mmol) in DMF (10 mL) and water (0.5 mL) was stirred at 80° C. under nitrogen atmosphere for 12 h. The mixture was poured into water (50 mL), extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine (50 mL), dried over Na2SO4 and concentrated in vacuum. The residue was purified by prep-HPLC to give the compound as light yellow oil (1020 mg, 56.2% yield). MS (ESI): m/z=326.0 [M−56+H]+.
  • BB53a tert-Butyl 3-[(4-chlorophenyl)methoxy]pyrrolidine-1-carboxylate
  • A solution of N-Boc-3-hydroxypyrrolidine (1.0 g, 5.34 mmol) and 4-chlorobenzyl bromide (1.32 g, 6.41 mmol) in ACN (10 mL) was added potassium carbonate (1.48 g, 10.68 mmol). The mixture was stirred at 80° C. for 15 h. Then the mixture was concentrated and diluted with water and extracted three times with EtOAc (10 mL each). The combined organic layers were concentrated to give the desired compound as colorless oil (326 mg, 19.6% yield) MS (ESI): m/z=256.0 [M−56+H]+.
  • Method D4 BB70 3-[4-(Trifluoromethyl)phenoxy]azetidine
  • To a solution of tert-butyl 3-[4-(trifluoromethyl)phenoxy]azetidine-1-carboxylate (500 mg, 1.58 mmol, BB70a) in DCM (3 mL) was added TFA (1.0 mL, 0.950 mmol) at 25° C., the reaction was stirred at this temperature for 12 h. The mixture was concentrated and the residue was purified to via prep-HPLC to provide the compound as colorless solid (150 mg, 0.690 mmol, 43.8%). MS (ESI): m/z=218.1 [M+H]+.
  • BB72a tert-Butyl 4-(4-chloro-3-cyclopropyl-phenoxy)piperidine-1-carboxylate
  • To a solution of tert-butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate (500 mg, 1.28 mmol, BB90), potassium carbonate (354 mg, 2.56 mmol) and cyclopropylboronic acid (121 mg, 1.41 mmol) in 1,4-dioxane (5 mL) and water (1 mL) was added [1,1′-bis(diphenylphosphino)ferrocene]dichloro palladium(II) (187.28 mg, 0.260 mmol). The mixture was stirred at 100° C. under nitrogen atmosphere for 12 h. The reaction mixture was filtered and the filtrate was diluted with EtOAc (30 mL), washed with water and then brine, the organic phase was dried over Na2SO4, concentrated. The residue was purified by silica gel column (eluting with a gradient of 5%-10% EtOAc-PE) to give the compound as light yellow oil (220 mg, 48.9%). MS (ESI): m/z=296.1 [M−56+H]+.
  • BB73a tert-Butyl 4-(4-chloro-3-morpholino-phenoxy) piperidine-1-carboxylate
  • To a solution of tert-butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate (500 mg, 1.28 mmol, BB90), cesium carbonate (834 mg, 2.56 mmol), (R)-(+)-2,2′-bis(diphenylphosphino)-1,1′-binaphthalene (159 mg, 0.260 mmol) and morpholine (112 mg, 1.28 mmol) in DMF (10 mL) was added tris(dibenzylideneacetone)dipalladium(0) (187 mg, 0.260 mmol) and the mixture was stirred at 110° C. under nitrogen atmosphere for 12 h. The reaction mixture was filtered, the filtrate was diluted with EtOAc (30 mL), washed with water and then brine, the organic phase was dried over Na2SO4, and concentrated. The residue was purified by silica gel column (eluting with a gradient of 5%-10% EtOAc-PE) to give the desired compound (360 mg, 70.9% yield) as light yellow oil. MS (ESI): m/z=397.1 [M+H]+.
  • BB74a tert-Butyl 4-[2-methyl-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[2-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (2.0 g, 4.71 mmol, BB74b) in THF (40 mL) was added lithium methide (11.8 mL, 18.9 mmol) dropwise at −70° C. The mixture was stirred at −70° C. for 1 h and then stirred at 20° C. for 12 h. The mixture was poured into ice water (100 mL) and extracted three times with EtOAc (50 mL each). The combined organic layer was washed with brine (100 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum to yield the compound as light yellow solid (780 mg, 46%). MS (ESI): m/z=260.1 [M−100+H]+.
  • BB75a tert-Butyl 4-[2-cyano-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • To a solution of zinc cyanide (2214 mg, 18.9 mmol) and tert-butyl 4-[2-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (1600 mg, 3.77 mmol, BB74b) in DMA (30 mL) was added dppf (627 mg, 1.13 mmol), N,N-diisopropylethylamine (1.97 mL, 11.3 mmol), Zinc dust (245 mg, 3.77 mmol) and Pd2(dba)3 (1036 mg, 1.13 mmol) at 20° C., then the mixture was stirred at 140° C. under nitrogen atmosphere for 4 h. The mixture was filtered. The filtrate was poured into water (100 mL) and extracted three times with EtOAc (50 mL each). The combined organic layer was washed with brine (50 mL), dried over Na2SO4 and filtered. The filtrate was concentrated over vacuum to give the title compound as light brown solid (2.3 g, crude). MS (ESI): m/z=315.0 [M−56+H]+.
  • BB76a tert-Butyl 4-(oxazolo[5,4-c]pyridin-2-ylmethyl)piperidine-1-carboxylate
  • To a solution of hexachloroethane (2.47 g, 10.4 mmol) in toluene (20 mL) was added triphenylphosphine (3.28 g, 12.5 mmol) and NEt3 (4.65 mL, 33.4 mmol). The mixture was stirred at 80° C. for 5 min, then tert-butyl 4-[2-[(3-hydroxy-4-pyridyl)amino]-2-oxo-ethyl]piperidine-1-carboxylate (1.4 g, 4.17 mmol) was added and stirred at 80° C. for 12 h. The mixture was concentrated to remove toluene, then diluted with water (100 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude was purified by silica gel chromatography (PE:EtOAc=10:1 to 1:0) to give the compound as a yellow oil (814 mg, 21% yield). MS (ESI): m/z=318.1 [M+H]+.
  • Step a) tert-Butyl 4-[2-[(3-hydroxy-4-pyridyl)amino]-2-oxo-ethyl]piperidine-1-carboxylate
  • A solution of 4-aminopyridin-3-ol (3.0 g, 27.3 mmol) and 1-Boc-4-piperidylacetic acid (7.95 g, 32.7 mmol) in DMF (30 mL) was added HOBt (6.26 g, 40.9 mmol), EDCI (6.34 g, 40.87 mmol) and NEt3 (11.39 mL, 81.74 mmol). The mixture was stirred at 20° C. for 15 h. Then the mixture was concentrated, the residue taken up in water (100 mL), and then extracted three times with EtOAc (20 mL each). The organic phase was washed with brine, dried over Na2SO4 and concentrated. The residue was purified by reversed phase chromatography and lyophilized to give two batches of the desired compound. Batch 1 as colorless solid (1.2 g, 85% purity, 11.1%), and batch 2 as colorless solid (520 mg, 76.7% purity, 4.4% yield). MS (ESI): m/z=336.1 [M+H]+ for both batches.
  • BB77 4-Chloro-3-(2-piperidin-4-ylethynyl)pyridine
  • Intermediate BB77 was prepared in analogy to BB18, but using 3-bromo-4-chloro-pyridine in step a), to give the title compound as an orange solid. MS (ESI): m/z=221.1 [M+H]+.
  • BB78 3-Chloro-2-(2-piperidin-4-ylethynyl)pyridine
  • Intermediate BB78 was prepared in analogy to BB18, but using 2-bromo-3-chloro-pyridine in step a), to give the title compound as a yellow solid. MS (ESI): m/z=221.1 [M+H]+.
  • BB79 4-[2-(2-Chloro-4-fluorophenyl)ethynyl]piperidine
  • Intermediate BB79 was prepared in analogy to BB18, but using 1-bromo-2-chloro-4-fluoro-benzene in step a), to give the title compound as a white solid. MS (ESI): m/z=238.1 [M+H]+.
  • BB80 4-[2-(3-Chlorophenyl)ethynyl]piperidine
  • Intermediate BB80 was prepared in analogy to BB18, but using 1-bromo-3-chlorobenzene in step a), to give the title compound as a colorless amorphous solid. MS (ESI): m/z=220.2 [M+H]+.
  • BB81 4-[2-(4-Chlorophenyl)ethynyl]piperidine
  • Intermediate BB81 was prepared in analogy to BB18, but using 1-bromo-4-chlorobenzene in step a), to give the title compound as a yellow amorphous solid. MS (ESI): m/z=220.2 [M+H]+.
  • BB82 4-[2-(2-Chloro-4-chlorophenyl)ethynyl]piperidine
  • Intermediate BB82 was prepared in analogy to BB18, but using 1-bromo-2,4-dichloro-benzene in step a), to give the title compound as a light yellow amorphous solid. MS (ESI): m/z=254.1 [M+H]+.
  • BB83 4-[2-(2-Chlorophenyl)ethynyl]piperidin-4-ol
  • Intermediate BB83 was prepared in analogy to BB18, but using tert-butyl 4-ethynyl-4-hydroxypiperidine-1-carboxylate (CAS RN 275387-83-2) in step a), to give the title compound as a yellow amorphous solid. MS (ESI): m/z=218.1 [M−H2O+H]+.
  • BB84 3-[2-(2-Chlorophenyl)ethynyl]azetidine
  • To a solution tert-butyl 3-[2-(2-chlorophenyl)ethynyl]azetidine-1-carboxylate (0.035 g, 0.120 mmol) in DCM (0.6 mL) was added TFA (0.92.4 mL, 1.2 mmol) and the reaction mixture was stirred at room temperature for 2 h. The mixture was diluted with DCM, poured into a saturated aq. NaHCO3 solution and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4, filtered, evaporated and further dried on the high vacuum to give the crude title compound (0.02 g, 87%) as a light yellow oil. MS (ESI): m/z=192.0 [M+H]+.
  • Step a) tert-Butyl 3-[2-(2-chlorophenyl)ethyl]azetidine-1-carboxylate
  • The compound was prepared in analogy to intermediate BB 18, but using tert-butyl 3-ethynylazetidine-1-carboxylate (CAS RN 287193-01-5) in step a), to give the title compound as a white solid. MS (ESI): m/z=236.1 [M−56+H]+.
  • BB85 3-[2-(2,4-Dichlorophenyl)ethynyl]azetidine
  • Intermediate BB85 was prepared in analogy to intermediate BB84, but using 1-bromo-2,4-dichloro-benzene in step a), to give the title compound as a light yellow oil. MS (ESI): m/z=226.1 [M+H]+.
  • BB86 3-[2-(2-Chloro-4-fluoro-phenyl)ethynyl]azetidine
  • Intermediate BB86 was prepared in analogy to intermediate BB84, but using 1-bromo-2-chloro-4-fluoro-benzene in step a), to give the title compound as a yellow oil. MS (ESI): m/z=210.1 [M+H]+.
  • In analogy to BB9a the following building blocks were prepared from the respective building blocks
  • BB No. Systematic Name Starting materials MS, m/z
    BB54a tert-Butyl 4-[[2-methyl-4- tert-Butyl 4- 302.1
    (trifluoromethyl)phenyl]methyl] methylenepipendine-1- [M + H − 56]+
    piperidine-1-carboxylate carboxylate
    4-Bromo-3-methyl
    benzotrifluoride
    BB55a tert-Butyl 4-[[2-chloro-4- tert-Butyl 4- 322.0
    (trifluoromethyl)phenyl]methyl] methylenepiperidine-1- [M + H − 56]+
    piperidine-1-carboxylate carboxylate
    4-Bromo-3-
    chlorobenzotrifluoride
  • In analogy to BB15a the following building blocks were prepared from the respective building blocks.
  • BB No. Systematic Name Starting materials MS, m/z
    BB49a tert-Butyl 3-[(2- 2-Chlorobenzyl bromide 256.0
    chlorophenyl)methoxy]pyrrolidine- N-Boc-3-hydroxypyrrolidine [M − 56 + H]+
    1-carboxylate
    BB50a tert-Butyl 3[(3- 3-Chlorobenzyl bromide 256.0
    chlorophenyl)methoxy]pyrrolidine- N-BOC-3-hydroxypyrrolidine [M − 56 + H]+
    1-carboxylate
  • In analogy to BB9 step a, the following building blocks were prepared from the respective starting materials.
  • BB No. Systematic Name Starting materials MS, m/z
    BB47a tert-Butyl 3-[(2- 2-Chlorophenol 256.0
    chlorophenoxy)methyl]pyrrolidine- tert-Butyl 3- [M − 56 + H]+
    1-carboxylate (hydroxymethyl)pyrrolidine-1-
    carboxylate
    BB52a tert-Butyl 3-[(4- 4-Chlorophenol 256.0
    chlorophenoxy)methyl]pyrrolidine- tert-Butyl 3- [M − 56 + H]+
    1-carboxylate (hydroxymethyl)pyrrolidine-1-
    carboxylate
    BB70a tert-Butyl 3-[4- 4-(Trifluoromethyl)phenol Used
    (trifluoromethyl)phenoxy]azetidine- tert-Butyl 3-hydroxyazetidine-1- without
    1-carboxylate carboxylate further
    purification
    BB71a tert-Butyl 4-[4-chloro-3- 1 -Boc-4-hydroxypiperidine 324.0
    (trifluoromethyl)phenoxy]piperidine- 4-Chloro-3- [M − 56 + H]+
    1-carboxylate (trifluoromethyl)phenol
    BB74b tert-Butyl 4-[2-bromo-4- 2-Bromo-4- 369.9
    (trifluoromethyl)phenoxy]piperidine- (trifluoromethyl)phenol [M − 56 + H]+
    1-carboxylate 1-BOC-4-hydroxypiperidine
    BB89a tert-Butyl 3-[3- 3-Chlorophenol 256.0
    chlorophenoxy)methyl]pyrrolidine- tert-Butyl 3- [M − 56 + H]+
    1-carboxylate (hydroxymethyl)pyrrolidine-1-
    carboxylate
    BB90 tert-Butyl 4-(3-bromo-4-chloro- 3-Bromo-4-chlorophenol 336.0
    phenoxy) piperidine-1- 1-BOC-4-hydroxypiperidine [M − 56 + H]+
    carboxylate
  • Method D5 BB51 4-[[2-Cyclopropyl-4-(trifluoromethyl)phenyl]methyl]piperidine formic acid salt
  • To a mixture of tert-butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate (940 mg, 2.45 mmol, BB51a) in DCM (10 mL) was added TFA (2.0 mL, 2.45 mmol). The mixture was stirred at 20° C. for 12 h. The mixture was concentrated under vacuum. The residue was purified twice by prep-HPLC to furnish the desired compound as light yellow gum (111 mg, 12.4%). MS (ESI): m/z=284.2 [M+H]+.
  • Step a) 2-Bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene
  • A mixture of 2-bromo-1-methyl-4-(trifluoromethyl)benzene (5.5 g, 23.0 mmol, CAS RN 128-08-5), benzoyl peroxide (835 mg, 3.45 mmol) and NBS (4.07 g, 23.0 mmol) in CCl4 (50.0 mL, 23.0 mmol) was stirred at 70° C. for 5 h. The mixture was poured into water (20 mL) and extracted twice with DCM (20 mL each). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated in vacuum to give the compound as light yellow oil (7.1 g, 97%) which was used in the next step without further purification.
  • Step b) 2-Bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene
  • A mixture of 2-bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene (7.1 g, 22.3 mmol) and triethyl phosphite (30.0 mL) was stirred at 155° C. for 5 h. The mixture was concentrated in vacuum to remove triethyl phosphite. The residue was diluted with water (100 mL) and extracted three times with EtOAc (100 mL each). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (PE:EA=100:1 to 10:1) to give the title compound as light yellow oil (8 g, 21.3 mmol, 95.5%) which was used in the subsequent step without further purification.
  • Step c) tert-Butyl 4-(2-bromo-4-(trifluoromethyl)benzylidene)piperidine-1-carboxylate
  • A mixture of 2-bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene (6.9 g, 18.4 mmol) in THF (100 mL) was added NaH (2.21 g, 55.2 mmol) at 0° C. The mixture was stirred at 0° C. for 1 h, then 1-Boc-4-piperidone (7.33 g, 36.8 mmol, CAS RN 79099-07-3) was added and the mixture was stirred at 20° C. for 12 h. The mixture was poured into water (100 mL) and extracted three times with EtOAc (100 mL each). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (PE:EA=100:1 to 50:1) to yield the desired compound as off-white solid (4 g, 9.52 mmol, 51.7%). MS (ESI): m/z=365.9 [M−56+H]+.
  • Step d) tert-Butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[[2-bromo-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (2.0 g, 4.76 mmol), cyclopropylboronic acid (818 mg, 9.52 mmol, CAS RN 411235-57-9) and potassium carbonate (1973 mg, 14.3 mmol) in DMF (10 mL) and water (0.5 mL) was stirred at 80° C. for 12 h under nitrogen atmosphere. The mixture was poured into water (50 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine (50 mL), dried over Na2SO4 and concentrated in vacuum. The residue was purified by prep-HPLC to give the compound as light yellow oil (1020 mg, 56.2% yield) MS (ESI): m/z=326.0 [M−56+H]+.
  • Step e) tert-Butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[[2-cyclopropyl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (1000 mg, 2.62 mmol) and PtO2 (100 mg, 0.440 mmol) in EtOAc (20 mL) was stirred at 20° C. for 12 h under hydrogen atmosphere (1520 mm Hg). Then the mixture was filtered and the filtrate was concentrated to yield the compound as light yellow solid (940 mg, 93.5% yield). MS (ESI): m/z=328.2 [M+H]+.
  • Method D6 BB92 N-methyl-N-[4-(trifluoromethyl)phenyl]piperidin-4-amine; trifluoroacetate salt
  • To a solution of tert-butyl 4-[N-methyl-4-(trifluoromethyl)anilino]piperidine-1-carboxylate (150 mg, 0.420 mmol) in DCM (1 mL) was added TFA (0.1 mL) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction mixture was concentrated in vacuum. The residue was purified by pre-HPLC (in the presence of TFA) to give the desired product as yellow solid (120 mg, 77.0%). MS (ESI): m/z=259.2 [M+H]+.
  • Step a) tert-Butyl 4-[4-(trifluoromethyl)anilino]piperidine-1-carboxylate
  • To a solution of p-trifluoromethylaniline (1.17 mL, 9.31 mmol, CAS RN 455-14-1) in DCM (30 mL) was added AcOH (0.560 g, 9.31 mmol) and 1-BOC-4-piperidone (2.78 g, 14.0 mmol, CAS RN 79099-07-3). Then 1M BH3/THF solution (27.9 mL, 27.9 mmol) was added carefully at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at 25° C. for 12 h. The mixture was poured into saturated aqueous NH4Cl solution (30 mL) and extracted three times with EtOAc. The combined organic layers were washed twice with water H2O, and then brine, dried over Na2SO4 and concentrated in vacuum to afford yellow residue, which was purified by silica gel column eluting with a gradient of PE:EtOAc (20:1 to 5:1) to give the desired product as white solid (2.0 g, 62.4%). MS (ESI): m/z=289.1 [M−56+H]+.
  • Step b) tert-Butyl 4-[N-methyl-4-(trifluoromethyl)anilino]piperidine-1-carboxylate
  • To a solution of NaH (52.3 mg, 60.0% wt %, 1.31 mmol) in DMF (5 mL) was added tert-butyl 4-[4-(trifluoromethyl)anilino]piperidine-1-carboxylate (300 mg, 0.870 mmol) at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 15 min, and then iodomethane (371 mg, 2.61 mmol) was added. The reaction mixture was stirred at 80° C. for 12 hrs. The reaction mixture was poured into water (20 mL) and extracted three times with EtOAc, the combined organic layers were washed twice with water and brine, dried over sodium sulfate and concentrated in vacuum to afford light yellow residue, which was purified by silica gel column eluting with a gradient of PE:EtOAc (20:1 to 5:1) to give the desired product as white solid (160 mg, 51.3%). MS (ESI): m/z=303.1 [M−56+H]+.
  • BB93 N-methyl-N-(4-(trifluoromethyl)phenyl)azetidin-3-amine (trifluoroacetic acid salt)
  • The title compound was prepared in analogy to method D6 from tert-butyl 3-[N-methyl-4-(trifluoromethyl)anilino]azetidine-1-carboxylate (48%). MS (ESI): m/z=231.1 [M+H]+.
  • Step a) tert-Butyl 3-[4-(trifluoromethyl)anilino]azetidine-1-carboxylate
  • To a solution of p-trifluoromethylaniline (0.780 mL, 6.21 mmol, CAS RN 455-14-1), AcOH (1.86 g, 31.0 mmol) and 1-BOC-3-azetidinone (2.13 g, 12.4 mmol, CAS RN 398489-26-4) in EtOH (10 mL) was added NaBH3CN (1.95 g, 31.0 mmol) at 25° C. The mixture was stirred at 25° C. for 12 h. The reaction mixture was poured into saturated aqueous NH4Cl solution (20 mL) and extracted twice with EtOAc. The combined organic layers were washed twice with H2O and brine, dried over sodium sulfate and concentrated in vacuum to afford yellow residue, which was purified by silica gel column eluting with a gradient of PE:EtOAc (10:1 to 5:1) to give the desired product as white solid (340 mg, 17.3%). MS (ESI): m/z=261.1 [M−56+H]+.
  • Step b) tert-Butyl 3-[N-methyl-4-(trifluoromethyl)anilino]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[4-(trifluoromethyl)anilino]azetidine-1-carboxylate (300 mg, 0.950 mmol) in DMF (5 mL) was added NaH (45.5 mg, 60% wt %/o, 1.14 mmol) at 0° C. The mixture was stirred for 15 min, and then iodomethane (404 mg, 2.85 mmol) was added. The reaction mixture was stirred at 25° C. for 12 h. The reaction mixture was poured into H2O (20 mL) and extracted twice with EtOAc. The combined organic layers were washed three times with H2O and brine, dried over Na2SO4 and concentrated in vacuum to afford yellow residue. The residue was purified by silica gel column eluting with a gradient of PE:EtOAc (10:1 to 5:1) to give the desired product as white solid (310 mg, 98.9%). MS (ESI): m/z=275.2 [M−56+H]+.
  • Method D7 BB94 N-methyl-N-(piperidin-4-yl)-2-(3-(trifluoromethyl)phenyl)acetamide hydrochloride
  • To a solution of tert-butyl 4-[methyl-[2-[3-(trifluoromethyl)phenyl]acetyl]amino]piperidine-1-carboxylate (0.080 g, 200 μmol) in DCM (I mL) was added a 2 M HCl solution in diethyl ether (999 μL, 2 mmol). The reaction mixture was stirred at RT overnight and then concentrated in vacuo to afford the title compound (67 mg, 199 μmol) as an off-white solid. MS (ESI): m/z=301.2 [M+H]+.
  • Step a) tert-Butyl 4-[methyl-[2-[3-(rifluoromethyl)phenyl]acetyl]amino]piperidine-1-carboxylate
  • To a stirred mixture of 2-(3-(trifluoromethyl)phenyl)acetic acid (105 mg, 513 μmol, CAS RN 351-35-9) in DMF (5 mL) was added HATU (195 mg, 513 μmol) and DIPEA (181 mg, 244 μL, 1.4 mmol). After 15 min. stirring, tert-butyl 4-(methylamino)piperidine-1-carboxylate (0.100 g, 467 μmol, CAS RN 147539-41-1) was added and the reaction mixture was stirred at RT for 2 h. The reaction mixture was diluted with DCM and washed with H2O. The org, phase was concentrated to give a crude product which was purified by flash chromatography on a 20 g SiO2 column, using an eluent mixture of n-heptane and EtOAc (0% to 100%) to afford the desired compound as a light yellow oil (85 mg, 213 μmol). MS (ESI): m/z=459.259 [M+CH3CN+NH4]+.
  • Method D8 BB194 3-(4-Chloro-3-cyclopropylphenoxy)azetidine
  • To a solution of tert-butyl 3-(4-chloro-3-cyclopropylphenoxy)azetidine-1-carboxylate (0.023 g, 0.057 mmol) in DCM (1 mL) was added TFA (0.088 mL, 1.14 mmol) and the reaction mixture stirred at room temperature for 18 hours. The mixture was diluted with DCM, poured into a sat. aq. NaHCO3 solution and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4, filtered and evaporated to dryness to yield the crude title compound (0.007 g, 35%) as a colorless oil. MS (ESI): m/z=224.1 [M+H]+.
  • Step a) tert-Butyl 3-(3-bromo-4-chlorophenoxy)azetidine-1-carboxylate
  • In a sealed tube, 3-bromo-4-chlorophenol (0.1 mg, 0.482 mmol) and tert-butyl 3-hydroxyazetidine-1-carboxylate (0.083 g, 0.482 mmol) were dissolved in toluene (1.5 mL). The vial was degassed with argon, then (tributylphosphoranylidene)acetonitrile (CAS RN 157141-27-0, 0.195 mL, 0.723 mmol) was added and the reaction mixture heated to 100° C. for 30 minutes. The mixture was diluted with EtOAc, poured into sat. aq. NaHCO3 solution and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography eluting with a 0 to 20% EtOAc/heptane gradient to give the title compound (0.116 g, 53%) as a yellow oil. MS (ESI): m/z=308.1 [M−56+H]+.
  • Step b) tert-Butyl 3-(4-chloro-3-cyclopropylphenoxy)azetidine-1-carboxylate
  • In a microwave vial, tert-butyl 3-(3-bromo-4-chlorophenoxy)azetidine-1-carboxylate (0.075 g, 0.165 mmol), cyclopropylboronic acid (0.021 g, 0.248 mmol) and K2CO3 (0.046 g, 0.331 mmol) were mixed in dioxane (1.6 mL). Then, water (0.4 mL) was added followed by bis(triphenylphosphine)palladium (II) chloride (0.012 g, 0.016 mmol) and the reaction mixture heated at 130° C. under microwave irradiation for 1 hour. The reaction mixture was diluted with EtOAc, poured into water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel flash chromatography, eluting with a 0 to 10% EtOAc/heptane gradient to give the title compound (0.023 g, 43%) as a colorless oil. MS (ESI): m/z=268.2 [M−56+H]+.
  • Method D9 BB197 3-(2-Chloro-3-cyclopropylphenoxy)azetidine, trifluoroacetate salt
  • To a solution of tert-butyl 3-(2-chloro-3-cyclopropyl-phenoxy)azetidine-1-carboxylate (0.1 g, 0.310 mmol) in DCM (2.5 mL) was added TFA (0.25 mL) and the reaction mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo to give the crude title compound (0.083 g, 80% yield) as a dark brown oil. MS (ESI): m/z=224.6 [M+H]+.
  • Step a) tert-Butyl 3-(3-bromo-2-chloro-phenoxy)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (0.5 g, 2.89 mmol) and 3-bromo-2-chloro-phenol (0.5 g, 2.41 mmol) in THF (10 mL) were added PPh3 (0.948 g, 3.62 mmol) followed by diethyl azodicarboxylate (0.47 mL, 3.62 mmol) and the reaction mixture was stirred at room temperature for 12 hours. The mixture was purified by reversed phase HPLC to give the title product (0.4 g, 46%) as a light yellow oil. MS (ESI): m/z=308.3 [M−56+H]+.
  • Step b) tert-Butyl 3-(2-chloro-3-cyclopropylphenoxy)azetidine-1-carboxylate
  • In a sealed tube, cyclopropylboronic acid (0.071 g, 0.830 mmol), tert-butyl 3-(3-bromo-2-chloro-phenoxy)azetidine-1-carboxylate (0.2 g, 0.550 mmol) and Na2CO3 (0.117 g, 1.1 mmol) were mixed in 1,4-dioxane (5 mL) and water (1 mL). Then, Pd(dppf)Cl2 (0.040 g, 0.060 mmol) was added and the mixture was stirred to 110° C. for 12 hours. The mixture was purified by reversed phase HPLC to give the title compound (0.12 g, 67%) as a light yellow oil. MS (ESI): m/z=268.1 [M−56+H]+.
  • Method D10 BB202 5-(4-Piperidyloxy)-2-(trifluoromethyl)benzonitrile, trifluoroacetate
  • To a solution of tert-butyl 4-[3-cyano-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (0.05 g, 0.140 mmol) in DCM (1.5 mL) was added TFA (0.2 mL) and the reaction mixture stirred at room temperature for 12 hours. The mixture was concentrated in vacuo to give the crude title compound (0.051 g, 98%) as a light brown oil. MS (ESI): m/z=271.6 [M+H]+.
  • Step a) tert-Butyl 4-[3-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • To a solution of 3-bromo-4-(trifluoromethyl)phenol (0.5 g, 2.54 mmol) and 1-Boc-4-hydroxypiperidine (0.512 g, 2.54 mmol) in THF (8.5 mL) were added PPh3 (1 g, 3.82 mmol) followed by diethyl azodicarboxylate (0.665 g, 3.82 mmol) and the reaction mixture was stirred at room temperature for 12 hours. The mixture was purified by silica gel flash chromatography, eluting with PE:EtOAc 5:1 to give the title compound (0.5 g, 47%) as a light yellow oil. MS (ESI): m/z=370.2 [M−56+H]+.
  • Step b) tert-Butyl 4-[3-cyano-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • In a sealed tube, tert-butyl 4-[3-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (0.2 g, 0.470 mmol), Zn(CN)2 (0.111 g, 0.940 mmol), CuI (0.09 g, 0.470 mmol) were mixed in DMF (10 mL). Then, Pd(PPh3)4 (0.109 g, 0.090 mmol) was added and the reaction mixture stirred to 130° C. for 16 hours. The mixture was purified by reversed phase HPLC to give the title product (0.05 g, 29%) as a colorless oil. MS (ESI): m/z=315.5 [M−56+H]+.
  • Method E Example 263 (+)-5-[1-[(4aR,8aS)-3-Oxo-4,4a,5,7,88a-hexahydropyrido[4,3-b][1,4]oxazine-6-carbonyl]azetidin-3-yl]oxy-2-(trifluoromethyl)benzonitrile
  • Figure US20200308190A1-20201001-C00308
  • In a sealed tube, (+)-(4aR,8aS)-6-[3-[3-bromo-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (BB 205, 0.2 g, 0.420 mmol), Zn(CN)2 (0.098 g, 0.840 mmol), Zn (0.027 g, 0.420 mmol), dppf (0.232 g, 0.420 mmol). Hünig's base (0.108 g, 0.840 mmol) were mixed in DMA (10 mL) and the mixture was degassed. Then, Pd2(dba)3 (76.59 mg, 0.080 mmol) was added and the reaction mixture was stirred at 130° C. for 16 h. The mixture was purified by reversed phase HPLC to give the title compound (0.055 g, 30%) as a light yellow solid. MS (ESI): m/z=425.3 [M+H]+.
  • Method F Example 265 (+)-(4aR,8aS)-6-[3-[3-(2-Azaspiro[3.3]heptan-2-yl)-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one
  • Figure US20200308190A1-20201001-C00309
  • In a sealed tube, (+)-(4aR,8aS)-6-[3-[3-bromo-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one (BB203, 0.2 g, 0.420 mmol), 2-azaspiro[3.3]heptane (CAS RN 665-04-03, 0.117 g, 0.630 mmol), BINAP (0.052 g, 0.080 mmol) and K2CO3 (0.173 g, 1.25 mmol) were mixed in DMF (10 mL) and the mixture was degassed. Then, Pd2(dba)3 (76.59 mg, 0.080 mmol) was added and the reaction mixture was stirred to 110° C. for 16 hours. The reaction mixture was filtered off, the filtrate diluted with water (50 mL) and extracted with EtOAc (3×20 mL). Combined organics were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by reversed phase HPLC to give the title compound (0.06 g, 29%) as a white solid. MS (ESI): m/z=495.1 [M+H]+.
  • Method G Example 293 (4aR,8aS)-6-(3-(4-Hydroxy-2-(trifluoromethyl)phenethyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one
  • Figure US20200308190A1-20201001-C00310
  • Boron tribromide (11.3 mg, 4.29 μL, 45.3 μmol) was added to an ice cooled solution of (4aR,8aS)-6-(3-(4-methoxy-2-(trifluoromethyl)phenethyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one (Example 216, 20 mg, 45.3 μmol) in DCM (0.5 mL). The reaction mixture was stirred at ambient temperature for 3 h. A saturated solution of aqueous NaHCO3 was added and the mixture was extracted with AcOEt. The layers were separated, the organic layer was dried over Na2SO4, filtered and the solvent removed under reduced pressure. The crude product was purified by prep. HPLC to give the title compound (19%) as colorless solid. MS (ESI): m/z=427.2 [M+H]+.
  • The following examples listed in the table below were prepared in analogy to the procedure described for the preparation of Example 265 by using the indicated intermediates and/or commercially available compounds and using the mentioned purification method such as reversed-phase HPLC or silica gel flash chromatography.
  • Ex Systematic Name/Structure Intermediates MS, m/z
    266
    Figure US20200308190A1-20201001-C00311
    BB203 BH66 and 3-Methylazetidine hydrochloride (CAS RN 935669-28-6) 469.2 [M + H]+
    267
    Figure US20200308190A1-20201001-C00312
    BB203 and Difluoroazetidine hydrochloride (CAS RN 288315-03-7) 491.2 [M + H]+
    268
    Figure US20200308190A1-20201001-C00313
    BB203 and 3-Fluoro-3-methyl- azetidine hydrochloride (CAS RN 1427379-42-7) 487.3 [M + H]+
    269
    Figure US20200308190A1-20201001-C00314
    BB203 and 6,6-Difluoro-2- azaspiro[3.3]heptane (CAS RN 1354952-05-8) 531.1 [M + H]+
    270
    Figure US20200308190A1-20201001-C00315
    BB203 and 5-Oxa-2- azaspiro[3.5]nonane (CAS RN 138387-19-6) 525.3 [M + H]+
    271
    Figure US20200308190A1-20201001-C00316
    Example 258 and 2-Azaspiro[3.3]heptane (CAS RN 665-04-03) 461.1 [M + H]+
    272
    Figure US20200308190A1-20201001-C00317
    Example 258 and 3-Methylazetidine hydrochloride (CAS RN 935669-28-6) 435.1 [M + H]+
    273
    Figure US20200308190A1-20201001-C00318
    Example 258 and 3-Fluoro-3-methyl- azetidine hydrochloride (CAS RN 1427379-42-7) 453.2 [M + H]+
    274
    Figure US20200308190A1-20201001-C00319
    Example 258 and 3-tert-Butoxyazetidine (CAS RN 1147530-63-9) 493.2 [M + H]+
    275
    Figure US20200308190A1-20201001-C00320
    Example 258 and 5-Oxa-2- azaspiro[3.4]octane (CAS RN 145309-24-6) 477.2 [M + H]+
    276
    Figure US20200308190A1-20201001-C00321
    Example 258 and 5-Oxa-2- azaspiro[3.5]nonane (CAS RN 138387-19-6) 491.2 [M + H]+
    277
    Figure US20200308190A1-20201001-C00322
    BB204 and 2-Azaspiro[3.3]heptane (CAS RN 665-04-03) 461.3 [M + H]+
    278
    Figure US20200308190A1-20201001-C00323
    BB204 and Pyrrolidine 435.3 [M + H]+
  • In analogy to the methods described herein above, the following building blocks were prepared from the respective starting material indicated in the table below.
  • BB No. Systematic Name Starting material Method MS, m/z
    BB47  3-[(2- tert-Butyl 3-[(2- D3 212.1
    Chlorophenoxy)methyl]pyrrolidine; chlorophenoxy)methyl]pyrrolidine- [M + H]+
    hydrochloride salt 1-carboxylate BB47a
    BB48  4-[[2-Fluoro-4- tert-Butyl 4-[[2-fluoro-4- D2 262.1
    (trifluoromethyl)phenyl]methyl]piperidine; (trifluoromethyl)phenyl]methyl] [M + H]+
    formic acid salt piperidine-1-carboxylate BB48a
    BB49  3-[(2- tert-Butyl 3-[(2- D3 212.1
    Chlorophenyl)methoxy]pyrrolidine; chlorophenyl)methoxy]pyrrolidine- [M + H]+
    hydrochloride salt 1-carboxylate BB49a
    BB50  3-[(3- tert-Butyl 3-[(3- D3 212.1
    Chlorophenyl)methoxy]pyrrolidine; chlorophenyl)methoxy]pyrrolidine- [M + H]+
    hydrochloride salt 1-carboxylate BB50a
    BB52  3-[(4- tert-Butyl 3-[(4- D3 212.1
    Chlorophenyl)methoxy]pyrrolidine; chlorophenyl)methoxy]pyrrolidine- [M + H]+
    hydrochloride salt 1-carboxylate BB52a
    BB53  3-[(4- tert-Butyl 3-[(4- D2 Used
    Chlorophenyl)methoxy]pyrrolidine chlorophenyl)methoxy]pyrrolidine- without
    formic acid salt 1-carboxylate BB53a further
    purification
    BB54  4-[[2-Methyl-4- tert-Butyl 4-[[2-methyl-4- D2 258.2
    (trifluoromethyl)phenyl]methyl]piperidine; (trifluoromethyl)phenyl]methyl] [M + H]+
    formic acid salt piperidine-1-carboxylate BB54a
    BB55  4-[[2-Chloro-4- tert-Butyl 4-[[2-chloro-4- D1 278.0
    (trifluoromethyl)phenyl]methyl]piperidine; (trifluoromethyl)phenyl]methyl] [M + H]+
    trifluoroacetate salt piperidine-1-carboxylate BB55a
    BB71  4-[4-Chloro-3- tert-Butyl 4-[4-chloro-3- D3 280.0
    (trifluoromethyl)phenoxy]piperidine; (trifluoromethyl)phenoxy]piperidine- [M + H]+
    hydrochloride salt 1-carboxylate BB71a
    BB72  4-(4-Chloro-3-cyclopropyl- tert-Butyl 4-(4-chloro-3- D1 Used
    phenoxy) piperidine; cyclopropyl- without
    trifluoroacetate salt phenoxy)piperidine-1- further
    carboxylate BB72a purification
    BB73  4-[2-Chloro-5-(4-piperidyloxy)phenyl] tert-Butyl 4-(4-chloro-3- D3 Used
    morpholine hydrochloride morpholino- without
    phenoxy)piperidine-1- further
    carboxylate BB73a purification
    BB74  4-[2-Methyl-4- tert-Butyl 4-+2-methyl-4- D1 260.2
    (trifluoromethyl)phenoxy]piperidine; (trifluoromethyl)phenoxy]piperidine- [M + H]+
    trifluoroacetate salt 1-carboxylate BB74a
    BB75  2-(4-Piperidyloxy)-5- tert-Butyl 4-[2-cyano-4- D4 271.1
    (trifluoromethyl)benzonitrile (trifluoromethyl)phenoxy]piperidine- [M + H]+
    1-carboxylate BB75a
    BB76  2-(4- tert-Butyl 4-(oxazolo[5,4-c]pyridin- D1 218.1
    Piperidylmethyl)oxazolo[5,4-c]pyridine; 2-ylmethyl)piperidine-1- [M + H]+
    trifluoroacetate salt carboxylate BB76a
    BB89  3-[(3- tert-Butyl 3-[(3- D3 212.1
    Chlorophenoxy)methyl]pyrrolidine; chlorophenoxy)methyl]pyrrolidine- [M + H]+
    hydrochloride salt 1-carboxylate BB89a
    BB192 4-[2-Fluoro-4- tert-Butyl 4- D3 264.2
    (trifluoromethyl)phenoxy]piperidine; hydroxypiperidine-1- [M + H]+
    hydrochloride salt carboxylate
    BB193 4-[3-Chloro-4- tert-Butyl 4-[3-chloro-4- D3 280.1
    (trifluoromethyl)phenoxy]piperidine; (trifluoromethyl)phenoxy]piperidine- [M + H]+
    hydrochloride salt 1-carboxylate
    BB195 4-[2-Chloro-3- tert-Butyl 4-[2-chloro-3- D8 280.1
    (trifluoromethyl)phenoxy]piperidine; (trifluoromethyl)phenoxy]piperidine [M + H]+
    hydrochloride salt 1-carboxylate
    BB196 3-(3-Bromo-2-chloro- tert-Butyl 3-(3-bromo-2- D1 263.0
    phenoxy)azetidine; chloro-phenoxy)azetidine-1- [M + H]+
    trifluoroacetate salt carboxylate
    BB198 3-[3-Bromo-4- tert-Butyl 3[3-bromo-4- D9 296.4
    (trifluoromethyl)phenoxy]azetidine; (trifluoromethyl)phenoxy]azetidine- [M + H]+
    trifluoroacetate salt 1-carboxylate
    BB199 3-[3-Cyclopropyl-4- tert-Butyl 3-[3-cyclopropyl-4- D9 258.1
    (trifluoromethyl)phenoxy]azetidine; (trifluoromethyl)phenoxy]azetidine- [M + H]+
    trifluoroacetate salt 1-carboxylate
    BB200 3-[3-Chloro-4- tert-Butyl 3-[3-chloro-4- D1 252.5
    (trifluoromethyl)phenoxy]azetidine; (trifluoromethyl)phenoxy]azetidine- [M + H]+
    trifluoroacetate salt 1-carboxylate
    BB201 3-(3-Bromo-5-chloro- tert-Butyl 3-(3-bromo-5- D1 263.9
    phenoxy)azetidine; trifluoroacetate salt chloro-phenoxy)azetidine-1- [M + H]+
    carboxylate
    BB205 3-(3-Bromo-4-chloro- tert-Butyl 3-(3-bromo-4- D1 263.9
    phenoxy)azetidine; chlorophenoxy)azetidine-1- [M + H]+
    trifluoroacetate salt carboxylate
  • BB91 4-[[2-Pyrrolidin-1-yl-4-(trifluoromethyl)phenyl]methyl]piperidine; formic acid salt
  • A solution of tert-butyl 4-[[2-pyrrolidin-1-yl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate (500 mg, 1.21 mmol) in 6 M HCl in MeOH solution (10.0 mL) was stirred at 20° C. for 1 h. The mixture was concentrated under vacuum, purified by reversed phase column to give the title compound as an orange oil (84.4 mg, 21.8% yield). MS (ESI): m/z=313.2 [M+H]+.
  • Step a) Tert-butyl 4-[[2-pyrrolidin-1-yl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[[2-bromo-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (800 mg, 1.90 mmol; BB51, step c), pyrrolidine (163 mg, 2.28 mmol), Ruphos (4.25 mg, 0.010 mmol) and potassium tert-butoxide (320 mg, 2.86 mmol) in toluene (15 mL) was added palladium(II) acetate (1.28 mg, 0.010 mmol). The mixture was stirred at 80° C. for 15 h under N2 atmosphere. The mixture was filtered and concentrated under vacuum to remove toluene. The mixture was diluted with H2O (40 mL) and extracted three times with EtOAc (40 mL each). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EtOAc=1:0 to 8:1) to give the compound as light yellow oil (552 mg, 1.34 mmol, 36.7%) MS (ESI): m/z=411.1 [M+H]+.
  • Step b) Tert-butyl 4-[[2-pyrrolidin-1-yl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[[2-pyrrolidin-1-yl-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (525 mg, 0.660 mmol) in MeOH (20 mL) was added wet Pd/C (˜52 mg) and the mixture was stirred at 20° C. under H2 atmosphere (balloon) for 1 h. The mixture was filtered and concentrated under vacuum to give the desired compound as colorless oil (500 mg) which was used in the next step without further purification.
  • BB95 3-[2-[2-fluoro-6-(trifluoromethyl)phenyl)phenyl]ethyl]azetidine 4-methylbenzenesulfonate
  • To an solution of 3-[2-[2-fluoro-6-(trifluromethyl)phenyl]ethyl]azetidine-1-carboxylate (50 mg, 144 μmol, Eq: 1) in EtOAc (0.8 mL) was added 4-methylbenzenesulfonic acid monohydrate (29.7 mg, 173 μmol, Eq: 1.2) and the mixture was heated at reflux for 1.5 hours. The clear, colorless solution was allowed to cool down to RT. No precipitation occurred. The solution was evaporated to give the desired product as a colorless foam. MS (ESI): m/z=248.1 [M−TsOH+H]+.
  • Step a) tert-butyl 3-[(E)-2-[2-fluoro-6-(trifluoromethyl)phenyl]ethenyl]azetidine-1-carboxylate
  • To an ice-cold solution of diethyl (2-fluoro-4-(trifluoromethyl)benzyl)phosphonate (300 mg, 955 μmol) in THF (2 mL) was added sodium hydride 55% in mineral oil (41.7 mg, 955 μmol) and the mixture was stirred at this temperature for 30 minutes. To the light brown mixture was added dropwise a solution of tert-butyl 3-formylazetidine-1-carboxylate (177 mg, 955 μmol) in THF (1 mL). This led to an immediate discolouration of the reaction mixture. Stirring was continued for 1 hours at ice-bath temperature followed by stirring at RT for 1.5 hours. The reaction mixture was poured into water and ethyl acetate and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic layers were washed once with brine, dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 12 g column using an MPLC system eluting with a gradient of n-heptane:ethyl acetate (100:0 to 25:75) to get the desired compound as a colorless solid (0.108 g; 32.8%). MS (ESI): m/z=290.2 [M−56+H]+.
  • Step b) tert-butyl 3-[2-[2-fluoro-6-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl (E)-3-(2-fluoro-4-(trifluoromethyl)styryl)azetidine-1-carboxylate (105 mg, 304 μmol) in MeOH (1 mL) and Ethyl acetate (1 mL) was added Pd/C 10% (11 mg, 304 μmol) and the mixture was stirred under a hydrogen atmosphere at 1.7 bar and RT for 30 minutes. The suspension was filtered. The filtrate was evaporated to get the desired compound as a colorless oil (0.104 g; 98.5%). MS (ESI): m/z=292.2 [M−56+H]+.
  • BB96 4-((2-chloro-4-fluorophenoxy)methyl)azepane hydrochloride
  • To a solution of tert-butyl 4-((2-chloro-4-fluorophenoxy)methyl)azepane-1-carboxylate (620 mg, 1.73 mmol) in DCM (7.5 ml) was added HCl in ether 2M (10 ml, 20 mmol) and the reaction mixture was stirred overnight at rt. The mixture was concentrated in vacuo, the crude material collected as a white solid (490 mg, 1.67 mmol, 96.1%) and used directly on the next step. LC-MS (ESI): m/z: 258.2 [M+H]+
  • Step a) tert-butyl 4-((2-chloro-4-fluorophenoxy)methyl)azepane-1-carboxylate
  • In a 25 ml four-necked sulphonation flask under argon, tert-butyl 4-(hydroxymethyl)azepane-1-carboxylate (480 mg, 2.09 mmol) was dissolved in THF (10 ml). Subsequently, 2-chloro-4-fluorophenol (337 mg, 251 μl, 2.3 mmol) and triphenylphosphine (604 mg, 2.3 mmol) were added and the clear solution was stirred for 5 min at rt. The mixture was cooled to 0° C. and DEAD (401 mg, 365 μl, 2.3 mmol) was added in portions over 10 min. The mixture was stirred for 1 hr at 0° C., then overnight at rt. The mixture was taken up into EtOAc (50 ml), washed with water (2×25 ml), organic phase washed with 1M NaOH (3×25 ml), brine (20 ml), dried with Na2SO4, filtered and concentrated in vacuo. Residue was dissolved in n-Heptane/diethylether and the mixture stirred for 30 min, the TPPO precipitate filtered and the crude concentrated in vacuo. The crude material was adsorbed on Isolute® and purified by flash column chromatography (0-30% EtOAc/Heptane) over silica gel (50 g) to afford the desired product (630 mg, 1.76 mmol, 84.1%) as a yellow oil. LC-MS (ESI): m/z: 302.1 [M−56+H]+
  • BB97 4-[[4-(trifluoromethyl)phenyl]methyl]azepane hydrochloride
  • To a solution of tert-butyl 4-(4-(trifluoromethyl)benzyl)azepane-1-carboxylate (88 mg, 246 μmol, Eq: 1) in DCM (1.5 ml) was added HCl in ether 2M (3.08 ml. 6.16 mmol) and the reaction mixture was stirred overnight at room temperature. The mixture was concentrated in vacuo, the crude material collected as a white solid (71 mg, 0.24 mmol, 98.2%) and used directly on the next step. LC-MS (ESI): m/z: 258.2 [M+H]+
  • Step a: Triphenyl(4-(trifluoromethyl)benzyl)phosphonium bromide
  • Triphenylphosphine (1.84 g, 7 mmol) and 1-(bromomethyl)-4-(trifluoromethyl)benzene (1.61 g, 6.74 mmol) were dissolved in xylene (35 ml). The reaction mixture was heated to reflux at 155° C. for 3.5 h and then cooled to room temperature. The precipitated white crystalline solid was collected by filtration, washed with diethyl ether and dried in vacuo. The final compound (3.30 g, 6.58 mmol, 97.7% yield) was obtained as a white powder and directly used on the next step. LC-MS (ESI): m/z: 421.2 [M+H]+
  • Step b: tert-butyl (E)-4-(4-(trifluoromethyl)benzylidene)azepane-1-carboxylate
  • A suspension of sodium hydride (88.6 mg, 2.22 mmol) in DMF (7.5 ml) was cooled in an ice bath, then triphenyl(4-(trifluoromethyl)benzyl)phosphonium bromide (1.11 g, 2.22 mmol) was added. The suspension was stirred at 0° C. for 5 min. then for 25 min at rt. tert-butyl 4-oxoazepane-1-carboxylate (315 mg, 1.48 mmol) was added and the resulting mixture was stirred at 80° C. for 28 h. The mixture was concentrated in vacuo, diluted with water (50 ml) and EtOAc (40 ml) and extracted EtOAc (3×30 ml). The combined organic fractions were washed with water, 10% LiCl solution, dried with Na2SO4 and concentrated in vacuo. The residual oil was treated with Et2O in order to precipitate the triphenylphosphoxide that was filtered off The solution was concentrated in vacuo and the residue was purified by flash column chromatography (0-35% EtOAc/Heptane) over silica gel (50 g) to afford the desired product (92 mg, 259 μmol, 17.5% yield) as a yellow oil. LC-MS (ESI): m/z: 300.2 [M−56+H]+
  • Step c: tert-butyl 4-(4-(trifluoromethyl)benzyl)azepane-1-carboxylate
  • A solution of tert-butyl (E)-4-(4-(trifluoromethyl)benzylidene)azepane-1-carboxylate (90 mg, 253 μmol) was dissolved in MeOH (2.5 ml). The reaction vessel was evacuated and back-filled with argon 5 times. Under argon. Pd—C (13.5 mg, 12.7 μmol) was added and the atmosphere was replaced with hydrogen three times. The reaction was stirred under a hydrogen atmosphere at 1 bar for 24 h. The atmosphere was replaced with argon and the reaction mixture was filtered over a pad of Dicalite. The filter cake was washed with methanol. The filtrate was concentrated in vacuo to give the desired product (89 mg, 249 μmol, 98.3% yield) as a colorless oil which was used without further purification. LC-MS (ESI): m/z: 302.2 [M−56+H]+
  • BB98 3-((2-Chloro-4-(trifluoromethyl)phenyl)thio)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)thio)azetidine-1-carboxylate (110 mg, 299 μmol) was dissolved in DCM (2 mL) and TFA (273 mg, 184 μL, 2.39 mmol) was added. The reaction mixture was stirred at RT for 3 h. Volatiles were removed in vacuo to yield 110 mg of a light yellow solid (96%). MS (ESI): m/z=268.1 [M+H]+.
  • Step a) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)thio)azetidine-1-carboxylate
  • In a 20 mL glastube, a solution of 2-chloro-4-(trifluoromethyl)benzenethiol (440 mg, 2.07 mmol) in dry THF (6 mL) was added potassium tert-butoxide IM solution in THF (2.17 ml, 2.17 mmol) and the yellow reaction mixture was stirred at RT for 15 min followed by addition of tert-butyl 3-bromoazetidine-1-carboxylate (489 mg, 2.07 mmol). The reaction mixture was then stirred at RT for 5 h and over night at 70° C. The crude reaction was diluted with EtOAc and extracted with H2O, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over Na2SO4 and evaporated down to dryness. The residue was purified by chromatography (SiO2, n-eptane/EtOAc (0 to 40% over 40 min) yielded the product as a viscous oil (467 mg, 61%). MS (ESI): m/z=312.1 [M−56]+.
  • BB99 3-((2-Chloro-4-(trifluoromethyl)phenyl)sulfonyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)sulfonyl)azetidine-1-carboxylate (100 mg, 250 μmol) was dissolved in DCM and TFA (228 mg, 154 μL, 2 mmol) was added. The reaction mixture was stirred at RT for 8 h. Volatiles were removed in vacuo to yield the desired compound as light yellow solid (102 mg, 98%). MS (ESI): m/z=300.0 [M+H]+.
  • Step a) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)thio)azetidine-1-carboxylate
  • In a 20 mL glastube, a solution of 2-chloro-4-(trifluoromethyl)benzenethiol (440 mg, 2.07 mmol) in dry THF (6 mL) was added potassium tert-butoxide 1M solution in THF (2.17 mL, 2.17 mmol) and the yellow reaction mixture was stirred at r.t for 15 min followed by addition of tert-butyl 3-bromoazetidine-1-carboxylate (489 mg, 2.07 mmol). The reaction mixture was then stirred at r.t for 5 h and over night at 70° C. The crude reaction was diluted with EtOAc and extracted with H2O, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over Na2SO4 and evaporated down to dryness. The residue was purified by column chromatography (SiO2, n-eptane/EtOAc (0 to 40% over 40 min) to yield the desired product as a viscous oil (467 mg, 61%). MS (ESI): m/z=312.1 [M−56+H]+.
  • Step b) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)sulfonyl)azetidine-1-carboxylate
  • mCPBA (347 mg, 1.41 mmol) was added in one portion to a stirred solution of tert-butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)thio)azetidine-1-carboxylate (345 mg, 938 μmol) in DCM (6 mL) in an ice bath. The reaction was stirred at RT for 20 min. The reaction mixture was poured into 5 mL saturated Na2CO3 solution and extracted twice with DCM (20 mL each). The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude material was purified by preparative HPLC (YMC-Triart C 18, ACN/H2O+0.1% HCOOH) to furnish the product as a white powder (253 mg, 67.5%) MS (ESI): m/z=344.0 [M−56+H]+.
  • BB100 3-((2-Chloro-4-(trifluoromethyl)phenyl)sulfinyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)sulfinyl)azetidine-1-carboxylate (50 mg, 130 μmol) was dissolved in DCM (1.5 mL), TFA (149 mg, 100 μL, 1.3 mmol) was added and the reaction mixture was stirred at RT for 8 h. Volatiles were removed in vacuo to yield the compound as white solid (51 mg, 98%). MS (ESI): m/z=284.1 [M+H]+.
  • Step a) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)thio)azetidine-1-carboxylate
  • The compound was prepared in analogy to BB99, step a, and used in the next step without further purification.
  • Step b) tert-Butyl 3-((2-chloro-4-(trifluoromethyl)phenyl)sulfinyl)azetidine-1-carboxylate
  • The sulfoxide intermediate was isolated from the synthesis of the according sulfone building block BB99, step b. The product was obtained as a white lyophilized powder (50 mg, 13.9%) MS (ESI): m/z=328.1 [M−56+H]+.
  • BB101 3-(((2-Chloro-4-(trifluoromethyl)phenyl)thio)methyl)azetidine 2,2,2-trifluoroacetate
  • To a solution of tert-butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)thio)methyl)azetidine-1-carboxylate (0.200 g, 524 μmol) in DCM (3 mL) was added TFA (478 mg, 323 μL, 4.19 mmol) and the reaction mixture was stirred at RT for 3 h. Volatiles were removed in vacuo to yield the compound as light yellow oil that was used in the next step without further purification (267 mg). MS (ESI): m/z=282.2 [M+H]+.
  • Step a) tert-Butyl 3-[[2-chloro-4-(trifluoromethyl)phenyl]sulfanylmethyl]azetidine-1-carboxylate
  • To a solution of 2-chloro-4-(trifluoromethyl)benzenethiol (0.400 g, 1.88 mmol) in dry THF (6 mL) was added potassium tert-butoxide IM solution in THF (1.98 mL, 1.98 mmol) and the turbid reaction mixture was stirred at RT for 15 min followed by addition of tert-butyl 3-(bromomethyl)azetidine-1-carboxylate (471 mg, 1.88 mmol). The reaction mixture was then stirred at RT for 19 h. The crude reaction was diluted with EtOAc and extracted with aq. 1 M NaHCO3 solution, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over Na2SO4 and evaporated down to dryness to yield the crude product which was used in the next step without further purification (762 mg). MS (ESI): m/z=326.1 [M-56+H].
  • BB102 3-((2-Fluoro-6-(trifluoromethyl)benzyl)sulfonyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-((2-fluoro-6-(trifluoromethyl)phenyl)methylsulfonyl)azetidine-1-carboxylate (0.047 g, 118 μmol) was dissolved in DCM (0.5 mL) and TFA (108 mg, 72.9 μL, 946 μmol) was added. The reaction mixture was stirred at RT for 2 h. Volatiles were removed in vacuo to yield the compound as a yellow oil (56 mg) that was used in the next step without further purification. MS (ESI): m/z=298.2 [M+H]+.
  • step a) tert-Butyl 3-((2-fluoro-6-(trifluoromethyl)benzyl)thio)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-mercaptoazetidine-1-carboxylate (0.400 g, 2.11 mmol) in dry THF (5 mL) was added potassium tert-butoxide 1M solution in THF (2.22 mL, 2.22 mmol) and the reaction mixture was stirred at RT for 15 min followed by addition of 2-(bromomethyl)-1-fluoro-3-(trifluoromethyl)benzene (CAS RN 239087-08-2). The reaction mixture was then stirred at RT for 14 h. The crude reaction was diluted with EtOAc and extracted with aq. 1 M NaHCO3 solution, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over NaSO4 and evaporated down to dryness to yield the crude product (805 mg) which was used in the next step without further purification. MS (ESI): m/z=310.2 [M−56+H]+.
  • Step b) tert-Butyl 3-[[2-fluoro-6-(trifluoromethyl)phenyl]methylsulfonyl]azetidine-1-carboxylate
  • 3-Chlorobenzoperoxoic acid (283 mg, 1.64 mmol) was added in portion to a stirred solution of tert-butyl 3-((2-fluoro-6-(trifluoromethyl)benzyl)thio)azetidine-1-carboxylate (0.300 g, 821 μmol) in DCM (5 mL) in an ice bath. The reaction mixture was stirred at RT for 15 min and poured into 5 mL saturated aqueous NaHCO3 solution and extracted twice with DCM (10 mL each). The organic layers were combined, washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude material was purified by flash column chromatography (silica gel, 20 g, 0% to 100% EtOAc in n-heptane) to furnish the desired product as a colorless oil (47 mg, 15%). MS (ESI): m/z=415.1 [M+NH4]+.
  • BB103 3-((2-Fluoro-6-(trifluoromethyl)benzyl)sulfinyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-[[2-fluoro-6-(trifluoromethyl)phenyl]methylsulfinyl]azetidine-1-carboxylate (0.086 g, 225 μmol) was dissolved in DCM (1 mL) and TFA (206 mg, 139 μL, 1.8 mmol) was added. The reaction mixture was stirred at RT for 2 h. Volatiles were removed in vacuo to yield the compound as a yellow oil (93 mg) that was used in the next step without further purification. MS (ESI): m/z=282.2 [M+H]+.
  • Step a) tert-Butyl 3-[[2-fluoro-6-(trifluoromethyl)phenyl]methylsulfinyl]azetidine-1-carboxylate
  • The sulfoxide intermediate was isolated from the synthesis of BB 102, step b, as a colorless oil (86 mg, 28%). MS (ESI): m/z=404.1 [M+Na]+.
  • BB104 3-(((2-Chloro-4-(trifluoromethyl)phenyl)sulfonyl)methyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)sulfonyl)methyl)azetidine-1-carboxylate (0.145 g, 350 μmol) was dissolved in DCM (2 mL) and TFA (320 mg, 216 μL, 2.8 mmol) was added. The reaction mixture was stirred at RT for 2 h. Volatiles were removed in vacuo to yield the compound as a yellow oil (181 mg) that was used in the next step without further purification. MS (ESI): m/z=314.1 [M+H]+.
  • Step a) tert-Butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)sulfonyl)methyl)azetidine-1-carboxylate
  • 3-Chlorobenzoperoxoic acid (352 mg, 1.57 mmol) was added in portions to a stirred solution of tert-butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)thio)methyl)azetidine-1-carboxylate (BB 101, step a) (0.300 g, 786 μmol) in DCM (5 mL) in an ice bath. The reaction mixture was stirred at RT for 15 min and poured into 5 mL saturated aqueous NaHCO3 solution and extracted twice with DCM (10 mL each). The organic layers were combined, washed with brine, dried over NaSO4 and concentrated in vacuo. The crude material was purified by flash chromatography (silica gel, 20 g, 0% to 100% EtOAc in n-heptane) to provide the desired product as a colorless oil (145 mg, 45%). MS (ESI): m/z=314.0 [M−56+H]+.
  • BB105 3-(((2-Chloro-4-(trifluoromethyl)phenyl)sulfinyl)methyl)azetidine 2,2,2-trifluoroacetate
  • tert-Butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)sulfinyl)methyl)azetidine-1-carboxylate (0.086 g, 216 μmol) was dissolved in DCM (1 mL) and TFA (197 mg, 133 μL, 1.73 mmol) was added. The reaction mixture was stirred at RT for 2 h. Volatiles were removed in vacuo to yield the compound as a yellow oil (99 mg) that was used in the next step without further purification. MS (ESI): m/z=298.1 [M+H]+.
  • Step a) tert-Butyl 3-(((2-chloro-4-(trifluoromethyl)phenyl)sulfinyl)methyl)azetidine-1-carboxylate
  • The sulfoxide intermediate was isolated from the synthesis of BB104, step a. The desired product was obtained as a yellow oil (80 mg, 25.6%). MS (ESI): m/z=398.1 [M+H]+.
  • BB106 3-((2-Fluoro-4-(trifluoromethyl)benzyl)thio)azetidine 2,2,2-trifluoroacetate
  • To a solution of tert-butyl 3-((2-fluoro-4-(trifluoromethyl)benzyl)thio)azetidine-1-carboxylate (0.282 g, 772 μmol) in DCM (3 mL) was added TFA (880 mg, 595 μL, 7.72 mmol) and the reaction mixture was stirred at RT for 3 h. Volatiles were removed in vacuo to yield the desired compound as a colorless oil (302 mg) that was used in the next step without further purification. MS (ESI): m/z=266.2 [M+H]+.
  • Step a) tert-Butyl 3-[[2-fluoro-4-(trifluoromethyl)phenyl]methylsulfonyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-mercaptoazetidine-1-carboxylate (0.200 g, 1.06 mmol) in dry THF (2 mL) was added potassium tert-butoxide 1M solution in THF (1.11 mL, 1.11 mmol) and the reaction mixture was stirred at RT for 15 min followed by addition of 1-(bromomethyl)-2-fluoro-4-(trifluoromethyl)benzene (272 mg, 1.06 mmol, CAS RN 239087-07-1). The reaction mixture was then stirred at RT for 14 h. The crude reaction was diluted with EtOAc and extracted with aq. 1 M NaHCO3 solution, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over NaSO4 and evaporated down to dryness and purified by flash chromatography (silica gel, 20 g, 0% to 80% EtOAc in n-heptane) to furnish the desired product as a colorless oil (288 mg, 75%). MS (ESI): m/z=310.2 [M−56+H]+.
  • In analogy to BB84, the following intermediates were prepared from the respective commercially available starting materials.
  • BB No. Systematic Name Starting material MS, m/z
    BB107 3-[2-(2,6- 1,3-Dichloro-2-iodobenzene 226.1
    Dichlorophenyl)ethynyl]azetidine [M + H]+
    BB108 3-[2-[2-Fluoro-4- 1-Bromo-2-fluoro-4- 244.2
    (trifluoromethyl)phenyl]ethynyl]azetidine (trifluoromethyl)benzene [M + H]+
    BB109 3-[2-(2,6- 1,3-Difluoro-2-iodobenzene 194.2
    Difluorophenyl)ethynyl]azetidine [M + H]+
    BB110 3-[2-[3-Chloro-4- 4-Bromo-2-chloro-1- 260.2
    (trifluoromethyl)phenyl]ethynyl]azetidine (trifluoromethyl)benzene [M + H]+
    BB111 3-[2-(2-Chloro-6-fluoro- 2-Bromo-1-chloro-3- 210.1
    phenyl)ethynyl]azetidine fluorobenzene [M + H]+
    BB112 3-[2-(2-Chloro-4-cyclopropyl- 1-Bromo-2-chloro-4- 232.2
    phenyl)ethynyl]azetidine cyclopropylbenzene [M + H]+
    BB113 3-[2-(2- 1-Bromo-2-methoxybenzene 188.2
    Methoxyphenyl)ethynyl]azetidine [M + H]+
    BB114 3-[2-[4-Chloro-2- 4-Chloro-1-iodo-2- 260.1
    (trifluoromethyl)phenyl]ethynyl]azetidine (trifluoromethyl)benzene [M + H]+
    BB115 3-[2-(3-Chlorophenyl)ethynyl]azetidine 1-Bromo-3-chlorobenzene 192.1
    [M + H]+
    BB116 3-[2-[4- 1-bromo-4- 242.2
    (Trifluoromethoxy)phenyl]ethynyl]azetidine (Trifluoromethoxy)benzene [M + H]+
    BB117 3-[2-[4- 1-Bromo-4- 226.2
    (Trifluoromethoxy)phenyl]ethynyl]azetidine (trifluoromethyl)benzene [M + H]+
    BB118 3-[2-(3-Fluoro-2-methyl- 1-Bromo-3-fluoro-2- 190.2
    phenyl)ethynyl]azetidine methylbenzene [M + H]+
    BB119 3-[2-(2,6- 2-Iodo-1,3-dimethylbenzene 186.2
    Dimethylphenyl)ethynyl]azetidine [M + H]+
    BB120 3-[2-[2- 1-Bromo-2- 242.2
    (Trifluoromethoxy)phenyl]ethynyl]azetidine (trifluoromethoxy)benzene [M + H]+
    BB121 3-[2-(2-Bromophenyl)ethynyl]azetidine 1-Bromo-2-iodobenzene 236.1
    [M + H]+
    BB122 3-[2-(2-Chloro-3-fluoro- 1-Bromo-2-chloro-3- 210.1
    phenyl]ethynyl]azetidine fluorobenzene [M + H]+
    BB123 3-[2-(o-Tolyl)ethynyl]azetidine 1-Bromo-2-methylbenzene 172.2
    [M + H]+
    BB124 3-[2-(4-Chloro-2-fluoro- 4-Chloro-2-fluoro-1- 210.1
    phenyl)ethynyl]azetidine iodobenzene [M + H]+
    BB125 3-[2-[2- 1-(Difluoromethoxy)-2- 224.2
    (Difluorormethoxy)phenyl]ethynyl]azetidine iodobenzene [M + H]+
    BB126 2-[2-(Azetidin-3-yl)ethynyl]-3-chloro- 2-Bromo-3- 217.2
    benzonitrile chlorobenzonitrile [M + H]+
    BB127 3-[2-[4- 1-(Difluoromethoxy)-4- 224.2
    (Difluorormethoxy)phenyl]ethynyl]azetidine iodobenzene [M + H]+
    BB128 1-[4-[2-(Azetidin-3- 1-(4- 223.2
    yl)ethynyl]phenyl]cyclopropartecarbonitrile Bromophenyl)cyclopropane- [M + H]+
    1-carbonitrile
    BB129 3-[2-(4-Cyclopropylphenyl)prop-1- 1-Bromo-4-cyclopropyl- 198.2
    ynyl]lazetidine benzene [M + H]+
    BB130 1-[4-[2-(Azetidin-3- 1-(4- 214.2
    yl)ethynyl]phenyl]cyclopropanol Bromophenyl]cyclopropanol [M + H]+
    BB131 3[2-(3- 1-Iodo-3-methoxybenzene 188.2
    Methoxyphenyl)ethynyl]azetidine [M + H]+
    BB132 3-[2-[2- 1-Bromo-2- 208.2
    (Difluoromethyl)phenyl]ethynyl]azetidine (difluoromethyl)benzene [M + H]+
    BB133 3-[2-[3-Methoxy-2-methyl- 1-Iodo-3-methoxy-2- 202.2
    phenyl)ethynyl]azetidine methylbenzene [M + H]+
    BB134 3-[2-[2-Chloro-6-methyl- 1-Chloro-2-iodo-3- 206.1
    phenyl)ethynyl]azetidine methylbenzene [M + H]+
    BB135 3-[2-(2-Chloro-5-fluoro- 2-Bromo-1-chloro-4- 210.1
    phenyl)ethynyl]azetidine fluorobenzene [M + H]+
    BB136 3-[2-(4- 1-Bromo-4-methylsulfonyl- 236.2
    Methylsulfonylphenyl)ethynyl]azetidine benzene [M + H]+
    BB137 3-[2-(5-Chloro-2- 2-Bromo-5-chlorothiophene 198.1
    thienyl)ethynyl]azetidine [M + H]+
    BB138 3-[2-(5-Chloro-3- 4-Bromo-2-chlorothiophene 198.1
    thienyl)ethynyl]azetidine [M + H]+
    BB139 3-[2-[2-Chloro-6-fluoro-4- 2-Bromo-1-chloro-3-fluoro- 278.1
    (trifluoromethyl)phenyl]ethynyl]azetidine 5-(trifluoromethyl)benzene [M + H]+
    BB140 3[2-(2-Chlorophenyl)ethynyl]azetidin-3-ol Chloro-2-iodobenzene 208.1
    and [M + H]+
    tert-butyl 3-ethynyl-3-
    hydroxyazetidine-1-
    carboxylate
    (CAS
    RN 1259034-35-9)
    BB141 3-[2-[2- 1-Iodo-2- 202.2
    (Methoxymethyl)phenyl]ethynyl]lazetidine (methoxymethyl)benzene [M + H]+
    BB142 3-[2-[2-Chloro-4- 2-Chloro-1-iodo-4- 260.2
    (trifluoromethyl)phenyl]ethynyl]azetidine (tritluoromethyl)benzene [M + H]+
  • In analogy to BB18, the following intermediates were prepared from the respective commercially available starting materials.
  • BB No. Systematic Name Starting material MS, m/z
    BB143 4-[2-[2- 1-Bromo-2- 254.3
    (Trifluoromethyl)phenyl]ethynyl]piperidine (trifluoromethyl)benzene [M + H]+
    BB144 4-[2-(2- 1-Bromo-2-methoxybenzene 216.3
    Methoxyphenyl)ethynyl]piperidine [M + H]+
    BB145 4-[2-(o-Tolyl)ethynyl]piperidine 1-Bromo-2-methylbenzene 200.3
    [M + H]+
    BB146 4-[2-(2,6- 2-Iodo-1,3-dimethytbenzene 214.3
    Dimethylphenyl)ethynyl]piperidine [M + H]+
    BB147 4-[2-(2,4-Dichlorophenyl)ethynyl]-4- Bromo-2,4-dichlorobenzene 268.2
    methyl-piperidine and tert-Butyl 4-ethynyl-4- [M + H]+
    methylpiperidine-
    1-carboxylate
    (CAS RN 1363383-17-8)
    BB148 4-[2-(2-Chloro-4-fluoro- 2-Chloro-4-fluoro-1- 252.2
    phenyl)ethynyl]-4-methyl-piperidine iodobenzene and [M + H]+
    tert-Butyl 4-ethynyl-4-
    methylpiperidine-
    1-carboxylate
    (CAS RN 1363383-17-8)
  • BB149 1-[2-(Azetidin-3-yl)ethynyl]cyclopentanol hydrochloride
  • To a solution of tert-butyl 3-[2-(1-hydroxycyclopentyl)ethynyl]azetidine-1-carboxylate (0.02 g, 0.075 mmol) in dioxane (0.5 mL) was added 4 M HCl in dioxane (0.094 mL, 0.377 mmol) and the reaction mixture was stirred at RT for 18 h. The mixture was evaporated to dryness and the residue triturated in diisopropyl ether, filtered off and further dried under high vacuum to give the title compound as a white solid as the hydrochloride salt (0.013 g, 87%). MS (ESI): m/z=166.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-(1-hydroxycyclopentyl)ethynyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (0.2 g, 1.1 mmol) in THF (6.5 mL) at −78° C. was added nBuLi (0.759 mL, 1.21 mmol) dropwise and the reaction mixture was stirred at this temperature for 1 h. Then, cyclopentanone (0.107 mL, 1.21 mmol) in THF (3 mL) was added dropwise to the mixture which was stirred at −78° C. for 2 h. The mixture was allowed to warm up to 0° C., poured into a sat. NH4OH aqueous solution and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and evaporated. The residue was purified by silica gel flash chromatography, eluting with a gradient of EtOAc:n-heptane (0 to 100%) to yield the title compound as a light yellow oil (0.020 g, 7%). MS (ESI): m/z=192.2 [M−56−18+H]+.
  • BB150 4-[3-Pyrazol-1-yl-5-(trifluoromethyl)phenoxy]piperidine formate
  • A mixture of tert-butyl 4-[3-pyrazol-1-yl-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (400.0 mg, 0.970 mmol) and TFA (1.0 mL, 0.970 mmol) in DCM (10 mL) was stirred at 20° C. for 12 h. The mixture was purified by prep-HPLC (ACN and water containing 0.225% v/v FA) to give the desired product (300 mg, 94.4%) as colorless gum. MS (ESI): m/z=312.1 [M+H]+.
  • Step a: tert-Butyl 4-[3-bromo-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • To a solution of 3-bromo-5-(trifluoromethyl)phenol (2.0 g, 8.3 mmol), 1-BOC-4-hydroxypiperidine (1.84 g, 9.13 mmol, CAS RN 106-52-5) and PPh3 (2.61 g, 9.96 mmol) in THF (32.6 mL) was added diisopropyl azodicarboxylate (1.96 mL, 9.96 mmol) and the mixture was stirred at 20° C. for 15 h. The mixture was concentrated under vacuum. The residue was purified by prep-HPLC (ACN and water containing 0.225% v/v FA) and concentrated under vacuum to give the desired product (2.6 g, 73.9% yield) as light yellow oil. MS (ESI): m/z=367.9 [M−56+H]+.
  • Step b) tert-Butyl 4-[3-pyrazol-1-yl-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[3-bromo-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (500.0 mg, 1.18 mmol), pyrazole (160.47 mg, 2.36 mmol), CuI (22.37 mg, 0.120 mmol), cesium carbonate (1152 mg, 3.54 mmol) and N,N′-dimethylethylenediamine (519.15 mg, 5.89 mmol) in DMF (5 mL) was stirred at 110° C. for 12 h. The mixture was poured into H2O water (30 mL) and extracted three times with EtOAc (50 mL). The combined organic layer was washed with ammonia (10 mL), brine (50 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum to give the desired product (400 mg, 82.5% yield) as light yellow oil. MS (ESI): m/z=356.2 [M−56+H]+.
  • BB51 4-[[2-(2,2,2-Trifluoroethoxy)-4-(trifluoromethyl)phenyl]methyl]piperidine
  • A mixture of 4-[[2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)phenyl]methylene]piperidine (250.0 mg, 0.740 mmol) and Pd/C (50.0 mg, wt. 10%) in THF (10 mL) was stirred at 20° C. for 12 h under H2 (1520 mmHg). The mixture was filtered and concentrated under vacuum to give the desired compound (240 mg, 95.4%) as light brown gum. MS (ESI): m/z=342.1 [M+H]+.
  • Step a) tert-Butyl 4-(p-tolylsulfonylhydrazone)piperidine-1-carboxylate
  • To a solution of 4-methylbenzenesulfonhydrazide (9.35 g, 50.19 mmol, CAS RN 1576-35-8) in MeOH (100 mL) was added 1-BOC-4-piperidone (10.0 g, 50.19 mmol, CAS RN 17502-28-8) and the mixture was stirred at 25° C. for 12 h. The mixture was concentrated to give the desired product (18.4 g. 99.8%) as off-white solid. MS (ESI): m/z=368.2 [M+H]+.
  • Step b) 2-(2,2,2-Trifluoroethoxy)-4-(trifluoromethyl)benzaldehyde
  • A mixture of NaH (187.39 mg, 60% dispersion in mineral oil, 4.68 mmol) in 2,2,2-trifluoroethanol (16.67 mL, 228.74 mmol, CAS RN75-89-8) was stirred at 0° C. The cooling bath was removed and the mixture was stirred at 20° C. for 2 h, and then 2-fluoro-4-(trifluoromethyl)benzaldehyde (1.0 g, 5.21 mmol, CAS RN 763-93-9) was added and the mixture was stirred at 20° C. for 12 h. The mixture was poured into H2O (30 mL) and extracted twice with EtOAc (30 mL each). The combined organic layers were washed with brine (30 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum to give the desired product (1.2 g, 84.7%) as light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.44-10.34 (m, 1H), 7.93 (d, J=8.1 Hz, 1H), 7.75 (s, 1H), 7.55 (d, J=8.1 Hz, 1H), 5.11 (q, J=8.7 Hz, 2H).
  • Step c) tert-Butyl 4-[2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)benzoyl]piperidine-1-carboxylate
  • A mixture of 2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)benzaldehyde (1000.0 mg, 3.67 mmol), tert-butyl 4-(p-tolylsulfonylhydrazone)piperidine-1-carboxylate (1350.3 mg, 3.67 mmol) and cesium carbonate (1795.9 mg, 5.51 mmol) in 1,4-dioxane (30 mL) was stirred at 110° C. for 12 h under N2 atmosphere. The mixture was poured into H2O (50 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine (50 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and the residue was purified by prep-HPLC (MeCN and water containing 0.225% v/v FA) to give the desired product (980 mg, 58.6%) as light yellow gum. MS (ESI): m/z=400.1 [M−56+H]+.
  • Step d) tert-Butyl 4-[hydroxy-[2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)benzoyl]piperidine-1-carboxylate (900.0 mg, 1.98 mmol) in MeOH (45 mL) was added NaBH4 (149.54 mg, 3.95 mmol) at 0° C. and the mixture was stirred at 20° C. for 12 h. The mixture was purified by prep-HPLC (MeCN and water containing 0.225% v/v FA) (650 mg, 71.9%) as light yellow oil. MS (ESI): m/z=384.0 [M−56-OH+H]+.
  • Step e) 4-[[2-(2,2,2-Trifluoroethoxy)-4-(trifluoromethyl)phenyl]methylene]piperidine
  • A mixture of tert-butyl 4-[hydroxy-[2-(2,2,2-trifluoroethoxy)-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate (400.0 mg, 0.870 mmol) and MsOH (840.43 mg, 8.74 mmol) in DCM (4 mL) was stirred at 40° C. for 24 h. The mixture was poured into saturated aqueous Na2CO3 solution (5 mL) and extracted three times with EtOAc (10 mL each). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum to give the desired compound as light yellow oil (260 mg, 76.2%). MS (ESI): m/z=340.1 [M+H]+.
  • BB152 4-[3-(1,2,4-Triazol-1-yl)-5-(trifluoromethyl)phenoxyl]piperidine trifluoroacetate
  • To a mixture of tert-butyl 4-[3-(1,2,4-triazol-1-yl)-5-(trifluoromethyl)phenoxypiperidine-1-carboxylate (240.0 mg, 0.580 mmol) in DCM (10 mL) was added TFA (1.0 mL). The mixture was stirred at 20° C. for 12 h and then concentrated under vacuum to give 4-13-(1,2,4-triazol-1-yl)-5-(trifluoromethyl)phenoxy]piperidine 2,2,2-trifluoroacetic acid salt (240 mg, 96.7%) as light yellow gum. MS (ESI): m/z=313.1 [M+H]+.
  • Step a) tert-Butyl 4-[3-(1,2,4-triazol-1-yl)-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[3-bromo-5-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (500.0 mg, 1.18 mmol, BB98, intermediate a). 1,2,4-triazole (162.8 mg, 2.36 mmol) and CuI (22.37 mg, 0.120 mmol) in DMF (5 mL) was stirred at 110° C. for 12 h. The mixture was poured into H2O (20 mL) and extracted three times with EtOAc (30 mL each). The combined organic layers were washed with ammonia (20 mL), brine (20 mL, three times), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and the residue was purified by column chromatography (PE:EA=50:1-3:1) to give the desired product (240 mg, 49.4%) as light yellow solid. MS (ESI): m/z=357.1 [M−56+H]+.
  • BB153 3-[4-Chloro-3-(trifluoromethyl)phenoxy]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[4-chloro-3-(trifluoromethyl)phenoxy]azetidine-1-carboxylate (300.0 mg, 0.530 mmol) in DCM (7.5 mL) was added TFA (1.04 mL) at 0° C. and the mixture was stirred at 20° C. for 2 h. The mixture was concentrated to give the title compound as yellow oil (280 mg, 97%). MS (ESI): m/z=252.0 [M+H]+.
  • Step a) tert-Butyl 3-[4-chloro-3-(trifluoromethyl)phenoxy]azetidine-1-carboxylate
  • To a solution of 2-chloro-5-hydroxybenzotrifluoride (1 g, 5.1 mmol CAS RN 6294-93-5), ten-butyl 3-hydroxyazetidine-1-carboxylate (0.97 g, 5.6 mmol CAS RN 141699-55-0) and triphenylphosphine (1.6 g, 6.11 mmol) in THF (20 mL) was added diisopropyl azodicarboxylate (1.2 mL, 6.11 mmol) and the mixture was stirred at 20° C. for 15 h. The mixture was concentrated and purified by reversed phase chromatography (MeCN and water containing 0.225% v/v FA) to give the title compound (820 mg, 28.7%) as brown solid. MS (ESI): m/z=295.9 [M−56+H]+.
  • BB154 4-(4-Chloro-3-pyrazol-1-yl-phenoxy)piperidine trifluoroacetate
  • To a solution of tert-butyl 4-(4-chloro-3-pyrazol-1-yl-phenoxy)piperidine-1-carboxylate (260.0 mg, 0.690 mmol) in DCM (5.38 mL) was added TFA (1.34 mL, 17.46 mmol) at 0° C. and the mixture was stirred at 20° C. for 1 h. The mixture was concentrated to give the title compound as an orange oil (250 mg, 92.7 yield). MS (ESI): m/z=278.1 [M+H]+.
  • Step a) tert-Butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate
  • To a solution of 1-BOC-4-hydroxypiperidine (2.04 g, 10.12 mmol, CAS RN 106-52-5), 3-bromo-4-chlorophenol (2.0 g, 9.64 mmol, CAS RN 2402-82-6) and triphenylphosphine (3.03 g, 11.57 mmol) in THF (50 mL) was added diisopropyl azodicarboxylate (2.28 mL, 11.57 mmol) and the mixture was stirred at 20° C. for 15 h. Then the mixture was concentrated and the residue was purified by reversed flash chromatography (MeCN and water containing 0.1% v/v FA) to give the desired product (2.8 g, 74.3%) as light yellow oil. MS (ESI): m/z=335.9 [M−56+H]+.
  • Step b) tert-Butyl 4-(4-chloro-3-pyrazol-1-yl-phenoxy)piperidine-1-carboxylate
  • To a mixture of tert-butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate (1.0 g, 2.56 mmol), pyrazole (139.4 mg, 2.05 mmol), cesium carbonate (2501.8 mg, 7.68 mmol) and 1,10-phenanthroline (225.49 mg, 2.56 mmol) in DMF (20 mL) was added CuI (48.59 mg, 0.260 mmol) and the mixture was stirred at 110° C. for 12 h under N2 atmosphere. The mixture was concentrated, diluted with H2O (20 mL) and extracted three times with EtOAc (10 mL). The combined organic layers were concentrated and the residue purified by reversed phase chromatography (ACN and water containing 0.1% v/v FA) to give the desired product (265 mg, 22.5%, 82% purity) as yellow oil. MS (ESI): m/z=378.1 [M+H]+.
  • BB55 4-[5-(4-Piperidyloxy)-2-(trifluoromethyl)phenyl]morpholine trifluoroacetate
  • To a solution of tert-butyl 4-[3-morpholino-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (400.0 mg, 0.93 mmol) in DCM (3 mL) was added TFA (1.0 mL) and the reaction mixture was stirred at 25° C. for 12 h. The reaction was concentrated in vacuum to provide the crude product (300 mg) as yellow oil, which was used in next step without further purification. MS (ESI): m/z=331.2 [M+H]+.
  • Step a) tert-Butyl 4-(3-bromo-4-(trifluoromethyl)phenoxy)piperidine-1-carboxylate
  • To a solution of 3-bromo-4-(trifluoromethyl)phenol (500.0 mg, 2.54 mmol, CAS RN1214385-56-4) and 1-BOC-4-hydroxypiperidine (512 mg, 2.54 mmol, CAS RN 106-52-5) in THF (8.5 mL) was added PPh3 (1000.9 mg, 3.82 mmol) and diethyl azodicarboxylate (664.53 mg, 3.82 mmol) and the mixture was stirred at 25° C. for 12 h. The mixture was purified by silica gel chromatography using PE:EA=5:1 as eluant to provide the desired product (503 mg, 46.6% yield) as light yellow oil. MS (ESI): m/z=369.2 [M−56+H]+.
  • Step b) tert-Butyl 4-(3-morpholino-4-(trifluoromethyl)phenoxy)piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[3-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (500.0 mg, 1.18 mmol), morpholine (154 mg, 1.77 mmol, CAS RN 110-91-8), (R)-(+)-2,2′-bis(diphenylphosphino)-1,1′-binaphthalene (146.77 mg, 0.24 mmol, CAS RN 76189-55-4), cesium carbonate (1.15 g, 3.54 mmol) and tris(dibenzylideneacetone)dipalladium(0) (172.47 mg, 0.240 mmol, CAS RN 76971-72-7) in DMF (10 mL) was stirred at 110° C. for 12 h. The mixture was poured into H2O and extracted three times with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and the residue was purified by column chromatography (gradient of EtOAc in PE 5% to 33%) to give the desired product (480 mg, 94.6%) as light yellow solid. MS (ESI): m/z=431.1 [M+H]+.
  • BB156 4-(4-Chloro-3-(1,2,4-triazol-1-yl)phenoxy)piperidine trifluoroacetate
  • To a solution of tert-butyl 4-[4-chloro-3-(1,2,4-triazol-1-yl)phenoxy]piperidine-1-carboxylate (196.0 mg, 0.520 mmol) in DCM (5 mL) was added TFA (1.01 mL, 13.13 mmol) at 0° C. and the mixture was stirred at 20° C. for 1 h. The mixture was concentrated to give the title compound (178 mg, 87.6%) as brown oil. MS (ESI): m/z=279.1 [M+H]+.
  • Step a) tert-Butyl 4-[4-chloro-3-(1,2,4-triazol-1-yl)phenoxy]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-(3-bromo-4-chloro-phenoxy)piperidine-1-carboxylate (500.0 mg, 1.28 mmol. BB102, intermediate a). 1,2,4-triazole (176.8 mg, 2.56 mmol), CuI (24.3 mg, 0.130 mmol) and cesium carbonate (1250.9 mg, 3.84 mmol) and dimethyl glycine (1.0 mL, 1.28 mmol) in DMF (10 mL) was stirred at 120° C. for 12 h. The mixture was concentrated to remove the DMF, diluted with H2O (50 mL) and extracted three times with EtOAc (20 mL each). The combined organic layers were evaporated and the residue purified by reverse phase flash chromatography (ACN and water containing 0.1% v/v FA) to give the title compound (196 mg, 37.1%) as colorless oil. MS (EST): m/z=323.0 [M−56+H]+.
  • BB157 4-[3-Cyclopropyl-4-(trifluoromethyl)phenoxy]piperidine trifluoroacetate
  • To a mixture of tert-butyl 4-[3-cyclopropyl-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (360.0 mg, 0.930 mmol) in DCM (18 mL) was added TFA (1.8 mL). The mixture was stirred at 25° C. for 12 h. The mixture was concentrated under vacuum to provide the desired compound as light yellow gum (370 mg, 99.2%). MS (ESI): m/z=286.2 [M+H]+.
  • Step a) tert-Butyl 4-[3-cyclopropyl-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[3-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (500.0 mg, 1.18 mmol, BB103, intermediate b), cyclopropylboronic acid (151.86 mg, 1.77 mmol), Na2CO3 (374.74 mg, 3.54 mmol) and Pd(PPh3)4 (13.6 mg, 0.010 mmol) in 1,4-dioxane (10 mL) and H2O (1 mL) was stirred at 95° C. for 12 h. The mixture was poured into H2O (50 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine (50 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and purified by column chromatography (PE:EtOAc=20:1-5:1) to give the desired product (380 mg, 83.7%) as colorless gum. MS (ESI): m/z=330.1 [M−56+H]+.
  • BB358 4-[3-Pyrazol-1-yl-4-(trifluoromethyl)phenoxy]piperidine trifluoroacetate
  • To a solution of tert-butyl 4-[3-pyrazol-1-yl-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (180.0 mg, 0.440 mmol) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at 25° C. for 12 h and then concentrated under vacuum to give the desired product (180 mg, 96.7%) as light yellow gum. MS (ESI): m/z=312.1 [M+H]+.
  • Step a) tert-Butyl 4-[3-pyrazol-1-yl-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate
  • A mixture of tert-butyl 4-[3-bromo-4-(trifluoromethyl)phenoxy]piperidine-1-carboxylate (500.0 mg, 1.18 mmol, BB103, intermediate b), pyrazole (120.35 mg, 1.77 mmol), CuI (22.37 mg, 0.120 mmol), N,N′-dimethylethylenediamine (519.45 mg, 5.89 mmol) and Cs2CO3 (767.99 mg, 2.36 mmol) in DMF (10 mL) was stirred at 110° C. for 12 h. The mixture was poured into H2O (30 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with ammonia (20 mL), brine (50 mL), dried over Na2SO4 and filtered. The filtrate was concentrated and the crude product was purified by prep-TLC (PE:EA=5:1) to give the desired product (190 mg, 39.2%) as colorless oil. MS (ESI): m/z=356.1 [M−56+H]+.
  • BB159 4-[[2,6-Difluoro-4-(trifluoromethyl)phenyl]methyl]piperidine trifluoroacetate
  • To a solution of tert-butyl 4-[[2,6-difluoro-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate (70.0 mg, 0.180 mmol) in DCM (1 mL) was added TFA (0.2 mL) and the mixture was stirred at 20° C. for 1 h. The mixture was concentrated to give the title compound (50 mg, 68.9%) as brown oil. MS (ESI): m/z=280.1 [M+H]+.
  • Step a) 2-(Diethoxyphosphorylmethyl)-1,3-difluoro-5-(trifluoromethyl)benzene
  • A solution of 2-(bromomethyl)-1,3-difluoro-5-(trifluoromethyl)benzene (1.29 mL, 3.27 mmol, CAS RN 493038-91-8) in triethyl phosphite (5.44 g, 32.73 mmol) was stirred at 160° C. for 5 h. The mixture was concentrated under vacuum to provide the title compound (600 mg, 55.2%, colorless oil) which was used in the next step without further purification.
  • Step b) tert-Butyl 4-[[2,6-difluoro-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate
  • A mixture of 2-(diethoxyphosphorylmethyl)-1,3-difluoro-5-(trifluoromethyl)benzene (400.0 mg, 1.2 mmol) in THF (4 mL) was added to sodium hydride (144.49 mg, 3.61 mmol) in THF (4 mL) at 0° C. The mixture was stirred at 0° C. for 1 h, and then 1-BOC-4-piperidone (479.83 mg, 2.41 mmol, CAS RN 79099-07-3) was added to the above mixture. The mixture was stirred at 20° C. for 12 h. The mixture was poured into H2O (50 mL) and extracted three times with EtOAc (20 mL each). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography (PE:EA=1:0 to 2:1) to give the title compound (100 mg, 22.0%) as colorless oil. MS (ESI): m/z=322.0 [M−56+H]+.
  • Step c) tert-Butyl 4-[[2,6-difluoro-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carboxylate
  • To a solution of tert-butyl 4-[[2,6-difluoro-4-(trifluoromethyl)phenyl]methylene]piperidine-1-carboxylate (100.0 mg, 0.270 mmol) in MeOH (8 mL) was added Pd/C (10.0 mg, wt. 10%). The mixture was stirred at 20° C. for 1 h under H2 atmosphere, then filtered and concentrated to give the title compound as colorless oil (70 mg, 69.6%). MS (ESI): m/z=324.1[M−56+H]+.
  • BB160 4-[4-Chloro-3-(4-chlorophenyl)-2-fluoro-phenoxyl]piperidine trifluoroacetate
  • To a mixture of tert-butyl 4-[4-chloro-3-(4-chlorophenyl)-2-fluoro-phenoxy]piperidine-1-carboxylate (145.0 mg, 0.330 mmol) in DCM (10 mL) was added TFA (1.0 mL). The mixture was stirred at 20° C. for 5 h. The mixture was concentrated under vacuum to give the desired product (149 mg, 99.6%) as light brown gum. MS (ESI): m/z=340.1 [M+H]+.
  • Step a) 1-Chloro-2-(4-chlorophenyl)-3-fluoro-4-methoxy-benzene
  • A mixture of 4-bromochlorobenzene (1.41 g, 7.34 mmol, CAS RN 106-39-8), (6-chloro-2-fluoro-3-methoxy-phenyl) boronic acid (1.0 g, 4.89 mmol, CAS RN 867333-04-8) and K2CO3 (2.03 g, 14.68 mmol) in 1,4-dioxane (15 mL) and H2O (1.5 mL) was stirred under N2 atmosphere at 110° C. for 1 h in a microwave oven. The mixture was poured into H2O (20 mL) and extracted three times with EtOAc (20 mL each). The combined organic layers were washed with brine (20 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and the residue was purified by column chromatography using PE as eluant to give the desired product (110 mg, 8.3%) as colorless oil which was used in the next step without further purification.
  • Step b) 4-chloro-3-(4-chlorophenyl)-2-fluoro-phenol
  • To a mixture of 1-chloro-2-(4-chlorophenyl)-3-fluoro-4-methoxy-benzene (215.0 mg, 0.790 mmol) in DCM (7 mL) was added a solution of BBr3 (993.36 mg, 3.97 mmol) in DCM (7 mL) drop wise at −78° C. The mixture was stirred at 20° C. for 12 h. The reaction was quenched by adding MeOH (1 mL) followed by water (10 mL), and the mixture was extracted three times with DCM (10 mL each). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum to give the desired product (120 mg, 57.5%) as light brown solid which was used in the next step without further purification.
  • Step c) tert-Butyl 4-[4-cloro-3-(4-chlorophenyl)-2-fluoro-phenoxy]piperidine-1-carboxylate
  • A mixture of 4-chloro-3-(4-chlorophenyl)-2-fluoro-phenol (120.0 mg, 0.470 mmol), 1-BOC-4-hydroxypiperidine (187.88 mg, 0.930 mmol, CAS RN 106-52-5). PPh3 (244.85 mg, 0.930 mmol) and DIAD (0.18 mL, 0.930 mmol) in THF (12 mL) was stirred at 20° C. for 12 h. The mixture was poured into H2O and extracted three times with EtOAc. The combined organic layer was washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated under vacuum and the residue purified by column chromatography (PE:EA=1:0˜20:1) to give the desired product as light yellow gum (150 mg, 73%). MS (ESI): m/z=384.0 [M−56+H]+.
  • BB161 3-[2-Chloro-4-(Trifluoromethyl)phenoxy]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-chloro-4-(trifluoromethyl)phenoxy]azetidine-1-carboxylate (400.0 mg, 1.14 mmol) in DCM (10 mL) was added TFA (2.0 mL) at 20° C. After stirring for 2 h the mixture was concentrated to give the crude product (410 mg, 98.6%) as light yellow oil which was used in the next step without further purification.
  • Step a) tert-Butyl 3-[2-chloro-4-(trifluoromethyl)phenoxy]azetidine-1-carboxylate
  • To a solution of 2-chloro-4-(trifluoromethyl)phenol (100(0.0 mg, 5.09 mmol, CAS RN 35852-58-5) and tert-butyl 3-hydroxyazetidine-1-carboxylate (1057.5 mg, 6.11 mmol, CAS RN 141699-55-0) in THF (20 mL) was added PPh3 (1999.49 mg, 7.63 mmol) and diethyl azodicarboxylate (1329.05 mg, 7.63 mmol), the mixture was stirred at 25° C. for 12 h. The reaction mixture solution was evaporated in vacuum, the residue was purified by reverse-phase flash (0.1% v/v FA) to afford the desired product (800 mg, 2.27 mmol, 44.7% yield) as light yellow oil. MS (ESI): m/z=296.0 [M−56+H]+.
  • BB162 3-((2-Fluoro-6-(trifluoromethyl)benzyl)oxy)azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[[2-fluoro-6-(trifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate (400.0 mg, 1.15 mmol) in dry DCM (10 mL) was added TFA (2.0 mL) at 25° C. and the mixture was stirred at 25° C. for 12 h. The solvent was stripped off and the residue was dried under vacuum to afford the desired compound as yellow oil (300 mg, 22%). MS (ESI): m/z=250.0 [M+H]+.
  • Step a) tert-Butyl 3-[[2-fluoro-6-(trifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate
  • To a solution of 2-fluoro-6-(trifluoromethyl)benzyl bromide (1000.0 mg, 3.89 mmol, CAS RN 239087-08-2) and tert-butyl 3-hydroxyazetidine-1-carboxylate (673.92 mg, 3.89 mmol, CAS RN 141699-55-0) in dry THF (10 mL) at 25° C. was added t-BuOK (5.84 mL, 5.84 mmol; 1.0 M in dry THF) and the mixture was stirred at 25° C. for 12 h. The mixture was poured into H2O (10 mL) and extracted three times with EA (20 mL each). The combined organic layers were combined, dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure, purified by flash chromatography on silica gel (gradient PE:EA=10:1 to 2:8) to give the title compound as colorless oil (1100 mg, 80.9%). MS (ESI): m/z=294.0 [M−56+H]+.
  • BB163 3-[2-(2-Fluoro-4-methyl-phenyl)ethyl]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethyl]azetidine-1-carboxylate (350.0 mg, 1.19 mmol) in dry DCM (10 mL) at 25° C., was added TFA (1.0 mL, 1.19 mmol) and the mixture was stirred at 25° C. for 12 h. The reaction mixture was concentrated by reduced pressure and the residue was dried in vacuum to provide the desired compound as colorless oil (260 mg, 70.9%). MS (ESI): m/z=194.0 [M+H]+.
  • Step a) tert-Butyl 3-(2-trimethylsilylethynyl)azetidine-1-carboxylate
  • To a solution of trimethylsilylacetylene (9.97 g, 101.55 mmol, CAS RN 1066-54-2) in dry THF (200 mL) at 25° C. was added i-PrMgCl (48.57 mL, 97.14 mmol; 1.0 M in dry THF) and the mixture was stirred at 25° C. for 15 mins. Then a solution of 1-BOC-3-iodoazetidine (25.0 g, 88.3 mmol, CAS RN 254454-54-1) was added followed by FeCl2 (0.34 g, 2.65 mmol) in dry DMF (606 mL) and the mixture was stirred at 25° C. for 12 hrs. The mixture was poured into saturated aq. NH4Cl solution (200 mL) and extracted three times with EtOAc (150 mL each). The organic layers were combined, dried with anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (PE:EA=20:1 to 10:1) to give the desired product as black oil (18 g, 80.4%). 1H NMR (400 MHz, CHLOROFORM-d) δ=4.11 (t, J=8.4 Hz, 2H), 3.92 (dd, J=6.5, 8.1 Hz, 2H), 3.51-3.17 (m, 1H), 1.44 (s, 10H), 0.16 (s, 9H).
  • Step b) tert-Butyl 3-ethynylazetidine-1-carboxylate
  • To a solution of tert-butyl 3-(2-trimethylsilylethynyl)azetidine-1-carboxylate (6243 mg, 24.64 mmol) in dry MeOH (40 mL) was added potassium carbonate (1700 mg, 12.32 mmol) at 25° C. and the reaction mixture was stirred at 25° C. for 2 h. The mixture was filtered, the filtrate was poured into saturated aq. NH4Cl solution (100 mL) and extracted with EA (100 mL three times). The combined organic layers were dried with anhydrous Na2SO4, filtered and the filtrate concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (PE:EA=50:1 to 15:1) to afford the title compound as light yellow oil (4100 mg, 91.8%). 1H NMR (400 MHz, CHLOROFORM-d) δ=4.16-4.11 (m, 2H), 3.93 (dd, J=6.5, 8.2 Hz, 2H), 3.37-3.20 (m, 1H), 2.28 (d, J=2.4 Hz, 1H), 1.43 (s, 9H).
  • Step c) tert-Butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethynyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (1000.0 mg, 5.52 mmol) and 4-bromo-3-fluorotoluene (1251.58 mg, 6.62 mmol, CAS RN 452-74-4) in dry THF (20 mL) were added Pd(PPh3)4 (530.63 mg, 0.460 mmol). CuI (87.83 mg, 0.460 mmol) and TEA (4644.2 mg, 46.0 mmol) at 25° C. The mixture was purged with N2 for 1 min and then stirred at 60° C. under N2 atmosphere for 12 h. The mixture was poured into saturated aq. NH4Cl solution (50 mL) and extracted three times with EtOAc (30 mL each). The combined organic layers were dried with anhydrous Na2SO4, filtered, the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (PE:EA=20:1 to 10:1) to provide the desired compound as colorless oil (650 mg, 40.7%). 1H NMR (400 MHz, CHLOROFORM-d) δ=7.33-7.28 (m, 1H), 6.94-6.85 (m, 2H), 4.26-4.19 (m, 2H), 4.05 (dd, J=6.4, 8.1 Hz, 2H), 3.66-3.49 (m. 1H). 2.36 (s, 3H), 1.46 (s, 9H).
  • Step d) tert-Butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethyl]azetidine-1-carboxylate
  • Batch a: To a solution of tert-butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethynyl]azetidine-1-carboxylate (50.0 mg, 0.170 mmol, 1 eq) in EtOAc (5 mL) was added Pd/C (50.0 mg, wt. 10%) at 25° C. The mixture was stirred at 40° C. under a balloon of hydrogen gas for 12 h. LCMS analysis found 79.8% of desired product.
  • Batch b: To a solution of tert-butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethynyl]azetidine-1-carboxylate (500.0 mg, 1.73 mmol) in EtOAc (10 mL) was added Pd/C (250.0 mg, wt. 10%) at 25° C. and the mixture was stirred at 40° C. under a balloon of hydrogen gas for 6 h. LCMS found 80.4% of desired product. Batch a and b were combined, the reaction mixture was filtered through a pad of celite, the filtrate was concentrated under reduced pressure and the residue was dried in vacuum to give the compound as colorless oil (350 mg, 69.0%). MS (ESI): m/z=238.1 [M−56+H]+.
  • BB164 3-[2-[4-Methoxy-2-(trifluoromethyl)phenyl]ethyl]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethyl]azetidine-1-carboxylate (180.0 mg, 0.5 mmol) in dry DCM (10 mL) was added TFA (1.0 mL, 1.19 mmol) at 25° C. and the mixture was stirred at 25° C. for 12 h. The reaction mixture was concentrated under reduced pressure and the residue was dried in vacuum to give the title compound (150 mg, 80.2%) as colorless oil. MS (ESI): m/z=260.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-[4-methoxy-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (800.0 mg, 4.41 mmol, BB111, intermediate b) and 3-trifluoromethyl-4-bromoanisole (1350.9 mg, 5.3 mmol, CAS RN 400-72-6) in dry THF (30 mL) at 25° C., was added Pd(PPh3)4 (509.41 mg, 0.440 mmol), CuI (84.31 mg, 0.440 mmol) and TEA (4458.42 mg, 44.14 mmol). The mixture was purged with N2 for 1 min and then stirred at 60° C. under N2 atmosphere for 12 h. The mixture was poured into saturated aq. NH4Cl solution (100 mL) and extracted three times with EtOAc (50 mL each). The organic layers were combined, dried with anhydrous Na2SO4, filtered, the filtrate was concentrated with reduced pressure. The residue was purified by flash chromatography on silica gel (PE:EA=20:1 to 10:1) to provide the product as colorless oil (160 mg, 8.2%). MS (ESI): m/z=300.1 [M−56+H]+.
  • Step b) tert-Butyl 3-[2-[4-methoxy-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[2-(2-fluoro-4-methyl-phenyl)ethynyl]azetidine-1-carboxylate (230.0 mg, 0.65 mmol) in EtOAc (10 mL) at 25° C., was added Pd/C (150.0 mg, wt. 10%), the mixture was stirred at 40° C. under a balloon of H2 (about 15 psi) for 12 h. The reaction mixture was filtered through a pad of celite and the filtrate was concentrated under reduced pressure. The residue was dried under vacuum to furnish the desired compound as colorless oil (180 mg, 77.4%). MS (ESI): m/z=304.1 [M−56+H]+.
  • BB165 3-[[4-Methyl-2-(trifluoromethyl)phenyl]methoxy]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[[4-methyl-2-(trifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate (130.0 mg, 0.380 mmol) in DCM (6.5 mL) was added TFA (1.3 mL, 16.87 mmol) and the reaction was stirred at 20° C. After 12 h the mixture was evaporated to give the desired crude product as light brown oil (130 mg, 96.1%). MS (ESI): m/z=246.5 [M+H]+.
  • Step a) 4-Bromo-1-(Bromomethyl)-2-(trifluoromethyl)benzene
  • The solution of 5-bromo-2-methylbenzotrifluoride (2000 mg, 8.37 mmol, CAS RN 86845-27-4), N-bromosuccinimide (1489 mg, 8.37 mmol, CAS RN 128-08-5) and benzoyl peroxide (101.34 mg, 0.420 mmol, CAS RN 2685-64-5) in carbon tetrachloride (30 mL) was stirred at 90° C. for 12 h. The mixture was evaporated and the residue was purified by silica gel column chromatography (100% PE) to give the desired product as light brown oil (690 mg, 25.9%) which was used in the next step without further purification.
  • Step b) tert-Butyl 3-[[4-bromo-2-(Trifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (337.5 mg, 1.95 mmol, CAS RN 22214-30-8) in THF (9 mL) was added t-BuOK (1.95 mL, 1.95 mmol), then 4-bromo-1-(bromomethyl)-2-(trifluoromethyl) benzene (590.0 mg, 1.86 mmol) was added and the mixture was stirred at 20° C. for 12 h. The mixture was poured into aq. NH4Cl solution (200 mL) and extracted three times with EtOAc (50 mL). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated. The residue was purified by prep-HPLC (ACN and water containing 0.225% v/v FA) to give the desired product as light brown oil (300 mg, 39.4%). MS (ESI): m/z=356.3 [M−56+H]+.
  • Step c) tert-Butyl 3-[[4-Methyl-2-(rifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[[4-bromo-2-(trifluoromethyl)phenyl]methoxy]azetidine-1-carboxylate (250.0 mg, 0.610 mmol), trimethylboroxine (114.8 mg, 0.910 mmol). K2CO3 (168.5 mg, 1.22 mmol) in 1,4-dioxane (10 mL) and H2O (2.5 mL) was added Pd(dppf)Cl2 (89.18 mg, 0.120 mmol). The reaction was stirred at 100° C. for 12 h. The mixture was filtered, concentrated, and the residue was purified by reversed flash chromatography (ACN and water containing 0.1% v/v FA) to give the desired product as light brown oil (146 mg, 69.4%). MS (ESI): m/z=290.4 [M−56+H]+.
  • BB166 3-[2-[2-Methoxy-6-(trifluoromethyl)phenyl]ethyl]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-[2-methoxy-6-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate (300.0 mg, 0.830 mmol) in DCM (5 mL), TFA (1.0 mL) was added and stirred at 25° C. for 1 h. The reaction mixture was evaporated under reduced pressure to give the desired product (300 mg, 96.3%) as colorless oil. MS (ESI): m/z=260.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-[2-methoxy-6-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (710.6 mg, 3.92 mmol, B111, intermediate b) and 2-bromo-1-methoxy-3-(trifluoromethyl)benzene (500.0 mg, 1.96 mmol) in dry DMSO (17.5 mL) at 25° C., was added Pd(PPh3)2Cl2 (137.6 mg, 0.200 mmol) and Cs2C3 (1278 mg, 3.92 mmol). The mixture was purged with N2 for 1 min and then stirred at 110° C. under N2 atmosphere for 12 h. The mixture was filtered, the filtrate was concentrated and the residue was purified by silica gel (PE:EtOAc=20:1) to give the desired product as light yellow oil (600 mg, 86.1%) that was used in the next step without further purification.
  • Step b) tert-Butyl 3-[2-[2-methoxy-6-(trifluoromethyl) phenyl]ethynyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[2-[2-methoxy-6-(trifluoromethyl)phenyl]ethynyl]azetidine-1-carboxylate (400.0 mg, 1.13 mmol) in EtOAc (20 mL), wet Pd/C (50 mg, 10 wt. %) was added. The mixture was purged with H2 3 times and then stirred at 40° C. under H2 atmosphere (balloon) for 12 h. The mixture was filtered and the filtrate was concentrated to give the desired product as light yellow oil (300 mg, 74.2% yield) which was used in the next step without further purification.
  • BB167 3-[2-[4-Methyl-2-(trifluoromethyl)phenyl]ethyl]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-[4-methyl-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate (100.0 mg, 0.290 mmol) in DCM (4 mL) was added TFA (0.5 mL) and the mixture was stirred at 20° C. for 12 h. The reaction mixture was evaporated under reduced pressure to give the desired product as yellow oil (98 mg, 94.2%). MS (ESI): m/z=244.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-[4-methyl-2-(trifluoromethyl)phenyl]ethynyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (606.6 mg, 3.35 mmol) and 2-bromo-5-methylbenzotrifluoride (400.0 mg, 1.67 mmol) in dry DMSO (14.9 mL) at 25° C., was added Pd(PPh3)2Cl2 (117.46 mg, 0.170 mmol) and Cs2CO3 (1091 mg, 3.35 mmol). The mixture was purged with N2 for 1 min and then stirred at 110° C. under N2 atmosphere for 12 h. The reaction mixture was poured into H2O and extracted with EtOAc. The organic layer was evaporated and the residue was purified by silica gel column chromatography (PE:EtOAc=20:1) to give the desired compound as a yellow oil (390 mg, 68.7% yield). MS (ESI): m/z=284.1 [M−56+H]+.
  • Step b) tert-Butyl 3-[2-[4-methyl-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[2-[4-methyl-2-(trifluoromethyl)phenyl]ethynyl]azetidine-1-carboxylate (390.0 mg, 1.15 mmol) in EtOAc (19.5 mL), wet Pd/C (150 mg, 10 wt. %) was added, the mixture was purged 3 times with H2 and stirred at 40° C. under H2 atmosphere (balloon) for 12 h. The mixture was filtered and the filtrate was concentrated to give the desired product as light yellow oil (295 mg, 72.9% yield). MS (ESI): m/z=288.1 [M−56+H]+.
  • BB68 1-[2-[2-(Azetidin-3-yl)ethyl]-5-(trifluoromethyl)phenyl]ethanone trifluoroacetate
  • To a solution of tert-butyl 3-[2-[2-acetyl-4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate (50.0 mg, 0.130 mmol) in DCM (1 mL) was added TFA (0.2 mL) and the solution was stirred at 20° C. for 12 h. The mixture was concentrated to give the desired product as light brown oil (50 mg, 96.4% yield). MS (ESI): m/z=272.1 [M+H]+.
  • Step a) 2-Bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene
  • To a solution of [2-bromo-4-(trifluoromethyl)phenyl]methanol (500.0 mg, 1.96 mmol, CAS RN 497959-33-8) and PPh3 (770.5 mg, 2.94 mmol) in THF (10 mL) was added carbon tetrabromide (975.3 mg, 2.94 mmol), and the mixture was stirred at 25° C. for 12 h. The reaction was concentrated in vacuum and the residue was purified by silica gel column chromatography (PE:EA=0:1-20:1) to yield the desired product as colorless oil (600 mg, 96.3% yield). 1H NMR (400 MHz, CHLOROFORM-d) δ=7.78 (s, 1H), 7.55-7.46 (m, 2H). 4.53 (s, 2H).
  • Step b) 2-Bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene
  • A solution of 2-bromo-1-(bromomethyl)-4-(trifluoromethyl)benzene (600.0 mg, 1.89 mmol) in triethyl phosphite (3136 mg, 18.87 mmol) was stirred at 160° C. for 5 h. The mixture was concentrated at 100° C. under reduced pressure to remove most of the triethyl phosphite to give the crude product (700 mg) as light yellow oil. MS (ESI): m/z=375.2 [M+H]+.
  • Step c) tert-Butyl 3-[(E)-2-[2-bromo-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate
  • A mixture of 2-bromo-1-(diethoxyphosphorylmethyl)-4-(trifluoromethyl)benzene (600.0 mg, 1.6 mmol) in THF (10 mL) was added to another suspension of NaH (191.9 mg, 4.8 mmol, 60% dispersion in mineral oil) in THF (10 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. Then tert-butyl 3-formylazetidine-1-carboxylate (296.3 mg, 1.6 mmol) was added and the mixture was stirred at 20° C. for 11 h. The reaction mixture was poured into aq. NH4Cl solution (100 mL) and extracted three times with EtOAc (50 mL each). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (PE:EtOAc=20:1) to give the desired product as light yellow oil (450 mg, 69.3%). MS (ESI): m/z=352.0 [M56+H]+. 1H NMR (400 MHz, CHLOROFORM-d) δ=7.74 (d, J=0.8 Hz, 1H), 7.58-7.51 (m, 1H), 7.49-7.41 (m, 1H), 6.71 (d, J=15.8 Hz, 1H), 6.36 (dd, J=8.4, 15.8 Hz, 1H), 4.13 (t, J=8.5 Hz, 2H), 3.78 (dd, J=5.8, 8.6 Hz, 2H), 3.44-3.31 (m, 1H), 1.39 (s, 9H).
  • Step d) tert-Butyl 3-[(E)-2-[2-acetyl-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate
  • A solution of tributyl(1-ethoxyvinyl)tin (426.7 mg, 1.18 mmol), tert-butyl 3-[(E)-2-[2-bromo-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate (400.0 mg, 0.980 mmol) and Pd(Ph3P)2Cl2 (138.2 mg, 0.200 mmol) in THF (16 mL) was stirred at 80° C. under N2 atmosphere for 4 h. The mixture was cooled down to room temperature and aq. KF solution (10 mL) was added. The mixture was stirred for 10 mins. extracted three times with EtOAc (20 mL each) and the combined organic layers were concentrated. The residue was dissolved in THF (20 mL) and aq. HCl (0.6 N, 20 mL) was added. The mixture was stirred at 20° C. for 0.5 h, extracted three times with EtOAc (20 mL each) and the combined organic layers were concentrated. The residue was purified by silica gel column chromatography (PE:EtOAc=20:1) to give the desired product (280 mg, 77% yield) as light yellow oil. MS (ESI): m/z=314.1 [M−56+H]+.
  • Step e) tert-Butyl 3-[2-[2-acetyl-4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-[(E)-2-[2-acetyl-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate (50.0 mg, 0.140 mmol) in EtOAc (5 mL) was added wet Pd/C (20.0 mg, 10 wt. %) and the mixture was stirred at 20° C. under H2 (balloon) atmosphere for 12 h. The reaction was then warmed up to 50° C. and stirred for another 12 h. The mixture was filtered and the filtrate was concentrated to give the desired product (50 mg, 99.5%) as light yellow oil. MS (ESI): m/z=316.2 [M−56+H]+.
  • BB169 3-[2-[2-Bromo-4-(trifluoromethyl)phenyl]ethyl]azetidine trifluoroacetate
  • To a solution of tert-butyl 3-[2-[2-bromo-4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate (400.0 mg, 0.980 mmol) in DCM (10 mL) was added TFA (1.0 mL) and the mixture was stirred at 20° C. for 12 h. The reaction mixture was evaporated under reduced pressure to give the desired product (413 mg, 99.8% yield) as yellow oil. MS (ESI): m/z=308.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-[2-bromo-4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a suspension of tert-butyl 3-[(E)-2-[2-bromo-4-(trifluoromethyl) phenyl]vinyl]azetidine-1-carboxylate (600.0 mg, 1.48 mmol, BB116, intermediate c) and MgO (118.1 mg, 2.95 mmol) in EtOAc (20 mL) was added Pd/C (300.0 mg, 10 wt. %), the mixture was stirred at 25° C. under H2 atmosphere (balloon) for 1 h. The reaction mixture was filtered and the filtrate was evaporated under reduced pressure to give the desired product (500 mg, 82.9%) as light yellow oil. MS (ESI): m/z=352.0 [M−56+H]+.
  • BB174 2-(Azetidin-3-ylmethoxy)-5-(trifluoromethyl)pyridine 2,2,2-trifluoroacetate
  • Synthesis of BB174 was performed in analog) to BB57, starting from tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate and 2-bromo-5-(trifluoromethyl)pyridine. MS (ESI): m/z=233.1 [M+H]+.
  • BB75 3-Methyl-5-[[rac-(3R,4R)-3-methyl-4-piperidyl]methoxy]-2-(trifluoromethyl)pyridine dihydrochloride
  • tert-Butyl (rac-3R,4R)-3-methyl-4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carboxylate (198 mg, 510 μmol) was dissolved in DCM (2 mL) and HCl 2M in ether (1.53 mL, 3.06 mmol) was added. The reaction mixture was stirred at RT for 8 h. The reaction mixture was concentrated in vacuo to yield 180 mg of desired product as white solid (98%) MS (ESI): m/z=289.3 [M+H]+.
  • a) tert-Butyl (rac-3R, 4R)-4-(hydroxymethyl)-3-methylpiperidine-1-carboxylate
  • To a stirred solution of cis-N-BOC-3-methylpiperidine-4-carboxylic acid methyl ester (2 g, 7.77 mmol) in THF (10 ml) was added lithium borohydride (5.83 mL, 11.7 mmol) at 2-5° C. The reaction mixture was then heated at reflux for 3 h and then cooled to 2-5° C. Water was added and the aqueous layer was extracted twice with EtOAc (30 mL each). The organic layer was washed with water, NaHCO3 and brine, the layers were separated, and the organics dried over Na2SO4 and concentrated in vacuum. Purification by flash chromatography (gradient of EtOAc in n-heptane, 0 to 65%) provided the product as a colorless oil (930 mg, 50%). MS (ESI): m/z=174.1 [M−56+H]+.
  • b) tert-Butyl (rac-3R, 4R)-3-methyl-4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carboxylate
  • tert-butyl (3R,4R)-4-(hydroxymethyl)-3-methylpiperidine-1-carboxylate (239 mg, 1.04 mmol) was dissolved in DMF (4.17 mL) and NaH in mineral oil (60%, 45.8 mg, 1.15 mmol) was added at RT. The reaction was stirred for 20 min, then 5-bromo-3-methyl-2-(trifluoromethyl)pyridine (250 mg, 167 μL, 1.04 mmol) was added and stirring continued for 12 h at RT. The reaction was quenched with 10 mL sat. NH4Cl solution and extracted three times with water/EtOAc. The organic phases were combined and dried over MgSO4 and the solvent was removed in vacuo. Flash chromatography (gradient of EtOAc in n-heptane, 0 to 50%) yielded the product as white solid (148 mg, 49%). MS (ESI): m/z=333.2 [M−56+H]+.
  • BB176 3-((2-Fluoro-4-(trifluoromethyl)benzyl)oxy)-2-methylazetidine 2,2,2-trifluoroacetate
  • To a solution of tert-butyl 3-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)-2-methylazetidine-1-carboxylate (0.265 g, 729 μmol) in DCM (4 mL) was added TFA (832 mg, 562 μL, 7.29 mmol). The resultant reaction mixture was stirred at RT for 1 h. The reaction mixture was concentrated to give the title compound as a colorless oil. The crude product was used without further purification. MS (ESI): m/z=264.2 [M+H]+.
  • Step a) [2-Fluoro-4-(trifluoromethyl)phenyl]methyl methanesulfonate
  • To an ice-cold solution of (2-fluoro-4-(trifluoromethyl)phenyl)methanol (840 mg, 4.33 mmol) and triethylamine (1.31 g, 1.81 mL, 13 mmol) in DCM (8 mL) was added dropwise methanesulfonyl chloride (496 mg, 337 μL, 4.33 mmol) and the mixture was stirred at 0° C. for 1 h. The reaction mixture was poured on saturated aqueous NaHCO3 solution (10 mL) and DCM (20 mL) and the layers were separated. The aqueous layer was extracted once with DCM (20 mL). The organic layers were washed once with brine, dried over MgSO4, filtered and evaporated to yield the desired compound as a yellow oil (1.13 g, 96%).
  • Step b) tert-Butyl 3-[[2-fluoro-4-(trifluoromethyl)phenyl]methoxy]-2-methyl-azetidine-1-carboxylate
  • To an ice-cold solution of tert-butyl 3-hydroxy-2-methylazetidine-1-carboxylate (250 mg, 1.34 mmol) in DMF (3 mL) was added NaH (60% in mineral oil, 58.7 mg, 1.47 mmol) in portions and the mixture was stirred at ice-bath temperature for 5 min followed by stirring at RT for 40 min. A solution of 2-fluoro-4-(trifluoromethyl)benzyl methanesulfonate (436 mg, 1.6 mmol) in DMF (1 mL) was added dropwise to the mixture at RT. Stirring of the slurry was continued at RT for 16 h. The reaction mixture was poured on saturated aqueous NH4C solution (10 mL) and EtOAc (20 mL) and the layers were separated. The aqueous layer was extracted once with EtOAc (50 mL). The organic layers were washed twice with water, dried over Na2SO4, filtered, and concentrated. The crude compound was purified by silica gel chromatography (gradient of n-heptane:EtOAc 100:0 to 0:100) to get tert-butyl 3-[[2-fluoro-4-(trifluoromethyl)phenyl]methoxy]-2-methyl-azetidine-1-carboxylate as a colorless oil (0.265 g, 54.6% yield). MS (ESI): m/z=308.2 [M−56+H]+.
  • BB 177 2-(Azetidin-3-ylmethoxy)-4,5-bis(trifluoromethyl)pyridine 2,2,2-trifluoroacetate
  • Synthesis of BB 177 was performed in analogy to BB57, starting from tert-butyl 3-(hydroxymethyl)azetidine-1-carboxylate and 2-chloro-4,5-bis(trifluoromethyl)pyridine. MS (ESI): m/z=301.2 [M+H]+.
  • BB179 3-((4-Chloro-2-phenoxybenzyl)oxy)azetidine 2,2,2-trifluoroacetate
  • Synthesis of BB179 was done in analogy to BB39, starting from tert-butyl 3-hydroxyazetidine-1-carboxylate and 1-(bromomethyl)-4-chloro-2-phenoxybenzene (synthesis described below). MS (ESI): m/z=290.2 [M+H]+.
  • 1-(Bromomethyl)-4-chloro-2-phenoxybenzene
  • i) In a 10 mL round-bottomed flask, methyl 4-chloro-2-phenoxybenzoate (547 mg, 2.08 mmol) was diluted in toluene (3.82 mL) and the reaction mixture was cooled in an ice bath. Sodium bis(2-methoxyethoxy)aluminum hydride 70% in toluene (649 mg, 637 μL, 2.25 mmol) was added dropwise slowly at max. 15° C. to give a light yellow solution. The reaction mixture was stirred at r.t. for 30 min. The crude reaction mixture, containing the product (4-chloro-2-phenoxyphenyl)methanol was used directly in the next step.
  • ii) In a 25 mL round-bottomed flask, hydrobromic acid 48% in H2O (6.49 g, 4.35 mL, 38.5 mmol) was cooled in an ice bath. Then 4-chloro-2-phenoxyphenyl)methanol (crude, 488 mg, 2.08 mmol) was added dropwise slowly and the mixture was stirred at 50° C. for 2 h. Hydrobromic acid 48% in H2O (6.25 g, 2.18 mL, 19.25 mmol) was added and the mixture was stirred at 60° C. for 1 h, then cooled to RT. The aqueous phase was separated, the organic phase was washed four times with H2O and evaporated. The crude material was purified by flash column chromatography (gradient 0% to 25% EtOAc in hexanes) and was used in the next step without further purification. Yield: 85%.
  • BB 181 3-((1-(2,4-Dichlorophenyl)cyclopropyl)methoxy)azetidine 2,2,2-trifluoroacetate
  • To a solution of tert-Butyl 3-((1-(2,4-dichlorophenyl)cyclopropyl)methoxy)azetidine-1-carboxylate (165 mg, 443 μmol) in DCM (2 mL) was added TFA (202 mg, 137 μL, 1.77 mmol) and the reaction stirred at RT for 8 h. The mixture was concentrated in vacuo (azeotrop with toluene, EtOAc and n-heptane) to provide the compound as a colorless oil (170 mg, 99%). MS (ESI): m/z=272.2 [M+H]+.
  • Step a) 1-(2,4-Dichlorophenyl)cyclopropyl)methanol
  • In a 50 mL three-necked flask, 1-(2,4-dichlorophenyl)cyclopropane-1-carboxylic acid (1 g, 4.33 mmol) was combined with THF (20 mL) to give a colorless solution. At 0° C., borane tetrahydrofuran complex solution 1.0 M in THF (6.49 mL, 6.49 mmol) was added dropwise over a period of 15 min. The reaction was stirred at RT for 2 h. MeOH (2 mL) was added dropwise followed by 1M aq. HCl solution and stirred for 30 min. The reaction mixture was extracted twice with EtOAc (40 mL each) and the organic layers were washed with 10% aq Na2CO3 solution (40 mL) followed by brine (40 mL). The organic fractions were combined and dried over Na2SO4 and concentrated in vacuo. The crude material was purified by flash column chromatography (gradient EtOAc in n-heptane, 0% to 30%) to yield the compound as colorless oil (90%) MS (ESI): m/z=201.0 [M−16+H]+.
  • Step b) 1-(2,4-Dichlorophenyl)cyclopropyl]methyl methanesulfonate
  • To an ice-cold solution of (1-(2,4-dichlorophenyl)cyclopropyl)methanol (350 mg, 1.61 mmol) and TEA (326 mg, 449 μL, 3.22 mmol) in DCM (6 mL) was added dropwise methanesulfonyl chloride (185 mg, 126 μL, 1.61 mmol) and the mixture was stirred at 0° C. for 1 h, then at RT overnight. The reaction mixture was poured on saturated aqueous NaHCO3 solution (10 mL) and DCM (10 mL) and the layers were separated. The aqueous layer was extracted once with DCM (10 mL). The organic layers were washed with brine, dried over MgSO4, filtered and evaporated to furnish the desired intermediate mesylate compound as a yellow oil (435 mg, 91%). MS (ESI): m/z=201.0 [M−mesyl+H]+.
  • Step c) tert-Butyl 3-((1-(2,4-dichlorophenyl)cyclopropyl)methoxy)azetidine-1-carboxylate
  • To an ice-cold solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (220 mg, 1.27 mmol) in DMF (4 mL) was added sodium hydride in mineral oil (60%, 61 mg, 1.52 mmol) in portions and the mixture was stirred at ice-bath temperature for 5 min followed by stirring at RT for 40 min. A solution of 1-(2,4-dichlorophenyl)cyclopropyl)methyl methanesulfonate (431 mg, 1.46 mmol) was dissolved in DMF (1 mL) and added dropwise to the mixture at RT. Stirring of the slurry was continued at RT for 16 h, then at 55° C. for 2.5 h. The reaction mixture was poured on saturated aqueous NH4Cl solution (10 mL) and EtOAc (20 mL) and the layers were separated. The aqueous layer was extracted once with EtOAc (50 mL). The organic layers were washed twice with water, dried over MgSO4, filtered and evaporated. Flash Chromatography (gradient of EtOAc in n-heptane 0 to 40%) yielded the product as colorless oil (165 mg, 35%) MS (ESI): m/z=316.2 [M−56+H]+.
  • BB182 2-((Azetidin-3-yloxy)methyl)-6-(4-fluorophenoxy)-4-(trifluoromethyl)pyridine 4-methylbenzenesulfonate
  • Tert-butyl 3-((6-(4-fluorophenoxy)-4-(trifluoromethyl)pyridin-2-yl)methoxy)azetidine-1-carboxylate (150 mg, 339 μmol) was dissolved under argon in EtOAc (2 mL), p-toluenesulfonic acid monohydrate (77.4 mg, 407 μmol) was added and the mixture was stirred at RT for 5 min, then for 80° C. 3 h at and at RT over night. The reaction mixture was evaporated to provide the compound as 180 mg of a yellow oil which was used in the next step without further purification. MS (ESI): m/z=343.2 [M+H]+.
  • Step a) tert-Butyl 3-((6-bromo-4-(trifluoromethyl)pyridin-2-yl)methoxy)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (272 mg, 1.57 mmol) in dry THF (8 mL) was added potassium tert-butoxide IM in THF (1.57 mL, 1.57 mmol) and the turbid reaction mixture was stirred at RT for 30 min. 2-Bromo-6-(bromomethyl)-4-(trifluoromethyl)pyridine (500 mg, 1.57 mmol) was added at 0-2° C. and the reaction stirred at 0-2° C. for 20 min. The reaction mixture was then stirred at RT for 16 h. The reaction mixture was diluted with EtOAc, extracted with water, the organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic layers were dried over sodium sulfate and evaporated down to dryness. The crude material was purified by flash column chromatography (gradient of EtOAc in n-heptane, 0% to 40%) to provide the product as light yellow oil (41%) MS (ESI): m/z=355.1 [M−56+H]+.
  • Step b) tert-Butyl 3-[[6-(4-fluorophenoxy)-4-(trifluoromethyl)-2-pyridyl]methoxy]azetidine-1-carboxylate
  • tert-Butyl 3-((6-bromo-4-(trifluoromethyl)pyridin-2-yl)methoxy)azetidine-1-carboxylate (260 mg, 632 μmol) and 4-fluorophenol (70.9 mg, 632 μmol) were dissolved in DMF (2 mL), then K2CO3 (131 mg, 948 μmol) was added and the mixture was stirred at 80° for 30 h. The reaction mixture was evaporated under vacuum and the residue was dissolved in EtOAc and extracted with water and brine. The organic layers were dried over MgSO4, filtered and the solvent was removed under vacuum. The residue was purified by flash chromatography (gradient of EtOAc in n-heptane, 0 to 30%) to yield the product as a light yellow oil (93%). MS (ESI): m/z=443.4 [M+H]+.
  • BB183 6-((Azetidin-3-yloxy)methyl)-2-(4-fluorophenoxy)-3-(trifluoromethyl)pyridine 4-methylbenzenesulfonate
  • tert-Butyl 3-((6-(4-fluorophenoxy)-5-(trifluoromethyl)pyridin-2-yl)methoxy)azetidine-1-carboxylate (170 mg, 384 μmol) was dissolved under argon atmosphere in EtOAc (2.27 mL) and p-toluenesulfonic acid monohydrate (87.7 mg, 461 μmol) was added. The reaction was stirred at RT for 5 min, then at 80° C. for 3 h and stirred at RT over night. The reaction mixture was evaporated under reduced pressure to dryness to provide the desired product as light yellow oil (89%) MS (ESI): m/z=343.2 [M+H]+.
  • Step a) Methyl 6-(4-fluorophenoxy)-5-(trifluoromethyl)picolinate
  • Methyl 6-chloro-5-(trifluoromethyl)picolinate (800 mg, 3.34 mmol), 4-fluorophenol (412 mg, 3.67 mmol) and K2CO3 (692 mg, 5.01 mmol) were dissolved in DMF (6 mL) and stirred at 80° C. for 6 h. The reaction mixture was cooled to RT and extracted three times with water (20 mL each), twice with EtOAc (30 mL each), brine (20 mL), dried over MgSO4, filtered and evaporated in vacuo. The crude residue was purified by flash column chromatography (gradient of EtOAc in n-heptane, 0 to 50%) to provide the product as white solid (67%). MS (ESI): m/z=316.1 [M+H]+.
  • Step b) (6-(4-Fluorophenoxy)-5-(trifluoromethyl)pyridin-2-yl)methanol
  • To a stirred solution of methyl 6-(4-fluorophenoxy)-5-(trifluoromethyl)picolinate (705 mg, 2.24 mmol) in THF (8 mL) was added lithium borohydride 2M in THF (1.34 mL, 2.68 mmol) at 2-5° C. The reaction mixture was stirred at RT for 3 h and then cooled to 2-4° C. and quenched with 10 mL water (slowly added). The aqueous layer was extracted twice with EtOAc (30 mL each) and the combined organic layers were washed with water, 10 mL NaHCO3 solution and 10 mL brine. The organic layer was dried over Na2SO4 and concentrated in vacuum. Purification by flash column chromatography (gradient of EtOAc in n-heptane, 0 to 50%) yielded the product as a colorless solid (95%). MS (ESI): m/z=288.2 [M+H]+.
  • Step c) 6-(Bromomethyl)-2-(4-fluorophenoxy)-3-(trifluoromethyl)pyridine
  • To a solution of (6-(4-fluorophenoxy)-5-(trifluoromethyl)pyridin-2-yl)methanol (330 mg, 1.15 mmol) in dry DCM (5 mL) was added tetrabromomethane (457 mg, 1.38 mmol). The mixture was cooled to 0-3° C. and over 10 min triphenylphosphine (392 mg, 1.49 mmol) in 1 mL dry DCM was added. The mixture was stirred 1 hr at 2-4° C., then 20 mL DCM and silica gel was added. The solvent was removed in vacuo and the residue subjected to column flash chromatography (gradient of EtOAC in n-heptane. 0 to 40%) to yield the desired product as a colorless oil (94%). MS (ESI): m/z=350.0 [M+H]+.
  • Step d) tert-Butyl 3-((6-(4-fluorophenoxy)-5-(trifluoromethyl)pyridin-2-yl)methoxy)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (183 mg, 1.06 mmol) in dry THF (5 mL) was added potassium tert-butoxide 1M in THF (1.11 mL, 1.11 mmol) and the reaction mixture was stirred at RT for 15 min. Then, 6-(bromomethyl)-2-(4-fluorophenoxy)-3-(trifluoromethyl)pyridine (370 mg, 1.06 mmol) was added. The reaction mixture was stirred at RT for 1 h and then diluted with EtOAc and extracted with 1M aq. NaHCO3 solution. The organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over sodium sulfate and evaporated down to dryness. The residue was purified by column flash chromatography (gradient of EtOAc in n-heptane, 0 to 30%) to furnish the product as a colorless oil (34%). MS (ESI): m/z=387.2 [M−56+H]+.
  • BB184 2-((Azetidin-3-yloxy)methyl)-4-(4-fluorophenyl)thiazole 2,2,2-trifluoroacetate
  • To a solution of tert-butyl 3-((4-(4-fluorophenyl)thiazol-2-yl)methoxy)azetidine-1-carboxylate (150 mg, 412 μmol) in dry DCM (1.5 mL) under argon atmosphere was added TFA (282 mg, 190 μL, 2.47 mmol) and the solution was stirred at RT for 8 h. The reaction mixture was concentrated in vacuo (azeotrop with toluene, EtOAc and heptane) to yield the desired product as a yellow solid (98%). MS (ESI): m/z=265.2 [M+H]+.
  • Step a) (4-(4-Fluorophenyl)thiazol-2-yl)methanol
  • To a stirred solution of ethyl 4-(4-fluorophenyl)thiazole-2-carboxylate (835 mg, 3.32 mmol) in dry THF (10 mL) was added lithium borohydride 2M in THF (1.99 mL, 3.99 mmol) at 2-5° C. The reaction mixture was stirred at RT for 3 h, then cooled to 2-4° C. and quenched with water (10 mL slowly added). The aqueous layer was extracted twice with EtOAc (30 mL each) and the organic layers were washed with water, 10 mL NaHCO3 solution and 10 mL brine. The combined organic layers were dried over Na2SO4 and concentrated in vacuum. The residue was purified by column flash chromatography (gradient of EtOAc in n-heptane, 0 to 60%) to yield the desired product as a white solid (94%) MS (ESI): m/z=210.1 [M+H]+.
  • Step b) 2-(Bromomethyl)-4-(4-fluorophenyl)thiazole
  • To a solution of (4-(4-fluorophenyl)thiazol-2-yl)methanol (400 mg, 1.91 mmol) in dry DCM (7 mL) was added tetrabromomethane (761 mg, 2.29 mmol), the solution was cooled to 0-3° C. and triphenylphosphine (652 mg, 2.49 mmol) in 1 mL dry DCM was added over 10 min. The mixture was stirred at 2-4° C. for 1 h, then 20 mL DCM were added. The reaction mixture was extracted with water, saturated NH4Cl solution and brine. The organic phase was dryed over MgSO4, filtered and evaporated. The residue was purified by flash chromatography (gradient of EtOAc in n-heptane, 0 to 40%) to provide 480 mg of the title compound as a light yellow oil (83%). MS (ESI): m/z=273.9 [M+H]+.
  • Step c) tert-Butyl 3-((4-(4-fluorophenyl)thiazol-2-yl)methoxy)azetidine-1-carboxylate
  • To a solution of tert-butyl 3-hydroxyazetidine-1-carboxylate (293 mg, 1.69 mmol) in dry THF (6 mL) was added potassium tert-butoxide IM in THF (1.77 mL, 1.77 mmol) and the reaction mixture was stirred at RT for 15 min. After cooling down to 2-4° C. 2-(bromomethyl)-4-(4-fluorophenyl)thiazole (460 mg, 1.69 mmol) in 1 mL THF was added. The reaction mixture was stirred at RT for 1 h, diluted with EtOAc and extracted with 1M aq. NaHCO3 solution. The organic phase was collected and the aqueous phase was back-extracted with EtOAc. The combined organic phases were dried over Na2SO4 and evaporated down to dryness. The residue was purified by column flash chromatography (gradient of EtOAc in n-heptane, 0 to 40%) to furnish the desired product as a light yellow solid (89%/). MS (ESI): m/z=365.2 [M+H]+.
  • BB186 rac-(2R,3S)-3-(2-Bromo-5-(trifluoromethyl)phenoxy)-2-methylpyrrolidine 2,2,2-trifluoroacetate
  • To a solution of rac-tert-butyl (2R,3S)-3-(2-bromo-5-(trifluoromethyl)phenoxy)-2-methylpyrrolidine-1-carboxylate (225 mg, 530 μmol) in dry DCM (2 mL) under argon atmosphere was added TFA (242 mg, 163 μL, 2.12 mmol) and the solution was stirred at RT over night. The reaction mixture was concentrated in vacuo to dryness (azeotrop with n-heptane) to provide 233 mg of the title compound as a colorless oil (97%). MS (ESI): m/z=324.1 [M+H]+.
  • Step a) rac-tert-Butyl (2R,3S)-3-(2-bromo-5-(trifluoromethyl)phenoxy)-2-methylpyrrolidine-1-carboxylate
  • To a solution of rac-tert-butyl (2R,3S)-3-hydroxy-2-methylpyrrolidine-1-carboxylate (CAS: 1807941-04-3, 150 mg, 745 μmol) in dry THF (4 mL) under argon atmosphere was added potassium tert-butoxide 1M in THF (783 μL, 783 μmol). The mixture was stirred at RT for 15 min, then cooled down to 2-4° C. and a solution of 1-bromo-2-fluoro-4-(trifluoromethyl)benzene (181 mg, 745 μmol) in 0.5 mL dry THF was added slowly. The mixture was stirred at RT for 2 h and then extracted with EtOAc and aqueous 5% NaHCO3 solution followed by water and brine. The organic phase was dried over MgSO4, filtered off and evaporated to dryness. The residue was purified by column flash chromatography (gradient of EtOAc in n-heptane, 0 to 40%) to yield the product as light yellow oil (71%). MS (ESI): m/z=368 [M−56+H]+.
  • The following intermediates were synthesized from 4-nitrophenyl (4aR,8aS)-3-oxohexahydro-2H-pyrido[4,3-b][1,4]oxazine-6(5H)-carboxylate (BB7a) and the suitable building blocks in analogy to the reaction methods described herein.
  • BB No. Building block(s) MS, m/z Method
    BB203 BB198 480.1 [M + H]+ A10 without DMAP
    BB204 BB201 445.1 [M + H]+ A10 without DMAP
  • BB206 3-[2-[2-Fluoro-4-(trifluoromethyl)phenyl]ethyl]azetidine; 4-methylbenzenesulfonic acid
  • The compound was prepared in analogy to BB95 from tert-butyl 3-(2-fluoro-4-(trifluoromethyl)phenethyl)azetidine-1-carboxylate and 4-methylbenzenesulfonic acid monohydrate. Upon cooling a suspension formed which was filtered. The filter cake was washed with a small volume of EtOAc to provide the desired product as a colorless solid (71.6%). MS (ESI): m/z=248.2 [M+H]+.
  • Step a) Diethyl (2-fluoro-4-(trifluoromethyl)benzyl)phosphonate
  • The compound was prepared in analogy to BB159, step a, from 1-(bromomethyl)-2-fluoro-4-(trifluoromethyl)benzene and triethyl phosphite. Colorless oil (83.4%). MS (ESI): m/z=315.2 [M+H]+.
  • Step b) tert-Butyl 3-[(E)-2-[2-fluoro-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate
  • The compound was prepared in analogy to BB95, step a, from diethyl (2-fluoro-4-(trifluoromethyl)benzyl)phosphonate and tert-butyl 3-formylazetidine-1-carboxylate to yield the compound as a colorless oil (69.9%). MS (ESI): m/z=290.1 [M−56+H]+.
  • Step c) tert-Butyl 3-[2-[2-fluoro-4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • The compound was prepared in analogy to BB95, step b, from tert-butyl 3-[(E)-2-[2-fluoro-4-(trifluoromethyl)phenyl]vinyl]azetidine-1-carboxylate. Colorless oil (92.0%). MS (ESI): m/z=292.2 [M−56+H]+.
  • BB208 3-[2,2-Difluoro-2-[4-(trifluoromethyl)phenyl]ethyl]azetidine; 4-methylbenzenesulfonic acid
  • The compound was prepared in analogy to BB95 from tert-butyl 3-(2,2-difluoro-2-(4-(trifluoromethyl)phenyl)ethyl)azetidine-1-carboxylate and 4-methylbenzenesulfonic acid monohydrate and using the material isolated from the filtrate after evaporation, which was used without further purification (30%). MS (ESI): m/z=266.2 [M+H]+.
  • Step a) tert-Butyl 3-[2-[methoxy(methyl)amino]-2-oxo-ethyl]azetidine-1-carboxylate
  • To a suspension of 2-(1-(tert-butoxycarbonyl)azetidin-3-yl)acetic acid (2 g, 9.29 mmol) and HATU (3.89 g, 10.2 mmol) in DCM (65 mL) was added DIPEA (2.64 g, 3.57 mL, 20.4 mmol) and the mixture was stirred at RT for 30 min before N,O-dimethylhydroxylamine hydrochloride (906 mg, 9.29 mmol) was added. Stirring was continued at RT overnight. The reaction mixture was poured on saturated aqueous NH4Cl solution and EtOAc and the layers were separated. The aqueous layer was extracted twice with EtOAc. The organic layers were washed twice with water, dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 25 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (100:0 to 0:100) to furnish the desired compound as a colorless oil (100%) which was used in the next step without further purification. MS (ESI): m/z=203.2 [M−56+H]+.
  • Step b) tert-Butyl 3-[2-oxo-2-[4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To an ice-cold solution of tert-butyl 3-(2-(methoxy(methyl)amino)-2-oxoethyl)azetidine-1-carboxylate (0.8 g, 3.1 mmol) in THF (5 mL) in an argon-flushed and heat-dried 2-neck flask was added dropwise a turbid solution of (4-(trifluoromethyl)phenyl)magnesium bromide 2.22 M in THF (1.95 mL, 4.34 mmol). The brown solution was stirred in an ice-bath for 2.5 h allowing the temperature to rise to RT. The reaction mixture was poured on saturated aqueous NH4Cl solution and EtOAc and the layers were separated. The aqueous layer was extracted twice with EtOAc. The organic layers were dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 25 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (100:0 to 0:100) to provide the desired compound as a colorless solid (25.9%). MS (ESI): m/z=342.3 [M−H].
  • Step c) tert-Butyl 3-[2,2-difluoro-2-[4-(trifluoromethyl)phenyl]ethyl]azetidine-1-carboxylate
  • To a solution of tert-butyl 3-(2-oxo-2-(4-(trifluoromethyl)phenyl)ethyl)azetidine-1-carboxylate (50 mg, 146 μmol) in toluene (0.3 mL) under argon was added bis(2-methoxyethyl)aminosulphur trifluoride (50% solution in THF, 387 mg, 379 μL, 874 μmol) and the mixture was stirred at 80° C. for 19 h. The dark mixture was allowed to cool down and another batch of bis(2-methoxyethyl)aminosulphur trifluoride (50% solution in THF, 387 mg, 379 μL, 874 μmol) was added. Heating was continued at 80° C. for another 4 h. The reaction mixture was poured on saturated aqueous NaHCO3 solution and EtOAc and the layers were separated. The aqueous layer was extracted twice with EtOAc. The organic layers were dried over MgSO4, filtered, treated with silica gel and evaporated. The compound was purified by silica gel chromatography on a 4 g column using an MPLC system eluting with a gradient of n-heptane:EtOAc (100:0 to 50:50) to yield the desired compound as a light brown oil (45.1%). MS (ESI): m/z=266.1 [M+H]+.
  • BB209 3-[2-Fluoro-5-(trifluoromethyl)phenoxyl]pyrrolidine; 4-methylbenzenesulfonic acid
  • The compound was prepared in analogy to BB95 from tert-butyl 3-[2-fluoro-5-(trifluoromethyl)phenoxy]pyrrolidine-1-carboxylate. Colorless oil which was used in the next step without further purification. MS (ESI): m/z=250.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-fluoro-5-(trifluoromethyl)phenoxy]pyrrolidine-1-carboxylate
  • To a solution of 2-fluoro-5-(trifluoromethyl)phenol (321 mg, 1.78 mmol), tert-butyl 3-hydroxypyrrolidine-1-carboxylate (334 mg, 1.78 mmol, CAS RN: 103057-44-9) and triphenylphosphine (467 mg, 1.78 mmol) in THF (5 mL) was added (E)-diazene-1,2-diylbis(piperidin-1-ylmethanone) (450 mg, 1.78 mmol, CAS RN 10465-81-3) in portions and the mixture was stirred at RT for 40 h. Silica gel was added to the suspension and it was evaporated. The compound was purified by silica gel chromatography on a 24 g column using an MPLC (ISCO) system eluting with a gradient of n-heptane:EtOAc (100:0 to 75:25) to provide the desired compound as a colorless oil (8.3%) which was used in the next step without further purification. MS (ESI): m/z=294.1 [M−56+H]+.
  • BB210 3-[2-Chloro-5-(trifluoromethyl)phenoxy]pyrrolidine; 4-methylbenzenesulfonic acid
  • The compound was prepared in analogy to BB95 from tert-butyl 3-[2-chloro-5-(trifluoromethyl)phenoxy]pyrrolidine-1-carboxylate. Colorless oil. MS (ESI): m/z=266.1 [M+H]+.
  • Step a) tert-Butyl 3-[2-chloro-5-(trifluoromethyl)phenoxy]pyrrolidine-1-carboxylate
  • The compound was prepared in analogy to BB209, step a, from 2-chloro-5-(trifluoromethyl)phenol and tert-butyl 3-hydroxypyrrolidine-1-carboxylate. Colorless solid which was used after chromatography without further purification. MS (ESI): m/z=310.1 [M−56+H]+.
  • BB211 3-[(E)-2-(2-fluoro-4-methyl-phenyl)vinyl]azetidine; 4-methylbenzenesulfonic acid
  • The compound was prepared in analogy to BB95 from tert-butyl 3-[(E)-2-(2-fluoro-4-methyl-phenyl)vinyl]azetidine-1-carboxylate and 4-methylbenzenesulfonic acid monohydrate. Colorless solid (87%). MS (ESI): m/z=192.2 [M+H]+.
  • Step a) 1-(Diethoxyphosphorylmethyl)-2-fluoro-4-methyl-benzene
  • The compound was prepared in analogy to BB206, step a, from 1-(bromomethyl)-2-fluoro-4-methylbenzene and triethyl phosphite followed by silica gel chromatography on a 40 g column using an MPLC (ISCO) system eluting with a gradient of n-heptane:EtOAc (100:0 to 0:100). Colorless liquid (85%). MS (ESI): m % z=261.1 [M+H]+.
  • Step b) tert-Butyl 3-[(E)-2-(2-fluoro-4-methyl-phenyl)vinyl]azetidine-1-carboxylate
  • The compound was prepared in analogy to example BB206, step b, from tert-butyl 3-formylazetidine-1-carboxylate and 1-(diethoxyphosphorylmethyl)-2-fluoro-4-methyl-benzene. Colorless oil (7%). MS (ESI): m % z=236.2 [M−56+H]+.

Claims (52)

1-56. (canceled)
57. A compound of formula (I):
Figure US20200308190A1-20201001-C00324
or a pharmaceutically acceptable salt thereof,
wherein:
(i) X is C—R3; m is 0 or 1; n is selected from 0, 1 and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CH2OCH2—, —CF2CH2—, —CH2═CH2—, —(CR16R17)q—CH2O—, and —CH2CF2—; or
(ii) X is N; m is 1; n is 1 or 2; and L is —(CH2)p— or —CF2CH2—;
p is selected from 1, 2, and 3;
q is 0 or 1;
A is selected from:
(i) C6-C14-aryl substituted with R4, R5, and R6;
(ii) 5- to 14-membered heteroaryl substituted with R7, R8, and R9; and
(iii) 3- to 14-membered heterocycloalkyl substituted with R10, R11, and R12;
R1 is hydrogen or C1-6-alkyl;
R2 is selected from hydrogen, C1-6-alkyl, and hydroxy-C1-6-alkyl;
R3 is selected from hydrogen, halogen, hydroxy, C1-6-alkoxy, C1-6-alkyl, and halo-C1-6-alkyl;
each of R4, R5, R6, R7, R8, R9, R10, R11 and R12 is independently selected from hydrogen, halogen, cyano, hydroxy, C1-6-alkyl, halo-C1-6-alkyl, hydroxy-C1-6-alkyl, C1-6-alkanoyl, halo-C1-5-alkyl-CH(OH)—, C1-6-alkoxy, halo-C1-6-alkoxy, SF5, CH3SO2, C3-10-cycloalkyl, C3-10-cycloalkyl substituted with R13, 3- to 14-membered heterocycloalkyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, C6-C14-aryl, C6-C14-aryloxy, halo-C6-C14-aryl, and halo-C6-C14-aryloxy;
each of R13, R14, and R15 is independently selected from C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkyl, halo-C1-6-alkoxy, halogen, and hydroxy; and
R16 and R17, taken together with the carbon atom to which they are attached, form a C3-10-cycloalkyl.
58. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ia):
Figure US20200308190A1-20201001-C00325
wherein A, L, X, m, n, R1, and R2 are as defined in claim 57.
59. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is a compound of formula (Ib):
Figure US20200308190A1-20201001-C00326
wherein A, L, X, m, n, R1, and R2 are as defined in claim 57.
60. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein:
(i) X is C—R3; m is 0 or 1; n is selected from 0, 1, and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —CF2CH2—, —CH2═CH2—, —(CR16R17)q—CH2O—, and —CH2OCH2—; or
(ii) X is N; m and n are both 1; and L is —(CH2)p—.
61. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein:
X is C—R3;
m and n are both 0; or
m and n are both 1; and
L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—.
62. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein p is 1 or 2.
63. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein p is 2.
64. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein q is 0 or 1.
65. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein q is 0.
66. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
(i) C6-C14-aryl substituted with R4, R5 and R6; and
(ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9.
67. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein A is selected from:
(i) phenyl substituted with R4, R5, and R6;
(ii) oxazolyl substituted with R7, R8, and R9; and
(iii) pyridyl substituted with R7, R8, and R9.
68. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen.
69. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen or C1-6-alkyl.
70. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen or methyl.
71. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl.
72. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, halogen, and C1-6-alkyl.
73. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from hydrogen, fluoro, and methyl.
74. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from hydrogen, halogen, hydroxy, cyano, C1-6-alkyl, C1-6-alkanoyl, SF5, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, 3- to 14-membered heterocyclyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, C6-C14-aryloxy, and halo-C6-C14-aryl.
75. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from halogen, SF5, C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, and 3- to 14-membered heterocycloalkyl.
76. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from chloro, SF5, methyl, methoxy, OCF3, CF3, cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl.
77. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, C1-6-alkoxy, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl, 5- to 14-membered heteroaryl, and halo-C6-C14-aryl.
78. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl, and halo-C6-C14-aryl.
79. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF3, pyrrolidinyl, cyclopentyl, cyclopropyl, and chlorophenyl.
80. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen or halogen.
81. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen.
82. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R7 is selected from hydrogen, C1-6-alkyl, C6-C14-aryl, halo-C6-C14-aryl, halo-C6-C14-aryloxy, and halo-C1-6-alkyl.
83. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R7 is C1-6-alkyl or halo-C1-6-alkyl.
84. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R7 is tert-butyl or CF3.
85. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R8 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl.
86. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or halo-C1-6-alkyl.
87. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R8 is hydrogen or CF3.
88. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R9 is hydrogen.
89. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R10 is halogen.
90. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R11 is halogen.
91. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R12 is hydrogen.
92. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R14 is selected from C1-6-alkyl, C1-6-alkoxy, and halogen.
93. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein R15 is hydrogen or halogen.
94. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein:
(i) X is C—R3; m is 0 or 1; n is selected from 0, 1, and 2; and L is selected from —(CH2)p—, —O—, —OCH2—, —(CR16R17)q—CH2O—, —CH2OCH2—, —CF2CH2—, and —CH2═CH2—; or
(ii) X is N; m and n are both 1; and L is —(CH2)p—;
p is 1 or 2;
q is 0 or 1;
A is selected from:
(i) C6-C14-aryl substituted with R4, R5, and R6;
(ii) 5- to 14-membered heteroaryl substituted with R7, R8, and R9; and
(iii) 3- to 14-membered heterocycloalkyl substituted with R10, R11, and R12;
R1 is hydrogen or C1-6-alkyl;
R2 is hydrogen or C1-6-alkyl;
R3 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl;
R4 is selected from hydrogen, halogen, cyano, SF5, C1-6-alkyl, C1-6-alkanoyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, 3- to 14-membered heterocycloalkyl, 3- to 14-membered heterocycloalkyl substituted with R14 and R15, 5- to 14-membered heteroaryl, halo-C6-C14-aryl, and C6-C14-aryloxy;
R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, C1-6-alkoxy, 3- to 14-membered heterocycloalkyl, C1-10-cycloalkyl, 5- to 14-membered heteroaryl, and halo-C6-C14-aryl;
R6 is hydrogen or halogen;
R7 is selected from hydrogen, C1-6-alkyl, C6-C14-aryl, halo-C6-C14-aryl, halo-C6-C14-aryloxy, and halo-C1-6-alkyl;
R8 is selected from hydrogen, halogen, C1-6-alkyl, and halo-C1-6-alkyl;
R9 is hydrogen;
R10 is halogen;
R11 is halogen;
R12 is hydrogen;
R14 is selected from halogen, C1-6-alkyl, and C1-6-alkoxy;
R15 is hydrogen or halogen; and
R16 and R17, taken together with the carbon atom to which they are attached, form a C3-10-cycloalkyl.
95. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein:
X is C—R3;
m and n are both 0; or
m and n are both 1;
L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
p is 1 or 2;
A is selected from:
(i) C6-C14-aryl substituted with R4, R5 and R6; and
(ii) 5- to 14-membered heteroaryl substituted with R7, R8 and R9;
R1 is hydrogen;
R2 is hydrogen or C1-6-alkyl;
R3 is selected from hydrogen, halogen and C1-6-alkyl;
R4 is selected from halogen, SF5, C1-6-alkyl, C1-6-alkoxy, halo-C1-6-alkoxy, halo-C1-6-alkyl, C3-10-cycloalkyl, and 3- to 14-membered heterocycloalkyl;
R5 is selected from hydrogen, cyano, halogen, C1-6-alkyl, halo-C1-6-alkyl, 3- to 14-membered heterocycloalkyl, C3-10-cycloalkyl and halo-C6-C14-aryl;
R6 is hydrogen;
R7 is C1-6-alkyl or halo-C1-6-alkyl;
R8 is hydrogen or halo-C1-6-alkyl; and
R9 is hydrogen.
96. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein:
X is C—R3;
m and n are both 0; or
m and n are both 1;
L is selected from —(CH2)p—, —O—, —OCH2— and —CH2O—;
p is 1 or 2;
A is selected from:
(i) phenyl substituted with R4, R5, and R6;
(ii) oxazolyl substituted with R7, R8, and R9; and
(iii) pyridyl substituted with R7, R8, and R9;
R1 is hydrogen;
R2 is hydrogen or methyl;
R3 is selected from hydrogen, fluoro and methyl;
R4 is selected from chloro, SF5, methyl, methoxy, OCF3, CF3, cyclopropyl, and 2-azaspiro[3.3]heptan-2-yl;
R5 is selected from hydrogen, cyano, fluoro, chloro, methyl, CF3, pyrrolidinyl, cyclopentyl, cyclopropyl and chlorophenyl;
R6 is hydrogen;
R7 is selected from tert-butyl, methyl, and CF3;
R8 is hydrogen or CF3; and
R9 is hydrogen.
97. A compound of claim 57, or a pharmaceutically acceptable salt thereof, selected from the compounds disclosed in Table 1.
98. A compound of claim 57, wherein the compound is:
(+)-(4aR,8aS)-6-(4-((4-(tert-Butyl)oxazol-2-yl)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)-(4aR,8aS)-6-[4-[[4-(Trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(+)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
rac-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4-fluorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-(4-(Trifluoromethoxy)benzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-(4-Chloro-3-fluorobenzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-(2-Chloro-4-(trifluoromethyl)phenoxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-(3-(Trifluoromethyl)phenoxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-[4-[[2-Chloro-4-(trifluoromethoxy)phenoxy]methyl]piperidine-1-carbonyl]-4,4a 5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethyl)phenoxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((4-Chloro-2-fluorophenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((4-Fluoro-2-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((2-Fluoro-4-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-(2-(Pyrrolidin-1-yl)-4-(trifluoromethyl)benzyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-4-((2-Chloro-4-(trifluoromethyl)phenoxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-[4-[[2-Cyclopentyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(+)- or (−)-3-Chloro-4-((1-((4aR,8aS)-3-oxooctahydro-2H-pyrido[4,3-b][1,4]oxazine-6-carbonyl)piperidin-4-yl)methoxy)benzonitrile;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4-(trifluoromethyl)phenoxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)-4-fluoropiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((4′,6-Dichloro-[1,1′-biphenyl]-3-yl)oxy)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(cis-4-((2-Chloro-4-fluorophenoxy)methyl)-3-methylpiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Chloro-4-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(3-((2-Fluoro-4-(trifluoromethoxy)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(+)- or (−)-(4aR,8aS)-6-(4-((2-Chloro-4-fluorophenoxy)methyl)-4-methylpiperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-[3-[(2,4-Dichlorophenyl)methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[4-[[2-Fluoro-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[4-[[2-Cyclopropyl-4-(trifluoromethyl)phenyl]methyl]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[[3-Chloro-4-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[[2-Fluoro-5-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[2-[2-Fluoro-6-(trifluoromethyl)phenyl]ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-(3-(2-fluoro-4-(trifluoromethyl)phenethyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
6-(3-((2,4-bis(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-[4-[3-chloro-4-(trifluoromethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-(3-methyl-4-(((5-methyl-6-(trifluoromethyl)pyridin-3-yl)oxy)methyl)piperidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-((3,4-dichlorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-((2,5-dichlorobenzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
rac-(4aR,8aS)-6(2-methyl-3-((4-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-(((4,5-bis(trifluoromethyl)pyridin-2-yl)oxy)methyl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
rac-(4aR,8aS)-6-(3-((2-fluoro-4-(trifluoromethyl)benzyl)oxy)-2-methyl azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-((2-fluoro-4-(pentafluoro-16-sulfaneyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-((4-methyl-2-(trifluoromethoxy)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-[4-[3-cyclopropyl-4-(trifluoromethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[2-(2-fluoro-4-methyl-phenyl)ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[2-[4-methoxy-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[3-(2-azaspiro[3.3]heptan-2-yl)-4-(trifluoromethyl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[2-[4-methyl-2-(trifluoromethyl)phenyl]ethyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-(3-((4-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
(4aR,8aS)-6-(3-((2-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one;
rac-(4aR,8aS)-6-[2-methyl-3-[[2-methyl-4-(trifluoromethoxy)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
rac-(4aR,8aS)-6-[2-methyl-3-[[2-methyl-3-(trifluoromethyl)phenyl]methoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-(4-chloro-3-cyclopropylphenoxy)azetidine-1-carbonyl]-4,4# a!,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[4-[2-chloro-3-(trifluoromethyl)phenoxy]piperidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-(2-chloro-3-cyclopropyl-phenoxy)azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[3-(2-azaspiro[3.3]heptan-2-yl)-2-chloro-phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[2-chloro-3-(5-oxa-2-azaspiro[3.5]nonan-2-yl)phenoxy]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-[3-[(E)-2-(2-fluoro-4-methyl-phenyl)vinyl]azetidine-1-carbonyl]-4,4a,5,7,8,8a-hexahydropyrido[4,3-b][1,4]oxazin-3-one;
(4aR,8aS)-6-(3-((E)-2-fluoro-6-(trifluoromethyl)styryl)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one; or
(4aR,8aS)-6-(3-((4-methyl-3-(trifluoromethyl)benzyl)oxy)azetidine-1-carbonyl)hexahydro-2H-pyrido[4,3-b][1,4]oxazin-3(4H)-one,
or a pharmaceutically acceptable salt thereof.
99. A process of manufacturing a compound of claim 57, or a pharmaceutically acceptable salt thereof, the process comprising:
reacting a first amine of formula 1, wherein R1 is as described in claim 57:
Figure US20200308190A1-20201001-C00327
with a second amine 2, wherein A, L, m, n, X and R2 are as described in claim 57:
Figure US20200308190A1-20201001-C00328
in the presence of a base and a urea forming reagent,
to form said compound of claim 57, or a pharmaceutically acceptable salt thereof.
100. A compound of claim 57, or a pharmaceutically acceptable salt thereof, when manufactured by a process comprising:
reacting a first amine of formula 1, wherein R1 is as described in claim 57:
Figure US20200308190A1-20201001-C00329
with a second amine 2, wherein A, L, m, n, X and R2 are as described in claim 57:
Figure US20200308190A1-20201001-C00330
in the presence of a base and a urea forming reagent,
to form said compound of claim 57, or a pharmaceutically acceptable salt thereof.
101. The compound of claim 57, or a pharmaceutically acceptable salt thereof, wherein said compound has an IC50 for monoacylglycerol lipase below 10 μM.
102. A pharmaceutical composition, comprising a compound of claim 57, or a pharmaceutically acceptable salt thereof, and a therapeutically inert carrier.
103. A pharmaceutical composition, comprising a compound of claim 98, or a pharmaceutically acceptable salt thereof, and a therapeutically inert carrier.
104. A method for the treatment or prophylaxis of a condition in a mammal, wherein the condition is neuroinflammation, a neurodegenerative disease, pain, cancer, or a mental disorder, the method comprising administering an effective amount of a compound of claim 57, or a pharmaceutically acceptable salt thereof, to the mammal.
105. A method for the treatment or prophylaxis of a condition in a mammal, wherein the condition is neuroinflammation, a neurodegenerative disease, pain, cancer, or a mental disorder, the method comprising administering an effective amount of a compound of claim 98, or a pharmaceutically acceptable salt thereof, to the mammal.
106. A method for the treatment or prophylaxis of a condition in a mammal, wherein the condition is multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain, or spasticity associated with pain, the method comprising administering an effective amount of a compound of claim 57, or a pharmaceutically acceptable salt thereof, to the mammal.
107. A method for the treatment or prophylaxis of a condition in a mammal, wherein the condition is multiple sclerosis, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, traumatic brain injury, neurotoxicity, stroke, epilepsy, anxiety, migraine, depression, hepatocellular carcinoma, colon carcinogenesis, ovarian cancer, neuropathic pain, chemotherapy induced neuropathy, acute pain, chronic pain, or spasticity associated with pain, the method comprising administering an effective amount of a compound of claim 98, or a pharmaceutically acceptable salt thereof, to the mammal.
US16/844,262 2018-08-13 2020-04-09 Heterocyclic compounds as monoacylglycerol lipase inhibitors Abandoned US20200308190A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/749,496 US11802133B2 (en) 2018-08-13 2022-05-20 Heterocyclic compounds as monoacylglycerol lipase inhibitors
US18/475,065 US20240150373A1 (en) 2018-08-13 2023-09-26 Heterocyclic compounds as monoacylglycerol lipase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP18188681.3 2018-08-13
EP18188681 2018-08-13
PCT/EP2019/071520 WO2020035424A1 (en) 2018-08-13 2019-08-12 New heterocyclic compounds as monoacylglycerol lipase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/071520 Continuation WO2020035424A1 (en) 2018-08-13 2019-08-12 New heterocyclic compounds as monoacylglycerol lipase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/749,496 Continuation US11802133B2 (en) 2018-08-13 2022-05-20 Heterocyclic compounds as monoacylglycerol lipase inhibitors

Publications (1)

Publication Number Publication Date
US20200308190A1 true US20200308190A1 (en) 2020-10-01

Family

ID=63244467

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/844,262 Abandoned US20200308190A1 (en) 2018-08-13 2020-04-09 Heterocyclic compounds as monoacylglycerol lipase inhibitors
US17/749,496 Active US11802133B2 (en) 2018-08-13 2022-05-20 Heterocyclic compounds as monoacylglycerol lipase inhibitors
US18/475,065 Pending US20240150373A1 (en) 2018-08-13 2023-09-26 Heterocyclic compounds as monoacylglycerol lipase inhibitors

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/749,496 Active US11802133B2 (en) 2018-08-13 2022-05-20 Heterocyclic compounds as monoacylglycerol lipase inhibitors
US18/475,065 Pending US20240150373A1 (en) 2018-08-13 2023-09-26 Heterocyclic compounds as monoacylglycerol lipase inhibitors

Country Status (22)

Country Link
US (3) US20200308190A1 (en)
EP (1) EP3837263B1 (en)
JP (1) JP2021534139A (en)
KR (1) KR20210045992A (en)
CN (1) CN112867720A (en)
AR (1) AR116666A1 (en)
AU (1) AU2019322538B2 (en)
BR (1) BR112021002298A2 (en)
CA (1) CA3104928A1 (en)
CL (1) CL2021000362A1 (en)
CO (1) CO2021002382A2 (en)
CR (1) CR20210077A (en)
IL (1) IL279173A (en)
MA (1) MA53219A (en)
MX (1) MX2021001433A (en)
PE (1) PE20211089A1 (en)
PH (1) PH12021500013A1 (en)
RU (1) RU2769507C1 (en)
SG (1) SG11202013064WA (en)
TW (1) TWI812770B (en)
UA (1) UA127629C2 (en)
WO (1) WO2020035424A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11390610B2 (en) 2017-10-10 2022-07-19 Hoffmann-La Roche Inc. Piperazine derivatives as MAGL inhibitors
US11420961B2 (en) 2017-11-28 2022-08-23 Hoffmann-La Roche Inc. Heterocyclic compounds
CN115335365A (en) * 2020-03-26 2022-11-11 詹森药业有限公司 Aminocyclobutane as monoacylglycerol lipase modulators
US11608347B2 (en) 2018-01-08 2023-03-21 Hoffmann-La Roche Inc. Octahydropyrido[1,2-alpha]pyrazines as MAGL inhibitors
US11802133B2 (en) 2018-08-13 2023-10-31 Hoffmann-La Roche Inc. Heterocyclic compounds as monoacylglycerol lipase inhibitors
US11814375B2 (en) 2019-09-12 2023-11-14 Hoffmann-La Roche Inc. Heterocyclic compounds
US11981661B2 (en) 2020-09-03 2024-05-14 Hoffmann-La Roche Inc. Heterocyclic compounds

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190292188A1 (en) 2018-02-27 2019-09-26 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as a2a / a2b inhibitors
CA3100731A1 (en) 2018-05-18 2019-11-21 Incyte Corporation Fused pyrimidine derivatives as a2a / a2b inhibitors
CA3105721A1 (en) 2018-07-05 2020-01-09 Incyte Corporation Fused pyrazine derivatives as a2a / a2b inhibitors
TWI829857B (en) 2019-01-29 2024-01-21 美商英塞特公司 Pyrazolopyridines and triazolopyridines as a2a / a2b inhibitors
MX2022002720A (en) * 2019-09-05 2022-08-10 Lunan Pharmaceutical Group Corp Magl inhibitor, preparation method therefor and use thereof.
US20210094971A1 (en) * 2019-09-09 2021-04-01 Hoffmann-La Roche Inc. Heterocyclic compounds
CN114401969A (en) * 2019-09-23 2022-04-26 豪夫迈·罗氏有限公司 Heterocyclic compounds
US20210094973A1 (en) * 2019-09-24 2021-04-01 Hoffmann-La Roche Inc. Heterocyclic compounds
WO2021058444A1 (en) * 2019-09-24 2021-04-01 F. Hoffmann-La Roche Ag Heterocyclic compounds
BR112022006507A2 (en) * 2019-10-07 2022-06-28 De Shaw Res Llc ARYLMETHYLENE HETEROCYCLIC COMPOUNDS AS KV1.3 POTASSIUM AGITATOR CHANNEL BLOCKERS

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5214038A (en) * 1991-04-15 1993-05-25 Hoechst-Roussel Pharmaceuticals Inc. 1-(pyrido[3,4-b]-1,4-oxazinyl-4-yl)-1H-indoles and intermediates for the preparation thereof
US7160888B2 (en) * 2003-08-22 2007-01-09 Warner Lambert Company Llc [1,8]naphthyridin-2-ones and related compounds for the treatment of schizophrenia
US7579495B2 (en) 2003-12-19 2009-08-25 Momentive Performance Materials Inc. Active-releasing cyclic siloxanes
US7842810B2 (en) 2005-03-31 2010-11-30 Janssen Pharmaceutica, Nv Bicyclic pyrazole compounds as antibacterial agents
WO2007002057A1 (en) 2005-06-20 2007-01-04 Schering Corporation Carbon-linked substituted piperidines and derivatives thereof useful as histamine h3 antagonists
EP1945222B1 (en) 2005-11-02 2012-12-26 Bayer Pharma Aktiengesellschaft Pyrrolo[2,1-f] [1,2,4]-triazin-4-ylamines as igf-1r kinase inhibitors for the treatment of cancer and other hyperproliferative diseases
WO2007098418A1 (en) 2006-02-17 2007-08-30 Memory Pharmaceuticals Corporation Compounds having 5-ht6 receptor affinity
US7872028B2 (en) 2006-04-05 2011-01-18 Vitae Pharmaceuticals, Inc. Diaminopropanol renin inhibitors
KR102107954B1 (en) 2007-03-12 2020-05-07 인트라-셀룰라 써래피스, 인코퍼레이티드. Substituted heterocycle fused gamma-carbolines synthesis
FR2915197B1 (en) * 2007-04-18 2009-06-12 Sanofi Aventis Sa TRIAZOLOPYRIDINE CARBOXAMIDE DERIVATIVES, PREPARATION THEREOF AND THERAPEUTIC USE THEREOF.
TW200942537A (en) 2008-02-01 2009-10-16 Irm Llc Compounds and compositions as kinase inhibitors
MX2011003951A (en) 2008-10-29 2011-05-03 Hoffmann La Roche Novel phenyl amide or pyridil amide derivatives and their use as gpbar1 agonists.
BR112012005382A2 (en) 2009-09-10 2016-03-29 Hoffmann La Roche jak inhibitors
WO2011059118A1 (en) 2009-11-10 2011-05-19 Kim Hyun Jeen System for testing olfactory perception
WO2011058766A1 (en) * 2009-11-16 2011-05-19 Raqualia Pharma Inc. Aryl carboxamide derivatives as ttx-s blockers
US8420661B2 (en) 2010-04-13 2013-04-16 Hoffmann-La Roche Inc. Arylethynyl derivatives
US9493451B2 (en) 2011-05-16 2016-11-15 Bionomics Limited Amine derivatives as potassium channel blockers
US20140288323A1 (en) 2011-10-19 2014-09-25 The Trustees Of Columbia University In The City Of New York Cyclopropenimine catalyst compositions and processes
US9029368B2 (en) 2011-11-30 2015-05-12 Actelion Pharmaceuticals Ltd. 3,7-disubstituted octahydro-2H-pyrido[4,3-E][1,3]oxazin-2-one antibiotics
PL2794600T3 (en) 2011-12-22 2018-06-29 Novartis Ag 2,3-Dihydro-benzo[1,4]oxazine derivatives and related compounds as phosphoinositide-3 kinase (PI3K) inhibitors for the treatment of e.g. rheumatoid arthritis
LT2800565T (en) 2012-01-06 2020-07-27 Lundbeck La Jolla Research Center, Inc. Carbamate compounds and methods of making and using same
GB201209587D0 (en) 2012-05-30 2012-07-11 Takeda Pharmaceutical Therapeutic compounds
TW201422625A (en) 2012-11-26 2014-06-16 Novartis Ag Solid form of dihydro-pyrido-oxazine derivative
MX362338B (en) 2013-04-16 2019-01-11 Idorsia Pharmaceuticals Ltd Antibacterial biaromatic derivatives.
WO2016109501A1 (en) 2014-12-30 2016-07-07 Karos Pharmaceuticals, Inc. Amide compounds as tryptophan hydroxylase inhibitors
US10106556B2 (en) 2015-03-30 2018-10-23 Takeda Pharmaceutical Company Limited Heterocyclic compound
TW201713641A (en) 2015-05-13 2017-04-16 賽爾維他股份公司 Substituted quinoxaline derivatives
KR20180005250A (en) 2015-05-21 2018-01-15 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Benzimidazole derivatives as PAD4 inhibitors
US10919875B2 (en) 2015-06-18 2021-02-16 89Bio Ltd Substituted 4-benzyl and 4-benzoyl piperidine derivatives
WO2017087858A1 (en) 2015-11-20 2017-05-26 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
US10323038B2 (en) 2015-11-20 2019-06-18 Abide Therapeutics, Inc. Pyrazole compounds and methods of making and using same
WO2017170830A1 (en) 2016-03-31 2017-10-05 武田薬品工業株式会社 Heterocyclic compound
CN109715613B (en) 2016-03-31 2021-07-30 武田药品工业株式会社 Heterocyclic compounds
EP3478678A1 (en) 2016-06-30 2019-05-08 Basilea Pharmaceutica International AG Mitochondrial inhibitors for the treatment of proliferation disorders
JP6880243B2 (en) 2017-06-12 2021-06-02 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Heteroaromatic compounds as vanine inhibitors
WO2019065791A1 (en) 2017-09-29 2019-04-04 武田薬品工業株式会社 Heterocyclic compound
AR114136A1 (en) 2017-10-10 2020-07-29 Hoffmann La Roche HETEROCYCLIC COMPOUNDS
EP3717477B1 (en) 2017-11-28 2022-07-20 F. Hoffmann-La Roche AG New heterocyclic compounds
TW201930300A (en) 2017-12-15 2019-08-01 瑞士商赫孚孟拉羅股份公司 New heterocyclic compounds
TW201938164A (en) 2018-01-08 2019-10-01 瑞士商赫孚孟拉羅股份公司 New heterocyclic compounds
HUE061584T2 (en) 2018-03-22 2023-07-28 Hoffmann La Roche Oxazine monoacylglycerol lipase (magl) inhibitors
JP2021533093A (en) 2018-08-13 2021-12-02 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft A novel heterocyclic compound as a monoacylglycerol lipase inhibitor
WO2020035424A1 (en) 2018-08-13 2020-02-20 F. Hoffmann-La Roche Ag New heterocyclic compounds as monoacylglycerol lipase inhibitors
JP2022508203A (en) 2018-11-22 2022-01-19 キル・レガー・セラピューティクス・インコーポレーテッド GLP-1R agonist and its use
WO2020104494A1 (en) 2018-11-22 2020-05-28 F. Hoffmann-La Roche Ag New heterocyclic compounds
CN113767105A (en) 2019-04-09 2021-12-07 豪夫迈·罗氏有限公司 Heterocyclic compounds as inhibitors of monoacylglycerol lipases (MAGL)
WO2021001330A1 (en) 2019-07-03 2021-01-07 F. Hoffmann-La Roche Ag Heterocyclic monoacylglycerol lipase (magl) inhibitors
WO2021005034A1 (en) 2019-07-09 2021-01-14 F. Hoffmann-La Roche Ag New heterocyclic compounds
US20210094971A1 (en) 2019-09-09 2021-04-01 Hoffmann-La Roche Inc. Heterocyclic compounds
WO2021048241A1 (en) 2019-09-12 2021-03-18 F. Hoffmann-La Roche Ag Radiolabeled compounds
KR20220062515A (en) 2019-09-12 2022-05-17 에프. 호프만-라 로슈 아게 4,4A,5,7,8,8A-hexapyrido[4,3-B][1,4]oxazin-3-one compounds as MAGL inhibitors
CN114401969A (en) 2019-09-23 2022-04-26 豪夫迈·罗氏有限公司 Heterocyclic compounds
US20210094973A1 (en) 2019-09-24 2021-04-01 Hoffmann-La Roche Inc. Heterocyclic compounds
CN114514233A (en) 2019-09-24 2022-05-17 豪夫迈·罗氏有限公司 Fluorescent probes for monoacylglycerol lipases (MAGL)
WO2021058444A1 (en) 2019-09-24 2021-04-01 F. Hoffmann-La Roche Ag Heterocyclic compounds
CR20230115A (en) 2020-09-03 2023-04-11 Hoffmann La Roche HETEROCYCLIC COMPOUNDS

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11390610B2 (en) 2017-10-10 2022-07-19 Hoffmann-La Roche Inc. Piperazine derivatives as MAGL inhibitors
US11420961B2 (en) 2017-11-28 2022-08-23 Hoffmann-La Roche Inc. Heterocyclic compounds
US11608347B2 (en) 2018-01-08 2023-03-21 Hoffmann-La Roche Inc. Octahydropyrido[1,2-alpha]pyrazines as MAGL inhibitors
US11802133B2 (en) 2018-08-13 2023-10-31 Hoffmann-La Roche Inc. Heterocyclic compounds as monoacylglycerol lipase inhibitors
US11814375B2 (en) 2019-09-12 2023-11-14 Hoffmann-La Roche Inc. Heterocyclic compounds
CN115335365A (en) * 2020-03-26 2022-11-11 詹森药业有限公司 Aminocyclobutane as monoacylglycerol lipase modulators
US11981661B2 (en) 2020-09-03 2024-05-14 Hoffmann-La Roche Inc. Heterocyclic compounds

Also Published As

Publication number Publication date
RU2769507C1 (en) 2022-04-01
CA3104928A1 (en) 2020-02-20
WO2020035424A1 (en) 2020-02-20
BR112021002298A2 (en) 2021-05-04
AU2019322538A1 (en) 2021-01-07
AU2019322538B2 (en) 2021-09-30
CL2021000362A1 (en) 2021-07-09
EP3837263A1 (en) 2021-06-23
CN112867720A (en) 2021-05-28
US20240150373A1 (en) 2024-05-09
SG11202013064WA (en) 2021-01-28
MA53219A (en) 2021-11-17
US20230050901A1 (en) 2023-02-16
EP3837263B1 (en) 2024-07-03
PE20211089A1 (en) 2021-06-14
KR20210045992A (en) 2021-04-27
UA127629C2 (en) 2023-11-08
TW202021970A (en) 2020-06-16
TWI812770B (en) 2023-08-21
CO2021002382A2 (en) 2021-03-08
CR20210077A (en) 2021-03-11
IL279173A (en) 2021-01-31
AR116666A1 (en) 2021-06-02
JP2021534139A (en) 2021-12-09
MX2021001433A (en) 2021-04-12
PH12021500013A1 (en) 2021-09-13
US11802133B2 (en) 2023-10-31

Similar Documents

Publication Publication Date Title
US11802133B2 (en) Heterocyclic compounds as monoacylglycerol lipase inhibitors
US20210277020A1 (en) Heterocyclic compounds as monoacylglycerol lipase inhibitors
EP3883936B1 (en) New heterocyclic compounds
US20210107920A1 (en) Oxazine monoacylglycerol lipase (magl) inhibitors
EP4028403B1 (en) 4,4a,5,7,8,8a-hexapyrido[4,3-b][1,4]oxazin-3-one compounds as magl inhibitors
EP3953360B1 (en) Heterocyclic compounds as inhibitors of monoacylglycerol lipase (magl)
US20210094973A1 (en) Heterocyclic compounds
US11981661B2 (en) Heterocyclic compounds
US20220213093A1 (en) Heterocyclic monoacylglycerol lipase (magl) inhibitors
US20220242876A1 (en) Heterocyclic compounds
RU2801190C2 (en) New heterocyclic compounds as monoacylglycerol lipase inhibitors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BELL, CHARLES;BENZ, JOERG;GOBBI, LUCA;AND OTHERS;SIGNING DATES FROM 20180913 TO 20181121;REEL/FRAME:056389/0075

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KURATLI, MARTIN;REEL/FRAME:056389/0177

Effective date: 20190809

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:056389/0245

Effective date: 20190809

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION