US20210338653A1 - N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases - Google Patents

N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases Download PDF

Info

Publication number
US20210338653A1
US20210338653A1 US17/239,726 US202117239726A US2021338653A1 US 20210338653 A1 US20210338653 A1 US 20210338653A1 US 202117239726 A US202117239726 A US 202117239726A US 2021338653 A1 US2021338653 A1 US 2021338653A1
Authority
US
United States
Prior art keywords
ethyl
carboxamide
benzylpiperidin
methyl
piperazine
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
US17/239,726
Inventor
Nicole HARRIOTT
Nicholas Pagano
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.)
Neurocrine Biosciences Inc
Original Assignee
Neurocrine Biosciences 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 Neurocrine Biosciences Inc filed Critical Neurocrine Biosciences Inc
Priority to US17/239,726 priority Critical patent/US20210338653A1/en
Assigned to NEUROCRINE BIOSCIENCES, INC. reassignment NEUROCRINE BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HARRIOTT, NICOLE, PAGANO, Nicholas
Publication of US20210338653A1 publication Critical patent/US20210338653A1/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61P25/28Drugs 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/26Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • muscarinic receptor 4 M4
  • M4 muscarinic receptor 4
  • Muscarinic acetylcholine receptors are autonomic receptors that form G protein-receptor complexes in the cell membranes of certain neurons and other cell types (e.g., endothelial cells of blood vessels). Muscarinic receptors are located postsynaptically at the parasympathetic neuroeffector junction, from where the receptors function to increase or decrease the activity of the effector cells. Extrapyramidal symptoms are observed in patients treated with antipsychotic therapeutics and in patients who have neuroleptic malignant syndrome, brain damage (e.g., athetotic cerebral palsy), encephalitis, and meningitis.
  • Drugs other than antipsychotics also cause extrapyramidal symptoms, for example antidopaminergic drugs (e.g., the antiemetic metoclopramide and the antidepressant amoxapine) and selective serotonin reuptake inhibitors (SSR*), which indirectly decrease dopamine.
  • Conditions associated with extrapyramidal symptoms include acute dystonic reactions, akathisia, pseudoparkinsonism, and tardive dyskinesia.
  • Extrapyramidal symptoms caused by antipsychotic therapeutics are being treated with anticholinergic drugs that lack selectivity for any of the five muscarinic receptor subtypes (see, e.g., Erosa-Rivero et al., Neuropharmacology 81:176-87 (2014)). Because anticholinergic drugs that effect multiple muscarinic receptors may cause distinct and in certain instances opposing effects, therapeutics that exhibit selectivity for particular receptors are desired.
  • compounds that antagonize muscarinic receptors are provided that selectively antagonize muscarinic receptor 4 (M4).
  • M4 muscarinic receptor 4
  • Such compounds are useful in the treatment of a number of diseases and/or disorders, particularly neurological conditions, diseases, and disorders including cognitive disorders such as Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia.
  • methods are provided for treating or preventing movement disorders which may include Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, particularly chorea associate with Huntington's disease.
  • A, B, C, L 1 , L 2 , R 1 , R 2 , R 3 , R 4 , R 5 , w, x, y and z are as defined below. More specific embodiments are also described within Tables 1-14, as well as the more specific formulas noted herein below.
  • compositions comprising a compound of formula (I), including one or more of the specific compounds described herein, and at least one pharmaceutically acceptable excipient.
  • the compounds, as well as the pharmaceutical compositions comprising the compounds may be used for antagonizing a muscarinic receptor, such as muscarinic receptor 4 (M4).
  • M4 muscarinic receptor 4
  • the compound is a selective M4 antagonist.
  • a non-human animal may refer to one or more non-human animals, or a plurality of such animals
  • reference to “a cell” or “the cell” includes reference to one or more cells and equivalents thereof (e.g., plurality of cells) known to those skilled in the art, and so forth.
  • M4 muscarinic receptor 4
  • A, B and C are each independently a carbocycle or heterocycle
  • R 1 is, at each occurrence, H, C 1-4 alkyl, C( ⁇ O)OC 1-4 alkyl or aryl;
  • R 2 , R 3 , R 4 and R 5 are each independently —OH, —NH 2 , —NH(C 1-4 alkyl), —N(C 1-4 alkyl) 2 , —C ⁇ N, —C( ⁇ O)NH 2 , halo, C 1-4 alkyl, C 1-4 alkylOH, C 1-4 haloalkyl, C 1-4 alkoxy or C 1-4 haloalkoxy;
  • w, x, y and z are each independently 0, 1, 2 or 3;
  • L 1 is a heteroalkylene linker having at least one N, O or S heteroatom, and wherein the heteroalkylene may be a straight chain or cyclized and optionally substituted with oxo, —OH, C 1-4 alkyl or C 1-4 alkoxy; and
  • L 2 is an optional linker that is not present or, when present, is —O(CH 2 ) m — where m is 0 or 1.
  • R 1 is H at both occurrences.
  • one R 1 is H and the other R 1 is methyl.
  • R 1 is methyl at both occurrences.
  • A is a non-aromatic carbocyle, and more specifically cyclohexyl.
  • A is an aromatic carbocyle, and more specifically aryl.
  • A is phenyl or naphthyl.
  • A is phenyl
  • A is an aromatic heterocycle, and more specifically one of the following:
  • w is 0 and R 2 is not present.
  • w is 1, 2 or 3, and R 2 is at each occurrence —OH, —CN, halo, or C 1-4 alkyl.
  • x is 0 and R 3 is not present.
  • x is 1 or 2 and R 3 is at each occurrence —OH or C 1-4 alkyl-OH.
  • L 1 is a a heteroalkylene linker having at least one N or O heteroatom, and wherein the heteroalkylene may be a straight chain or cyclized and optionally substituted with oxo, —OH, C 1-4 alkyl or C 1-4 alkoxy, and more specifically one of the following:
  • B is a non-aromatic carbocyle, and more specifically cyclohexyl.
  • B is an aromatic carbocyle, and more specifically aryl.
  • B is phenyl or naphthyl.
  • B is phenyl
  • B is a non-aromatic heterocycle.
  • B is piperazinyl, piperadinyl or pyrrolidinyl, and more specifically one of the following:
  • B an aromatic heterocycle, and more specifically
  • y is 0 and R 4 is not present.
  • y is 1 and R 4 is methyl.
  • B is piperazinyl, y is 1 and R 4 is methyl and, in a more specific embodiment, such moiety has the following structure:
  • y is 2 and R 4 at both occurrences is methyl.
  • B is piperazinyl, y is 2 and R 4 at both occurrences is methyl and, in a more specific embodiment, such moiety has one of the following structures:
  • L 2 is not present.
  • L 2 is —O—
  • L 2 is —OCH 2 —.
  • C is an aromatic 5-12 membered carbocycle or heterocycle.
  • C is an aromatic carbocyle, and more specifically aryl.
  • C is phenyl
  • C is an aromatic heterocycle, and more specifically one of the following:
  • z is 0 and R 5 is not present.
  • z is 1, 2 or 3, and each occurrence of R 5 is independently-OH, —NH 2 , —NH(C 1-4 alkyl), —N(C 1-4 alkyl) 2 , —CN, halo, C 1-4 alkyl, C 1-4 alkyl-OH, C 1-4 haloalkyl, C 1-4 alkoxy or C 1-4 haloalkoxy.
  • compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 0.
  • compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 1 or 2, and R 4 is at each occurrence C 1-4 alkyl.
  • compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 1 or 2, and R 4 is at each occurrence methyl.
  • compounds are provided of formula (XV), (XVI) or (XVII) wherein —C(R 5 ) z is one of the following:
  • z is 0 and R 5 is not present.
  • z is 1, 2 or 3, and each occurrence of R 5 is independently-OH, —NH 2 , —NH(C 1-4 alkyl), —N(C 1-4 alkyl) 2 , —C ⁇ N, halo, C 1-4 alkyl, C 1-4 alkyl-OH, C 1-4 haloalkyl, C 1-4 alkoxy or C 1-4 haloalkoxy.
  • a compound of formula (I) is one or more of the following compounds (with cal. ion m/z listed in parentheses):
  • compositions that comprise a compound of Formula I, including one or more of the specific compounds described herein (see, e.g., Tables 1-19), and at least one pharmaceutically acceptable excipient.
  • a method for antagonizing a muscarinic receptor in a cell comprising contacting the cell and a compound of Formula I, including specific compounds described herein, for a time sufficient and under appropriate conditions to permit interaction between the cell and the compound.
  • the cell is in a subject who is in need of treatment with a compound disclosed herein.
  • the subject may have or be at risk for developing a neurological disease, condition, or disorder including cognitive and movement neurological diseases, conditions, and disorders.
  • methods are provided for preventing (i.e., reducing the likelihood of occurrence of) or treating Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia; Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, including chorea associate with Huntington's disease.
  • Parkinson's Disease drug induced Parkinsonism
  • dyskinesias dystonia
  • chorea levodopa induced dyskinesia
  • Huntington's disease including chorea associate with Huntington's disease.
  • a person skilled in the medical or neurological art will readily appreciate that many of the aforementioned neurological diseases have both cognitive deficits and movement deficiencies or difficulties associated with them.
  • C 1 -C 4 alkyl describes an alkyl group, as defined below, having a total of 1 to 4 carbon atoms
  • C 4 -C 12 cycloalkylalkyl describes a cycloalkylalkyl group, as defined below, having a total of 4 to 12 carbon atoms.
  • the total number of carbons in the shorthand notation does not include carbons that may exist in substituents of the group described. For example, the following terms have the meaning indicated.
  • C 1 -C 6 alkyl refers to an alkyl radical as defined below containing one to six carbon atoms. The C 1 -C 6 alkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 1 -C 4 alkyl refers to an alkyl radical as defined below containing one to four carbon atoms. The C 1 -C 4 alkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 2 -C 4 alkenyl refers to an alkenyl radical as defined below containing two to six carbon atoms.
  • the C 2 -C 12 alkenyl radical may be optionally substituted as defined below for an alkenyl group.
  • C 2 -C 6 alkynyl refers to an alknyl radical as defined below containing two to six carbon atoms.
  • the C 2 -C 12 alknyl radical may be optionally substituted as defined below for an alkenyl group.
  • C 1 -C 4 alkoxy refers to an alkoxy radical as defined below containing one to twelve carbon atoms.
  • the alkyl part of the C 1 -C 4 alkoxy radical may be optionally substituted as defined below for an alkyl group.
  • C 2 -C 6 alkoxyalkyl refers to an alkoxyalkyl radical as defined below containing two to six carbon atoms. Each alkyl part of the C 2 -C 6 alkoxyalkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 7 -C 12 aralkyl refers to an aralkyl group as defined below containing seven to twelve carbon atoms.
  • the aryl part of the C 7 -C 12 aralkyl radical may be optionally substituted as described below for an awl group.
  • the alkyl part of the C 7 -C 12 aralkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 7 -C 12 aralkenyl refers to an aralkenyl group as defined below containing seven to twelve carbon atoms.
  • the aryl part of the C 7 -C 12 aralkenyl radical may be optionally substituted as described below for an aryl group.
  • the alkenyl part of the C 7 -C 12 aralkenyl radical may be optionally substituted as defined below for an alkenyl group.
  • C 3 -C 12 cycloalkyl refers to a cycloalkyl radical as defined below having three to twelve carbon atoms.
  • the C 3 -C 12 cycloalkyl radical may be optionally substituted as defined below for a cycloalkyl group.
  • C 4 -C 12 cycloalkylalkyl refers to a cycloalkylalkyl radical as defined below having four to twelve carbon atoms.
  • the C 4 -C 12 cycloalkylalkyl radical may be optionally substituted as defined below for a cycloalkylalkyl group.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, one to eight carbon atoms, or one to six carbon atoms, or one to four carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, and the like.
  • Alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably two to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like.
  • Alkylene or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms or from one to four carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • Alkenylene or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like.
  • the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • the points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • Carbocyclyl refers to a stable 3- to 18-membered aromatic or non-aromatic ring radical which consists of 3 to 18 carbon atoms. Unless stated otherwise specifically in the specification, the carbocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems, and may be partially or fully saturated. Non-aromatic carbocyclyl radicals include cycloalkyl, while aromatice carbocyclyl redicals include aryl.
  • Cycloalkyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptly, and cyclooctyl.
  • Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo-[2.2.1]heptanyl, and the like.
  • Aryl refers to a hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring.
  • the aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene.
  • aryl is phenyl or naphthyl, and in another embodiment is phenyl.
  • Heterocyclyl refers to a stable 3- to 18-membered aromatic or non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated.
  • heteroaryl radicals examples include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, pyrazolopyrimidinyl, quinuclidiny
  • Heteroaryl refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, benzoxazolinonyl, benzimidazolthionyl, carbazolyl, cinnolin
  • each of alkyl, alkenyl, alkylene, alkenylene, carbocyclyl, cycloalkyl, aryl, heterocyclyl and heteroaryl as defined above may be optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, —R 41 —OR 40 , —R 41 —OC(O)—R 40 , —R 41 —N(R 40 ) 2 , —R 41 —C(O)R 40 , —R 41 —C(O)OR 40 , —R 41 —C(O)N(R
  • Amino refers to the —NH 2 radical.
  • Haldroxy refers to the —OH radical.
  • Niro refers to the —NO 2 radical.
  • Oxo refers to the ⁇ O substituent.
  • Thioxo refers to the ⁇ S substituent.
  • Trifluoromethyl refers to the —CF 3 radical.
  • Trifluoromethoxy refers to the —OCF 3 radical.
  • acyl refers to a radical —C(O)R, wherein R is alkyl, aralkyl, carbocyclyl, aryl, heteroaryl, or heterocyclyl, as defined herein.
  • R is methyl
  • the acyl group is also referred to as acetyl.
  • Heteroalkylene or “heteroalkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting of carbon and hydrogen and at least one heteroatom selected from N, O, and S.
  • Alkoxy refers to a radical of the formula —OR a where R a is an alkyl, or haloalkyl radical as defined above containing one to six carbon atoms.
  • Representative alkoxy groups include methoxy and ethoxy. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.
  • An alkoxy that is substituted with halo may be called herein a haloalkoxy, which includes for example trifluoromethoxy, trichloromethoxy and the like.
  • Heteroalkenylene or “heteroalkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting of carbon and hydrogen and at least one heteroatom selected from N, O, and S.
  • Alkyl refers to a radical of the formula —R b —R e where R b is an alkylene chain as defined above and R, is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like.
  • the alkylene chain part of the aralkyl radical may be optionally substituted as described above for an alkylene chain.
  • the aryl part of the aralkyl radical may be optionally substituted as described above for an aryl group.
  • Cycloalkylalkyl refers to a radical of the formula —R b R g where R b is an alkylene chain as defined above and R g is a cycloalkyl radical as defined above.
  • the alkylene chain and the cycloalkyl radical may be optionally substituted as defined above.
  • fused refers to any ring system described herein which is fused to an existing ring structure in the compounds of the invention.
  • the fused ring system is a heterocyclyl or a heteroaryl, any carbon in the existing ring structure which becomes part of the fused ring system may be replaced with a nitrogen.
  • Halo refers to bromo, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like.
  • the alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Haloalkenyl refers to an alkenyl radical, as defined above, that is substituted by one or more halo radicals, as defined above.
  • the alkenyl part of the haloalkyl radical may be optionally substituted as defined above for an alkenyl group.
  • Haloalkoxy refers to an alkoxy radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethoxy, difluoromethoxy, trichloromethoxy, 2,2,2-trifluoroethoxy, 3-bromo-2-fluoropropyloxy, and the like.
  • the alkoxy part of the haloalkoxy radical may be optionally substituted as defined above for an alkoxy group.
  • N-heterocyclyl refers to a heterocyclyl radical as defined above containing at least one nitrogen.
  • An N-heterocyclyl radical may be optionally substituted as described above for heterocyclyl radicals.
  • Heterocyclylalkyl refers to a radical of the formula —R b R h where R b is an alkylene chain as defined above and R h is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heterocyclylalkyl radical may be optionally substituted as defined above for an alkyene chain.
  • the heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
  • N-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical.
  • An N-heteroaryl radical may be optionally substituted as described above for heteroaryl radicals.
  • Heteroarylalkyl refers to a radical of the formula —R b R i where R b is an alkylene chain as defined above and R t is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkylene chain part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkylene chain.
  • Hydroalkyl refers to a radical of the formula —R b OH where R b is an alkylene chain as defined above.
  • the —OH group can be attached to any carbon in the alkylene chain.
  • the alkylene chain part of the heteroarylalkyl radical may additionally be optionally substituted as defined above for an alkylene chain.
  • the compounds described herein may generally be used as the free acid or free base. Alternatively, the compounds may be used in the form of acid or base addition salts. Acid addition salts of the free amino compounds may be prepared by methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic, acetic, trifluoroacetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids.
  • Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids.
  • Base addition salts included those salts that form with the carboxylate anion and include salts formed with organic and inorganic cations such as those chosen from the alkali and alkaline earth metals (for example, lithium, sodium, potassium, magnesium, barium and calcium), as well as the ammonium ion and substituted derivatives thereof (for example, dibenzylammonium, benzylammonium, 2-hydroxyethylammonium, and the like).
  • the term “pharmaceutically acceptable salt” of the compounds described herein is intended to encompass any and all acceptable salt forms.
  • each anionic species described herein may sometimes be depicted as an anionic species.
  • the compounds exist with an equimolar ratio of cation.
  • the compounds can exist in the fully protonated form, or in the form of a salt such as sodium, potassium, ammonium or in combination with any inorganic base as described above.
  • each anionic species may independently exist as either the protonated species or as the salt species.
  • the compounds described herein may have one or more chiral (or asymmetric) centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers (e.g., cis or trans).
  • E and Z geometric isomers e.g., cis or trans
  • all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.
  • stereoisomers and mixtures thereof include “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • the compounds may occur in any isomeric form, including racemates, racemic mixtures, and as individual enantiomers or diastereomers.
  • crystalline forms of the compounds may exist as polymorphs, which are contemplated herein.
  • some of the compounds may also form solvates with water or other organic solvents. Such solvates are similarly included within the scope of the compounds described herein.
  • any of the aforementioned compounds may incorporate radioactive isotopes. Accordingly, also contemplated is use of isotopically-labeled compounds identical to those described herein, wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into these compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl respectively.
  • Certain isotopically-labeled compounds for example those into which radioactive isotopes such as 3 H and 14C are incorporated, are also useful in drug or substrate tissue distribution assays.
  • Tritiated hydrogen ( 3 H) and carbon-14 ( 14 C) isotopes are particularly preferred for their ease of preparation and detectability.
  • Substitution with heavier isotopes such as deuterium ( 2 H) can provide certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dose requirements and, therefore, may be preferred in some circumstances.
  • reference to an element, such as hydrogen (H) or carbon (C) is intended to encompass all isotopes of the same.
  • reference to H encompasses 1 H (protium), 2 H (deuterium) and 3 H (tritium), and reference to C encompasses 12 C, 13 C and 14 C.
  • compounds of formula (I) wherein both R 1 groups are 2 H (deuterium) are encompassed within the scope of this invention by reference to R 1 being, in one embodiment, hydrogen (H).
  • R 1 being, in one embodiment, hydrogen (H).
  • Isotopically-labeled compounds can generally be prepared by performing procedures routinely practiced in the art.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein.
  • prodrug refers to a metabolic precursor of a compound described herein that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound as described herein.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound described herein, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.
  • prodrugs are provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • prodrug is also meant to include any covalently bonded carriers which release the active compound as described herein in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound described herein may be prepared by modifying functional groups present in the compound described herein in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound described herein.
  • Prodrugs include compounds described herein wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, ester and amide derivatives of hydroxy, carboxy, mercapto or amino functional groups in the compounds described herein and the like.
  • the compounds used in the reactions described herein may be made according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature.
  • “Commercially available chemicals” may be obtained from standard commercial sources including Acros Organics (Pittsburgh Pa.), Aldrich Chemical (Milwaukee Wis., including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester Pa.), Crescent Chemical Co.
  • Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C., may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
  • a reference for the preparation and selection of pharmaceutical salts of the present disclosure is P. H. Stahl & C. G. Wermuth “Handbook of Pharmaceutical Salts,” Verlag Helvetica Chimica Acta, Zurich, 2002.
  • Methods are provided herein for treating or preventing (i.e., reducing the likelihood of occurrence of neurological condition, disease or disorder including but not limited to Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia; Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, including chorea associate with Huntington's disease. While some of these diseases are considered cognitive disorders (e.g., Alzheimer's disease), and other diseases are considered neurological movement diseases/disorders, several have both cognitive and movement deficiencies or conditions associated with them (e.g., Parkinson's disease, Huntington's disease).
  • a muscarinic receptor antagonist such as a selective M4 antagonist
  • One or any combination of diagnostic methods appropriate for the particular disease or disorder including physical examination, patient self-assessment, assessment and monitoring of clinical symptoms, performance of analytical tests and methods, including clinical laboratory tests, physical tests, and exploratory surgery, for example, may be used for monitoring the health status of the subject and the effectiveness of the inhibitor.
  • the effects of the methods of treatment described herein can be analyzed using techniques known in the art, such as comparing symptoms of patients suffering from or at risk of a particular disease or disorder that have received the pharmaceutical composition comprising an antagonist with those of patients who were not treated with the inhibitor or who received a placebo treatment.
  • the terms, “treat” and “treatment,” refer to medical management of a disease, disorder, or condition of a subject (i.e., patient) (see, e.g., Stedman's Medical Dictionary).
  • an appropriate dose and treatment regimen provide the M4 antagonist in an amount sufficient to provide therapeutic and/or prophylactic benefit.
  • Therapeutic benefit for subjects to whom the M4 antagonist compound(s) described herein are administered includes, for example, an improved clinical outcome, wherein the object is to prevent or slow or retard (lessen) an undesired physiological change associated with the disease, or to prevent or slow or retard (lessen) the expansion or severity of such disease.
  • effectiveness of the one or more M4 antagonists may include beneficial or desired clinical results that comprise, but are not limited to, abatement, lessening, or alleviation of symptoms that result from or are associated with the disease to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity that a subject will present symptoms on the basis of which a diagnosis of a disease is made); diminishment of extent of disease; stabilized (i.e., not worsening) state of disease; delay or slowing of disease progression; amelioration or palliation of the disease state; and remission (whether partial or total), whether detectable or undetectable; and/or overall survival.
  • beneficial or desired clinical results comprise, but are not limited to, abatement, lessening, or alleviation of symptoms that result from or are associated with the disease to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity that a subject will present symptoms on the
  • Treatment can also mean prolonging survival when compared to expected survival if a subject were not receiving treatment.
  • Subjects in need of treatment include those who already have the disease or disorder as well as subjects prone to have or at risk of developing the disease or disorder, and those in which the disease, condition, or disorder is to be prevented (i.e., decreasing the likelihood of occurrence or recurrence of the disease or disorder).
  • a subject i.e., patient, individual in need of treatment with an M4 antagonist as described herein may be a human or may be a non-human primate or other animal (i.e., veterinary use) who has developed symptoms of a hyperkinetic disease or disorder or who is at risk for developing a hyperkinetic disease or disorder.
  • Non-human animals that may be treated include mammals, for example, non-human primates (e.g., monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, rabbits), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, bovine, elephants, bears and other domestic, farm, and zoo animals.
  • rodents e.g., rats, mice, gerbils, hamsters, ferrets, rabbits
  • lagomorphs e.g., swine (e.g., pig, miniature pig)
  • swine e.g., pig, miniature pig
  • equine canine
  • feline bovine
  • elephants bears and other domestic, farm, and zoo animals.
  • compositions comprising any one of the M4 antagonist compounds described herein (a compound of Formula I, including specific compounds described herein) and a pharmaceutically acceptable excipient for use in the methods for treating hyperkinetic disorders.
  • a pharmaceutically acceptable excipient is a physiologically and pharmaceutically suitable non-toxic and inactive material or ingredient that does not interfere with the activity of the active ingredient; an excipient also may be called a carrier.
  • the formulation methods and excipients described herein are exemplary and are in no way limiting.
  • compositions are well known in the pharmaceutical art and described, for example, in Rowe et al., Handbook of Pharmaceutical Excipients: A Comprehensive Guide to Uses, Properties, and Safety, 5 th Ed., 2006, and in Remington: The Science and Practice of Pharmacy (Gennaro, 21 st Ed. Mack Pub. Co., Easton, Pa. (2005)).
  • exemplary pharmaceutically acceptable excipients include sterile saline and phosphate buffered saline at physiological pH. Preservatives, stabilizers, dyes, buffers, and the like may be provided in the pharmaceutical composition. In addition, antioxidants and suspending agents may also be used.
  • acceptable carriers and/or diluents include saline and sterile water, and may optionally include antioxidants, buffers, bacteriostats and other common additives.
  • the compositions can also be formulated as pills, capsules, granules, or tablets which contain, in addition to an M4 antagonist, diluents, dispersing and surface active agents, binders, and lubricants.
  • M4 antagonist diluents
  • dispersing and surface active agents binders, and lubricants.
  • One skilled in this art may further formulate the M4 antagonist in an appropriate manner, and in accordance with accepted practices, such as those disclosed in Remington, supra.
  • Methods of administration include systemic administration of an M4 antagonist described herein, preferably in the form of a pharmaceutical composition as discussed above.
  • systemic administration includes oral and parenteral methods of administration.
  • suitable pharmaceutical compositions include powders, granules, pills, tablets, and capsules as well as liquids, syrups, suspensions, and emulsions. These compositions may also include flavorants, preservatives, suspending, thickening and emulsifying agents, and other pharmaceutically acceptable additives.
  • the compounds of the present invention can be prepared in aqueous injection solutions which may contain, in addition to the M4 antagonist, buffers, antioxidants, bacteriostats, and other additives commonly employed in such solutions.
  • optimal doses are generally determined using experimental models and/or clinical trials.
  • the optimal dose of the M4 antagonist may depend upon the body mass, weight, blood volume, or other individual characteristics of the subject. For example, a person skilled in the medical art can consider the subject's condition, that is, stage of the disease, severity of symptoms caused by the disease, general health status, as well as age, gender, and weight, and other factors apparent to a person skilled in the medical art.
  • the amount of a compound described herein, that is present in a dose ranges from about 0.1 mg to about 2 mg per kg weight of the subject. In certain embodiments, a daily dose is about 10-150 mg. The use of the minimum dose that is sufficient to provide effective therapy is usually preferred.
  • Subjects may generally be monitored for therapeutic effectiveness by clinical evaluation and using assays suitable for the condition being treated or prevented, which methods will be familiar to those having ordinary skill in the art and are described herein.
  • the level of a compound that is administered to a subject may be monitored by determining the level of the compound in a biological fluid, for example, in the blood, blood fraction (e.g., plasma, serum), and/or in the urine, and/or other biological sample from the subject. Any method practiced in the art to detect the compound may be used to measure the level of compound during the course of a therapeutic regimen.
  • compositions comprising an M4 antagonist may formulated for timed release (also called extended release, sustained release, controlled release, or slow release). Such compositions may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.
  • Sustained-release formulations may contain the compound dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Excipients for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release. The amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition to be treated or prevented.
  • compositions described herein that comprise at least one of the M4 antagonist compounds described herein may be administered to a subject in need by any one of several routes that effectively deliver an effective amount of the compound.
  • Such administrative routes include, for example, oral, parenteral (e.g., subcutaneous, intravenous, intramuscular, intrasternal, intracavernous), enteral, rectal, intranasal, buccal, sublingual, intramuscular, and transdermal.
  • Kits with unit doses of one or more of the compounds described herein, usually in oral or injectable doses are provided.
  • Such kits may include a container containing the unit dose, an informational package insert describing the use and attendant benefits of the drugs in treating pathological condition of interest, and optionally an appliance or device for delivery of the composition.
  • Step 1A 5-[(3R)-3-methylpiperazin-1-yl]-2-(trifluoromethoxy)benzonitrile
  • this reaction is completed with stirring overnight, however if necessary, additional acid equivalents and/or gentle heat (50° C.) can be used to push the reaction.
  • Step 2A 2-[(3R)-3-methylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidine
  • this reaction is completed with stirring overnight, however if necessary, additional acid equivalents and/or gentle heat (50° C.) can be used to push the reaction.
  • Step 3A (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-cyano-4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
  • Triphosgene (2.1 g, 7.2 mmol, 0.40 eq) was dissolved in methylene chloride (50 mL) and a solution of 5-[(3R)-3-methylpiperazin-1-yl]-2-(trifluoromethoxy)benzonitrile 1a (5.2 g, 18 mmol, 1.0 eq) and N,N-diisopropylethylamine (6.0 mL, 36 mmol, 2.0 eq) in methylene chloride (50 mL) was added dropwise at room temperature.
  • Step 4A (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
  • Triphosgene (1.2 g, 4.0 mmol, 0.40 eq) was dissolved in methylene chloride (20 mL) and a solution of tert-butyl (3R)-3-methylpiperazine-1-carboxylate (2.0 g, 10 mmol, 1.0 eq) and N,N-diisopropylethylamine (1.6 mL, 10 mmol, 1.0 eq) in methylene chloride (30 mL) was added dropwise at room temperature.
  • reaction mixture was stirred for 10 min before a solution of 2-(1-benzylpiperidin-4-yl)ethan-1-amine (2.6 g, 12 mmol, 1.2 eq) and N,N-diisopropylethylamine (1.6 mL, 10 mmol, 1.0 eq) in methylene chloride (30 mL) was added. Stirring at room temperature, the reaction was complete within 1 hr. The reaction mixture was diluted further with methylene chloride and washed with sat. NH 4 Cl followed by sat. NaHCO 3 . The combined organic layers were dried over Na 2 SO 4 and concentrated in vacuo.
  • Step 4B (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
  • Step 5A (2R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
  • Step 6A (2R)-2-methyl-N-[2-(piperidin-4-yl)ethyl]-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • Triphosgene (1.3 g, 4.4 mmol, 0.40 eq) was dissolved in methylene chloride (30 mL) and a solution of (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 1l (2.5 g, 11 mmol, 1.0 eq) and N,N-diisopropylethylamine (3.6 mL, 22 mmol, 2.0 eq) in methylene chloride (30 mL) was added dropwise at room temperature.
  • reaction mixture was stirred for 10 min before a solution of tert-butyl 4-(2-aminoethyl)piperidine-1-carboxylate (2.9 g, 13 mmol, 1.2 eq) and N,N-diisopropylethylamine (3.6 mL, 22 mmol, 2.0 eq) in methylene chloride (30 mL) was added. Stirring at room temperature, the reaction was complete within 1 hr. The reaction mixture was diluted further with methylene chloride and washed with sat. NH 4 Cl followed by sat. NaHCO 3 .
  • Step 6B (2R)—N- ⁇ 2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl ⁇ -2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • reaction mixture was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (2R)—N- ⁇ 2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl ⁇ -2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 6-1.
  • 2R 2-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl ⁇ -2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 6-1.
  • Table below provides the observed (Obs) ion m/z ratio for 6-1 (first compound listed in Table 4) and other compounds that were made according to the procedure as described in this example.
  • Step 7A (2R)—N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • Step 7B (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 4-(2-aminoethyl)-1-benzylpiperidin-4-ol
  • Step 8A 4-Nitrophenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate
  • Step 8B N-[2-(1-Benzylpiperidin-4-yl)ethyl]-4-[4-(trifluoromethyl)phenyl]piperidine-1-carboxamide
  • Step 9A 1-[4-(Trifluoromethoxy)phenyl]piperidine-4-carboxylic acid
  • Step 9B N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • Step 10A N-[2-(1-benzylpiperidin-4-yl)ethyl]piperidine-4-carboxamide
  • a silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-10%) in methylene chloride over 20 min yielding tert-butyl 4- ⁇ [2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl ⁇ piperidine-1-carboxylate 10a.
  • a portion of the chromatographed 10a (1.0 g, 2.3 mmol) was dissolved in 20% TFA in methylene chloride (5 mL) and heated to 50° C. overnight. The reaction mixture was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate.
  • Step 10B N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[5-(trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide
  • Step 11A N-[2-(piperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • Step 11B N-(2- ⁇ 1-[(4-hydroxyphenyl)methyl]piperidin-4-yl ⁇ ethyl)-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • Step 12A 2-(1-Benzylpiperidin-4-yl)ethyl chloroformate
  • Step 12B 2-(1-benzylpiperidin-4-yl)ethyl 4-[4-(trifluoromethoxy)phenyl]piperazine-1-carboxylate
  • a silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-15%) in methylene chloride over 20 min to provide 2-(1-benzylpiperidin-4-yl)ethyl 4-[4-(trifluoromethoxy)phenyl]piperazine-1-carboxylate 12-1 (0.15 g, 0.31 mmol, 75%) as a foam.
  • the table below provides the observed (Obs) ion m/z ratio for 12-1 (first compound listed in Table 10) and other compounds that were made according to the procedure as described in this example.
  • Step 13A Ethyl (4E)-1-benzyl-4-(cyanomethylidene)piperidine-3-carboxylate
  • Step 14A 2-(1-benzyl-4-hydroxypiperidin-4-yl)acetonitrile
  • n-BuLi (2 M in pentane, 7.3 mL, 1.1 eq) was added dropwise to solution of MeCN (0.76 mL) in THF (6 mL) at ⁇ 78° C. and stirred 20 minutes.
  • a solution of 1-benzylpiperidine-4-one (2.5 g, 13.2 mmol, 1.0 eq) in 4 mL THF was added dropwise at ⁇ 78° C.
  • the reaction mixture was warmed to room temperature slowly overnight.
  • the mixture was diluted with EtOAc and extracted from water.
  • Step 14B 4-(2-aminoethyl)-1-benzylpiperidin-4-ol
  • Step 15A Ethyl 1-benzyl-4-(cyanomethyl)piperidine-4-carboxylate
  • Ethyl 1-benzylpiperidine-4-carboxylate (2.0 g, 8.1 mmol, 1.0 eq) was dissolved in anhydrous THF (20 mL) and cooled to ⁇ 78° C. Next, LDA (2.0M in THF, 8.1 mL, 2.0 eq) was added dropwise and stirred for one hour at ⁇ 78° C. A solution of bromoacetonitrile (1.9 g, 16.2 mmol, 2.0 eq) in THF (10 mL) was added dropwise at 78° C. and stirred at this temperature for 3 hours then warmed to room temperature. The reaction mixture was diluted with EtOAc and washed with sat. NH 4 Cl and brine.
  • Step 16A Benzyl 4-(oxiran-2-yl)piperidine-1-carboxylate
  • Trimethylsulfoxonium iodide (13.5 g, 60.7 mmol, 1.5 eq) was added in two portions to a solution of NaH (1.5 g, 60.7 mmol, 1.5 eq) in anhydrous DMSO (15 mL). This mixture was stirred for one hour at room temp. Then a solution of benzyl 4-formylpiperidine-1-carboxylate (10.0 g, 40.4 mmol, 1.0 eq) in anhydrous DMSO (20 mL) was added and the mixture stirred two hours. The reaction was poured into water and extracted twice with Et 2 O. The organic layer was dried over MgSO 4 , filtered, and concentrated.
  • Step 16B Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate
  • Benzyl 4-(oxiran-2-yl)piperidine-1-carboxylate 16a (6.8 g, 26.1 mmol, 1.0 eq) was dissolved in anhydrous THF (15 mL) and the catalyst dissolved in minimal anhydrous THF was added and cooled to 0° C. and water (0.26 mL, 14.6 mmol, 0.6 eq) was added dropwise. The reaction slowly warmed to room temperature and stirred overnight. LCMS showed slow reaction so additional water (0.05 mL, 2.8 mmol, 0.1 eq) was added and the reaction was stirred overnight.
  • reaction mixture was concentrated and purified by ISCO column chromatography eluting with a gradient of 0% to 70% EtOAc in hexanes.
  • Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate 16b (3.4 g, 12.9 mmol) was determined to be 99% ee by chiral chromatography compared to the racemic mixture.
  • Step 16C Benzyl 4-[(1S)-2-azido-1-hydroxyethyl]piperidine-1-carboxylate
  • Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate 16b (3.4 g, 12.9 mmol, 1.0 eq) was dissolved in EtOH (9 mL) and water (1 mL) and sodium azide (1.7 g, 25.8 mmol, 2.0 eq) and ammonium chloride (1.4 g, 25.8 mmol, 2.0 eq) were added. The reaction mixture was heated to 55° C. and stirred overnight. EtOH was removed in vacuo and the reaction mixture was diluted with DCM and extracted from sat. NaHCO 3 .
  • Step 16D Benzyl 4-[(1S)-2- ⁇ [(tert-butoxy)carbonyl]amino ⁇ -1-hydroxyethyl]-piperidine-1-carboxylate
  • Step 16E (1S)-2-amino-1-(1-benzylpiperidin-4-yl)ethan-1-ol
  • the crude amine (2.4 g, 9.8 mmol, 1.0 eq) was dissolved in EtOH (75 mL) and acetic acid (0.55 mL, 10.5 mmol, 1.1 eq) and benzaldehyde (1.6 mL, 15.8 mmol, 1.6 eq) were added followed by sodium cyanoborohydride (1.0 g, 15.8 mmol, 1.6 eq) and the reaction was stirred for 2 hours Additional benzaldehyde (1.0 mL, 8.0 mmol) was added and reaction complete after one hour of stirring. The reaction mixture was quenched with water and MeOH was removed in vacuo. The reaction mixture was redissolved in DCM and extracted with sat.
  • Step 17A Ethyl 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylate
  • Step 17B 7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxylic acid
  • Step 17C N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Step 18A N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Step 18B N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,4-difluorophenyl)-7-methyl-pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Step 19A 7-methyl-N-[2-(piperidin-4-yl)ethyl]-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Step 19B 7-methyl-N- ⁇ 2-[1-(pyridin-3-ylmethyl)piperidin-4-yl]ethyl ⁇ -2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Step 20A (3R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyrimidin-2-yl)-3-methylpiperidine-4-carboxamide
  • Step 21A (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N- ⁇ 2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl ⁇ -2,6-dimethylpiperazine-1-carboxamide
  • reaction mixture was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N- ⁇ 2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl ⁇ -2,6-dimethylpiperazine-1-carboxamide 21-1.
  • the table below provides the observed (Obs) ion m/z ratio for 21-1 (first compound listed in Table 15) and other compounds that were made according to the procedure as described in this example.
  • Step 22A (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide
  • methanesulfanato(2-di-t-butylphosphino-2′,4′,6′-tri-1-propyl-1,1′-biphenyl)(2′ amino-1,1′-biphenyl-2-yl) palladium (II) was used in place of bis(tri-tert-butylphosphine)palladium(0).
  • Step 23A (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin-5-yl)-2,6-dimethylpiperazine-1-carboxamide
  • Step 24A 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]pyrimidine-5-carbonitrile
  • Step 25A 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrazine
  • Step 26A (2R,6R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
  • Step 27A 2-[(3R,5S)-4- ⁇ [2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl ⁇ -3,5-dimethylpiperazin-1-yl]-N-cyclopropylpyrimidine-5-carboxamide
  • reaction mixture was passed through an HPLC filter diluting to 1 mL with MeOH and purified by preparative chromatography yielding 2-[(3R,5 S)-4- ⁇ [2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl ⁇ -3,5-dimethylpiperazin-1-yl]-N-cyclopropylpyrimidine-5-carboxamide 27-1.
  • the table below provides the observed (Obs) ion m/z ratio for 27-1 (first compound listed in Table 19) and other compounds that were made according to the procedure as described in this example.
  • Binding affinity (Ki) of compounds was measured by inhibition of radioligand binding to membranes from CHO cells expressing human M1, M2, M3, M4 and M5 receptors. Membranes were prepared by nitrogen cavitation and differential centrifugation as previously described (Hoare et al., Mol. Pharmacol. 2003 March; 63(3): 751-65). The radioligand employed was tritiated N-methylscopolamine, used at a concentration of 1.5 nM. A dose-response of twelve concentrations of compound was used, ranging from 10 ⁇ M to 32 pM.
  • the assay buffer was 50 mM HEPES, 100 mM NaCl, 5 mM MgCl 2 , 1 mM ethylenediaminetetraacetic acid, pH-adjusted to pH 7.4.
  • Membranes, radioligand and compound were incubated together for 90 minutes at 37° C., in a total volume of 150 ⁇ l in a 96-well plate.
  • Receptor-bound radioligand was then collected by harvesting the assay over glass fiber filters pretreated with polyethylenimine to trap the cell membranes, using rapid vacuum filtration. Harvesting and radioactivity counting was conducted as previously described (see, e.g., Hoare et al., Mot. Pharmacol. 2003 63(3):751-65); Erratum at Mol. Pharmacol. 2005 July; 68(1): 260).
  • Binding affinities of all the exemplified compounds, which are described in the examples and listed in the tables above, are less than 1 ⁇ M against the M4 receptor. More specifically, specificity for the M4 receptor for each of the compounds listed in Table 20 is as follows: (1) “+” means the compound had a Ki against the M4 receptor of less than 1 ⁇ M (1,000 nM) but greater or equal to 100 nM; (2) “++” means the compound had a Ki against the M4 receptor of less than 100 nM but greater or equal to 10 nM; and (3) “+++” means that the compound had a Ki against the M4 receptor of less than 10 nM.
  • acetylcholine binding to the muscarinic receptors activates G-proteins. Activation of G-proteins can be determined by their binding of the radiolabeled GTP analogue 35 S-GTP ⁇ S.
  • acetylcholine stimulates the binding of 35 S-GTP ⁇ S to G-proteins associated with cell membranes, and the incorporated 35 S-GTP ⁇ S can be collected by harvesting the membranes.
  • Antagonist activity of the compounds was determined as the IC50 for inhibition of the acetylcholine response.
  • the assay buffer used was 50 mM HEPES, 100 mM NaCl, 5 mM MgCl 2 , 1 mM ethylenediaminetetraacetic acid, pH-adjusted to pH 7.4.
  • Acetylcholine, compound (a dose-response of twelve concentrations ranging from 10 ⁇ M to 32 pM) and membranes from CHO cells expressing M4 or M2 receptors were incubated together in 150 ⁇ l buffer for 30 minutes at 30° C. in a 96-well plate.
  • 35 S-GTP ⁇ S was then added, to a final concentration of 0.2 nM and a final volume of 175 ⁇ l.
  • membranes were harvested by rapid vacuum filtration onto non-treated glass fiber filters, as previously described (see, e.g., Hoare et al., Mol. Pharmacol. 2003 63(3):751-65); Erratum at Mol. Pharmacol. 2005 July; 68(1): 260).
  • concentration of acetylcholine used was that which stimulated 80% of the maximal response (3 ⁇ M for the M4 receptor, 1 ⁇ M for M2). Many of the compounds described above have been evaluated in the functional assay.
  • Extracellular field potential recordings were made with an Axoprobe 1A amplifier (Axon Instruments Ltd., USA) via a Krebs'-filled glass micropipette (resistance 2-5 M12) positioned in the stratum radiatum of the CAI, digitized (10 kHz) via a CED1401 interface and stored on a computer with Spike2 software (Cambridge Electronic Design Ltd., Cambridge, UK).
  • Field excitatory postsynaptic potential (fEPSP) responses were evoked (pair of 0.02 ms pulses, separated by 40 ms; applied every 10 s; adjusted to approximately 60% of the maximal spike-free response) by a bipolar stimulating electrode positioned in the stratum radiatum near the CA3-CA1 border.
  • the cholinergic agonist carbachol (aza-acetylcholine, resistant to degradation by acetylcholinesterase) was used to stimulate muscarinic receptors.
  • the M1 muscarinic receptor was blocked using 5 ⁇ M VU0255035, a selective M1 antagonist.
  • the resulting inhibitory signal was primarily M4-mediated, based on its sensitivity to the M4 activator VU010010.
  • the effect of M4 antagonists on this M4-mediated inhibition of fEPSPs was measured by adding M4 compound 20 minutes prior to application of carbachol.
  • 6-OHDA Lesion protocol Male Sprague-Dawley rats were anesthetized with isoflurane and placed into the stereotaxic frame. Thirty minutes prior the injection of 6-OHDA, rats received desipramine (15 mg/kg, i.p.) to prevent the entry of the toxin into the noradrenergic cells. A unilateral lesion was induced by injections of 6-OHDA (8 ⁇ g/4 ⁇ l/site/rat; flow rate 1 ⁇ l/min; dissolved in 0.9% NaCl with 0.02% ascorbic acid) or vehicle into the left and right medial forebrain bundle at the following coordinates: AP ⁇ 4.4.
  • the rats were allowed to recover for 14 days and were then tested for locomotor activity induced by novelty (placing the rat in a new cage, 30 min) and for contraversive (contralateral) rotational behavior induced by apomorphine (0.2 mg/kg, s.c.).
  • mice Young adult male, Sprague-Dawley rats (240-250 g) were purchased from Charles River Laboratories and assessed in the Open Field (Kinder Scientific, CA) task during the light hours of a 12:12 L:D cycle and were tested under bright light conditions. Animals were allowed to acclimate to the facility for at least one week prior to use. On the day of testing, animals were acclimated to the test room for at least 1 hour and were then treated orally with the Neurocrine compound and placed into the test chamber 30 minutes later. Animals were allowed to freely ambulate for 60 minutes. Measurements taken included, but were not limited to, total horizontal and vertical beam breaks.

Abstract

Provided herein are small molecule compounds of the following formula (I): or a stereoisomer, tautomer, solvate, ester or pharmaceutically acceptable salt thereof, wherein A, B, C, Li, L2, Ri, R2, R3, R4, R5, w, x, y and z are as defined herein. Methods for treating diseases/disorders by antagonizing muscarinic receptors, including specifically antagonizing muscarinic receptor 4 (M4), are also disclosed. Such diseases/disorders are e.g. neurological diseases/disorders such as e.g. Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia, Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy, progressive supranuclear palsy, and Huntington's disease. Preferred compounds are e.g. N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1-carboxamide derivatives and related compounds wherein the pyrazine has been replaced by e.g. pyridazine, pyrimidine, pyridine or phenyl.

Description

    BACKGROUND Technical Field
  • Compounds are provided herein that selectively antagonize muscarinic receptors, in particular muscarinic receptor 4 (M4), as well as methods for treating diseases and/or disorders that would benefit from the same.
  • Description of the Related Art
  • Muscarinic acetylcholine receptors are autonomic receptors that form G protein-receptor complexes in the cell membranes of certain neurons and other cell types (e.g., endothelial cells of blood vessels). Muscarinic receptors are located postsynaptically at the parasympathetic neuroeffector junction, from where the receptors function to increase or decrease the activity of the effector cells. Extrapyramidal symptoms are observed in patients treated with antipsychotic therapeutics and in patients who have neuroleptic malignant syndrome, brain damage (e.g., athetotic cerebral palsy), encephalitis, and meningitis. Drugs other than antipsychotics also cause extrapyramidal symptoms, for example antidopaminergic drugs (e.g., the antiemetic metoclopramide and the antidepressant amoxapine) and selective serotonin reuptake inhibitors (SSR*), which indirectly decrease dopamine. Conditions associated with extrapyramidal symptoms include acute dystonic reactions, akathisia, pseudoparkinsonism, and tardive dyskinesia. Extrapyramidal symptoms caused by antipsychotic therapeutics are being treated with anticholinergic drugs that lack selectivity for any of the five muscarinic receptor subtypes (see, e.g., Erosa-Rivero et al., Neuropharmacology 81:176-87 (2014)). Because anticholinergic drugs that effect multiple muscarinic receptors may cause distinct and in certain instances opposing effects, therapeutics that exhibit selectivity for particular receptors are desired.
  • BRIEF SUMMARY
  • Provided herein are compounds that antagonize muscarinic receptors. In particular embodiments, compounds are provided that selectively antagonize muscarinic receptor 4 (M4). Such compounds are useful in the treatment of a number of diseases and/or disorders, particularly neurological conditions, diseases, and disorders including cognitive disorders such as Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia. In other embodiments, methods are provided for treating or preventing movement disorders which may include Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, particularly chorea associate with Huntington's disease.
  • In one embodiment, compounds are provided having the structure of formula (I):
  • Figure US20210338653A1-20211104-C00002
  • or a stereoisomer, tautomer, solvate, ester, prodrug, or pharmaceutically acceptable salt thereof, wherein A, B, C, L1, L2, R1, R2, R3, R4, R5, w, x, y and z are as defined below. More specific embodiments are also described within Tables 1-14, as well as the more specific formulas noted herein below.
  • In another embodiment, pharmaceutical compositions are provided comprising a compound of formula (I), including one or more of the specific compounds described herein, and at least one pharmaceutically acceptable excipient. The compounds, as well as the pharmaceutical compositions comprising the compounds, may be used for antagonizing a muscarinic receptor, such as muscarinic receptor 4 (M4). In certain embodiments, the compound is a selective M4 antagonist.
  • These and other embodiments will be apparent upon reference to the following detailed description. To this end, various references are set forth herein that describe in more detail certain background information, procedures, compounds and compositions, and are each hereby incorporated by reference in their entirety.
  • DETAILED DESCRIPTION
  • In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the invention may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments. Unless the context requires otherwise, throughout the specification and claims which follow, the word “comprise” and variations thereof, such as, “comprises” and “comprising,” are to be construed in an open, inclusive sense, that is, as “including, but not limited to.” In addition, the term “comprising” (and related terms such as “comprise” or “comprises” or “having” or “including”) is not intended to exclude that in other certain embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, described herein, may “consist of” or “consist essentially of” the described features. Headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed embodiments.
  • Reference throughout this specification to “one embodiment” or “an embodiment” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment. Thus, the appearances of the phrases “in one embodiment” or “in an embodiment” in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
  • Also, as used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to “a non-human animal” may refer to one or more non-human animals, or a plurality of such animals, and reference to “a cell” or “the cell” includes reference to one or more cells and equivalents thereof (e.g., plurality of cells) known to those skilled in the art, and so forth. When steps of a method are described or claimed, and the steps are described as occurring in a particular order, the description of a first step occurring (or being performed) “prior to” (i.e., before) a second step has the same meaning if rewritten to state that the second step occurs (or is performed) “subsequent” to the first step. The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 15% of the stated number or numerical range. It should also be noted that the term “or” is generally employed in its sense including “and/or” unless the content clearly dictates otherwise. The term, “at least one,” for example, when referring to at least one compound or to at least one composition, has the same meaning and understanding as the term, “one or more.”
  • Provided herein are compounds useful for treating diseases and/or disorders treatable by antagonizing one or more muscarinic receptors. In particular embodiments, compounds are provided that are selective for muscarinic receptor 4 (M4) (also referred herein as the M4 receptor).
  • Provided herein are compounds having a structure of the following formula (I):
  • Figure US20210338653A1-20211104-C00003
  • or a stereoisomer, tautomer, solvate, ester, prodrug, or pharmaceutically acceptable salt thereof, wherein:
  • A, B and C are each independently a carbocycle or heterocycle;
  • R1 is, at each occurrence, H, C1-4alkyl, C(═O)OC1-4alkyl or aryl;
  • R2, R3, R4 and R5 are each independently —OH, —NH2, —NH(C1-4alkyl), —N(C1-4alkyl)2, —C═N, —C(═O)NH2, halo, C1-4alkyl, C1-4alkylOH, C1-4haloalkyl, C1-4alkoxy or C1-4haloalkoxy;
  • w, x, y and z are each independently 0, 1, 2 or 3;
  • L1 is a heteroalkylene linker having at least one N, O or S heteroatom, and wherein the heteroalkylene may be a straight chain or cyclized and optionally substituted with oxo, —OH, C1-4alkyl or C1-4alkoxy; and
  • L2 is an optional linker that is not present or, when present, is —O(CH2)m— where m is 0 or 1.
  • In one embodiment of formula (I), R1 is H at both occurrences.
  • In one embodiment of formula (I), one R1 is H and the other R1 is methyl.
  • In one embodiment of formula (I), R1 is methyl at both occurrences.
  • In one embodiment of formula (I), A is a non-aromatic carbocyle, and more specifically cyclohexyl.
  • In one embodiment of formula (I), A is an aromatic carbocyle, and more specifically aryl.
  • In one embodiment of formula (I), A is phenyl or naphthyl.
  • In one embodiment of formula (I), A is phenyl.
  • In one embodiment of formula (I), A is an aromatic heterocycle, and more specifically one of the following:
  • Figure US20210338653A1-20211104-C00004
  • In one embodiment of formula (I), w is 0 and R2 is not present.
  • In one embodiment of formula (I), w is 1, 2 or 3, and R2 is at each occurrence —OH, —CN, halo, or C1-4alkyl.
  • In one embodiment, compounds are provided of formula (I) wherein -A(R2)w is:
  • Figure US20210338653A1-20211104-C00005
  • In one embodiment of formula (I), x is 0 and R3 is not present.
  • In one embodiment of formula (I), x is 1 or 2 and R3 is at each occurrence —OH or C1-4alkyl-OH.
  • In one embodiment of formula (I), L1 is a a heteroalkylene linker having at least one N or O heteroatom, and wherein the heteroalkylene may be a straight chain or cyclized and optionally substituted with oxo, —OH, C1-4alkyl or C1-4alkoxy, and more specifically one of the following:
  • Figure US20210338653A1-20211104-C00006
    Figure US20210338653A1-20211104-C00007
  • In one embodiment of formula (I), B is a non-aromatic carbocyle, and more specifically cyclohexyl.
  • In one embodiment of formula (I), B is an aromatic carbocyle, and more specifically aryl.
  • In one embodiment of formula (I), B is phenyl or naphthyl.
  • In one embodiment of formula (I), B is phenyl.
  • In one embodiment of formula (I), B is a non-aromatic heterocycle.
  • In one embodiment of formula (I), B is piperazinyl, piperadinyl or pyrrolidinyl, and more specifically one of the following:
  • Figure US20210338653A1-20211104-C00008
  • In one embodiment of formula (T), B an aromatic heterocycle, and more specifically
  • Figure US20210338653A1-20211104-C00009
  • In one embodiment of formula (I), y is 0 and R4 is not present.
  • In one embodiment of formula (I), y is 1 and R4 is methyl.
  • In one embodiment of formula (I), B is piperazinyl, y is 1 and R4 is methyl and, in a more specific embodiment, such moiety has the following structure:
  • Figure US20210338653A1-20211104-C00010
  • In one embodiment of formula (I), y is 2 and R4 at both occurrences is methyl.
  • In one embodiment of formula (I), B is piperazinyl, y is 2 and R4 at both occurrences is methyl and, in a more specific embodiment, such moiety has one of the following structures:
  • Figure US20210338653A1-20211104-C00011
  • In one embodiment of formula (I), L2 is not present.
  • In one embodiment of formula (I), L2 is —O—
  • In one embodiment of formula (I), L2 is —OCH2—.
  • In one embodiment of formula (I), C is an aromatic 5-12 membered carbocycle or heterocycle.
  • In one embodiment of formula (I), C is an aromatic carbocyle, and more specifically aryl.
  • In one embodiment of formula (I), C is phenyl.
  • In one embodiment of formula (I), C is an aromatic heterocycle, and more specifically one of the following:
  • Figure US20210338653A1-20211104-C00012
  • In one embodiment of formula (I), z is 0 and R5 is not present.
  • In one embodiment of formula (I), z is 1, 2 or 3, and each occurrence of R5 is independently-OH, —NH2, —NH(C1-4alkyl), —N(C1-4alkyl)2, —CN, halo, C1-4alkyl, C1-4alkyl-OH, C1-4haloalkyl, C1-4alkoxy or C1-4haloalkoxy.
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (TI) or (ITT):
  • Figure US20210338653A1-20211104-C00013
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (IV) or (V):
  • Figure US20210338653A1-20211104-C00014
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (VI):
  • Figure US20210338653A1-20211104-C00015
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (VII), (VIII) or (IX):
  • Figure US20210338653A1-20211104-C00016
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (X) or (XI):
  • Figure US20210338653A1-20211104-C00017
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (XII), (XIII) or (XIV):
  • Figure US20210338653A1-20211104-C00018
  • In one embodiment of formula (I), compounds are provided having a structure of the following formula (XV), (XVI) or (XVII):
  • Figure US20210338653A1-20211104-C00019
  • In one embodiment, compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 0.
  • In one embodiment, compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 1 or 2, and R4 is at each occurrence C1-4alkyl.
  • In one embodiment, compounds are provided of formula (XV), (XVI) or (XVII) wherein y is 1 or 2, and R4 is at each occurrence methyl.
  • In one embodiment, compounds are provided of formula (XV), (XVI) or (XVII) wherein —C(R5)z is one of the following:
  • Figure US20210338653A1-20211104-C00020
  • In one embodiment of the —C(R5)z groups noted immediately above, z is 0 and R5 is not present.
  • In one embodiment of the —C(R5)L groups noted immediately above, z is 1, 2 or 3, and each occurrence of R5 is independently-OH, —NH2, —NH(C1-4alkyl), —N(C1-4alkyl)2, —C═N, halo, C1-4alkyl, C1-4alkyl-OH, C1-4haloalkyl, C1-4alkoxy or C1-4haloalkoxy.
  • In one embodiment, a compound of formula (I) is one or more of the following compounds (with cal. ion m/z listed in parentheses):
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrazin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (462.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-cyanopyridazin-3-yl)-2,6-dimethylpiperazine-1-carboxamide (462.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin-5-yl)-2,6-dimethylpiperazine-1-carboxamide (462.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanophenyl)-2,6-dimethylpiperazine-1-carboxamide (460.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyridin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (461.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-4-methoxypyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (492.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-chloropyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (471.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-chloropyrazin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (471.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-chloropyridazin-3-yl)-2,6-dimethylpiperazine-1-carboxamide (471.3);
    • (2R,6R)-4-(4-amino-5-chloropyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide (486.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-chloropyridin-3-yl)-2,6-dimethylpiperazine-1-carboxamide (470.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[6-(trifluoromethyl)pyridazin-3-yl]piperazine-1-carboxamide 505.3
    • (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-methoxypyrimidin-2-yl)-2-methylpiperazine-1-carboxamide (453.3);
    • (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-methoxypyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (467.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-methoxypyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (467.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-methoxypyrazin-2-yl)-2,6-dimethylpiperazine-1-carboxamide (467.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-methoxypyridazin-3-yl)-2,6-dimethylpiperazine-1-carboxamide (467.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-methoxypyrimidin-5-yl)-2,6-dimethylpiperazine-1-carboxamide (467.3);
    • (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(difluoromethoxy)pyrimidin-2-yl]-2-methylpiperazine-1-carboxamide (489.3);
    • (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(difluoromethoxy)pyrimidin-2-yl]-2,6-dimethylpiperazine-1-carboxamide (503.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(difluoromethoxy)pyrimidin-2-yl]-2,6-dimethylpiperazine-1-carboxamide (503.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(difluoromethoxy)pyrazin-2-yl]-2,6-dimethylpiperazine-1-carboxamide (503.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[6-(difluoromethoxy)pyridazin-3-yl]-2,6-dimethylpiperazine-1-carboxamide (503.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2-(difluoromethoxy)pyrimidin-5-yl]-2,6-dimethylpiperazine-1-carboxamide (503.3);
    • (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5-(trifluoromethoxy)pyrimidin-2-yl]piperazine-1-carboxamide (507.3);
    • (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethoxy)pyrimidin-2-yl]piperazine-1-carboxamide (521.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethoxy)pyrimidin-2-yl]piperazine-1-carboxamide (521.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethoxy)pyrazin-2-yl]piperazine-1-carboxamide (521.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[6-(trifluoromethoxy)pyridazin-3-yl]piperazine-1-carboxamide (521.3);
    • (2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2-(trifluoromethoxy)pyrimidin-5-yl]piperazine-1-carboxamide (521.3);
    • N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-methoxypyrimidin-2-yl)piperazine-1-carboxamide (439.3);
    • N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(difluoromethoxy)pyrimidin-2-yl]piperazine-1-carboxamide (475.3); and
    • N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(trifluoromethoxy)pyrimidin-2-yl]piperazine-1-carboxamide (492.2).
  • In even more specific embodiments, specific compounds of formula I are as listed in Tables 1-19 herein.
  • In other embodiments, pharmaceutical compositions are provided that comprise a compound of Formula I, including one or more of the specific compounds described herein (see, e.g., Tables 1-19), and at least one pharmaceutically acceptable excipient.
  • In another embodiment, a method is provided for antagonizing a muscarinic receptor in a cell comprising contacting the cell and a compound of Formula I, including specific compounds described herein, for a time sufficient and under appropriate conditions to permit interaction between the cell and the compound. In certain embodiments, the cell is in a subject who is in need of treatment with a compound disclosed herein. For example, the subject may have or be at risk for developing a neurological disease, condition, or disorder including cognitive and movement neurological diseases, conditions, and disorders. In certain embodiments, methods are provided for preventing (i.e., reducing the likelihood of occurrence of) or treating Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia; Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, including chorea associate with Huntington's disease. A person skilled in the medical or neurological art will readily appreciate that many of the aforementioned neurological diseases have both cognitive deficits and movement deficiencies or difficulties associated with them.
  • As used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated.
  • Certain chemical groups named herein are preceded by a shorthand notation indicating the total number of carbon atoms that are to be found in the indicated chemical group. For example; C1-C4alkyl describes an alkyl group, as defined below, having a total of 1 to 4 carbon atoms, and C4-C12cycloalkylalkyl describes a cycloalkylalkyl group, as defined below, having a total of 4 to 12 carbon atoms. The total number of carbons in the shorthand notation does not include carbons that may exist in substituents of the group described. For example, the following terms have the meaning indicated.
  • “C1-C6alkyl” refers to an alkyl radical as defined below containing one to six carbon atoms. The C1-C6alkyl radical may be optionally substituted as defined below for an alkyl group. “C1-C4alkyl” refers to an alkyl radical as defined below containing one to four carbon atoms. The C1-C4alkyl radical may be optionally substituted as defined below for an alkyl group.
  • “C2-C4alkenyl” refers to an alkenyl radical as defined below containing two to six carbon atoms. The C2-C12alkenyl radical may be optionally substituted as defined below for an alkenyl group.
  • “C2-C6alkynyl” refers to an alknyl radical as defined below containing two to six carbon atoms. The C2-C12alknyl radical may be optionally substituted as defined below for an alkenyl group.
  • “C1-C4alkoxy” refers to an alkoxy radical as defined below containing one to twelve carbon atoms. The alkyl part of the C1-C4alkoxy radical may be optionally substituted as defined below for an alkyl group.
  • “C2-C6alkoxyalkyl” refers to an alkoxyalkyl radical as defined below containing two to six carbon atoms. Each alkyl part of the C2-C6alkoxyalkyl radical may be optionally substituted as defined below for an alkyl group.
  • “C7-C12aralkyl” refers to an aralkyl group as defined below containing seven to twelve carbon atoms. The aryl part of the C7-C12aralkyl radical may be optionally substituted as described below for an awl group. The alkyl part of the C7-C12aralkyl radical may be optionally substituted as defined below for an alkyl group.
  • “C7-C12aralkenyl” refers to an aralkenyl group as defined below containing seven to twelve carbon atoms. The aryl part of the C7-C12aralkenyl radical may be optionally substituted as described below for an aryl group. The alkenyl part of the C7-C12aralkenyl radical may be optionally substituted as defined below for an alkenyl group.
  • “C3-C12cycloalkyl” refers to a cycloalkyl radical as defined below having three to twelve carbon atoms. The C3-C12cycloalkyl radical may be optionally substituted as defined below for a cycloalkyl group.
  • “C4-C12cycloalkylalkyl” refers to a cycloalkylalkyl radical as defined below having four to twelve carbon atoms. The C4-C12cycloalkylalkyl radical may be optionally substituted as defined below for a cycloalkylalkyl group.
  • In addition to the foregoing, as used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated:
  • “Alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, one to eight carbon atoms, or one to six carbon atoms, or one to four carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, and the like.
  • “Alkenyl” refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably two to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like.
  • “Alkylene” or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms or from one to four carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • “Alkenylene” or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • “Carbocyclyl” refers to a stable 3- to 18-membered aromatic or non-aromatic ring radical which consists of 3 to 18 carbon atoms. Unless stated otherwise specifically in the specification, the carbocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems, and may be partially or fully saturated. Non-aromatic carbocyclyl radicals include cycloalkyl, while aromatice carbocyclyl redicals include aryl.
  • “Cycloalkyl” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptly, and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo-[2.2.1]heptanyl, and the like.
  • “Aryl” refers to a hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. The aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. In one embodiment, aryl is phenyl or naphthyl, and in another embodiment is phenyl.
  • “Heterocyclyl” refers to a stable 3- to 18-membered aromatic or non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated. Examples or aromatic hetercyclyl radicals are listed below in the definition of heteroaryls (i.e., heteroaryl being a subset of heterocyclyl). Examples of non-aromatic heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, pyrazolopyrimidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trioxanyl, trithianyl, triazinanyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl.
  • “Heteroaryl” refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, benzoxazolinonyl, benzimidazolthionyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, pteridinonyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyridinonyl, pyrazinyl, pyrimidinyl, pryrimidinonyl, pyridazinyl, pyrrolyl, pyrido[2,3-d]pyrimidinonyl, quinazolinyl, quinazolinonyl, quinoxalinyl, quinoxalinonyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, thieno[3,2-d]pyrimidin-4-onyl, thieno[2,3-d]pyrimidin-4-onyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl).
  • Unless stated otherwise specifically in the specification, each of alkyl, alkenyl, alkylene, alkenylene, carbocyclyl, cycloalkyl, aryl, heterocyclyl and heteroaryl as defined above may be optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, —R41—OR40, —R41—OC(O)—R40, —R41—N(R40)2, —R41—C(O)R40, —R41—C(O)OR40, —R41—C(O)N(R40)2, —R41—N(R40)C(O)OR42, —R41—N(R40)C(O)R42, —R41—N(R40)S(O)tR42 (where t is 1 to 2), —R41—N═C(OR40)R40, —R41—S(O)tOR42 (where t is 1 to 2), —R41—S(O)pR42 (where p is 0 to 2), and —R41—S(O)tN(R40)2 (where t is 1 to 2) where each R40 is independently hydrogen, alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; each R41 is independently a direct bond or a straight or branched alkylene or alkenylene chain; and each R42 is alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
  • “Amino” refers to the —NH2 radical.
  • “Cyano” refers to the —CN radical.
  • “Hydroxy” refers to the —OH radical.
  • “Nitro” refers to the —NO2 radical.
  • “Oxo” refers to the ═O substituent.
  • “Thioxo” refers to the ═S substituent.
  • “Trifluoromethyl” refers to the —CF3 radical.
  • “Trifluoromethoxy” refers to the —OCF3 radical.
  • “Acyl” refers to a radical —C(O)R, wherein R is alkyl, aralkyl, carbocyclyl, aryl, heteroaryl, or heterocyclyl, as defined herein. When R is methyl, the acyl group is also referred to as acetyl.
  • “Heteroalkylene” or “heteroalkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting of carbon and hydrogen and at least one heteroatom selected from N, O, and S.
  • “Alkoxy” refers to a radical of the formula —ORa where Ra is an alkyl, or haloalkyl radical as defined above containing one to six carbon atoms. Representative alkoxy groups include methoxy and ethoxy. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted. An alkoxy that is substituted with halo may be called herein a haloalkoxy, which includes for example trifluoromethoxy, trichloromethoxy and the like.
  • “Heteroalkenylene” or “heteroalkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting of carbon and hydrogen and at least one heteroatom selected from N, O, and S.
  • “Aralkyl” refers to a radical of the formula —Rb—Re where Rb is an alkylene chain as defined above and R, is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. The alkylene chain part of the aralkyl radical may be optionally substituted as described above for an alkylene chain. The aryl part of the aralkyl radical may be optionally substituted as described above for an aryl group.
  • “Cycloalkylalkyl” refers to a radical of the formula —RbRg where Rb is an alkylene chain as defined above and Rg is a cycloalkyl radical as defined above. The alkylene chain and the cycloalkyl radical may be optionally substituted as defined above.
  • “Fused” refers to any ring system described herein which is fused to an existing ring structure in the compounds of the invention. When the fused ring system is a heterocyclyl or a heteroaryl, any carbon in the existing ring structure which becomes part of the fused ring system may be replaced with a nitrogen.
  • “Halo” refers to bromo, chloro, fluoro or iodo.
  • “Haloalkyl” refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like. The alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
  • “Haloalkenyl” refers to an alkenyl radical, as defined above, that is substituted by one or more halo radicals, as defined above. The alkenyl part of the haloalkyl radical may be optionally substituted as defined above for an alkenyl group.
  • “Haloalkoxy” refers to an alkoxy radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethoxy, difluoromethoxy, trichloromethoxy, 2,2,2-trifluoroethoxy, 3-bromo-2-fluoropropyloxy, and the like. The alkoxy part of the haloalkoxy radical may be optionally substituted as defined above for an alkoxy group.
  • “N-heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen. An N-heterocyclyl radical may be optionally substituted as described above for heterocyclyl radicals.
  • “Heterocyclylalkyl” refers to a radical of the formula —RbRh where Rb is an alkylene chain as defined above and Rh is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkyl radical may be optionally substituted as defined above for an alkyene chain. The heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
  • “N-heteroaryl” refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. An N-heteroaryl radical may be optionally substituted as described above for heteroaryl radicals.
  • “Heteroarylalkyl” refers to a radical of the formula —RbRi where Rb is an alkylene chain as defined above and Rt is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group. The alkylene chain part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkylene chain.
  • “Hydroxyalkyl” refers to a radical of the formula —RbOH where Rb is an alkylene chain as defined above. The —OH group can be attached to any carbon in the alkylene chain. The alkylene chain part of the heteroarylalkyl radical may additionally be optionally substituted as defined above for an alkylene chain.
  • The compounds described herein may generally be used as the free acid or free base. Alternatively, the compounds may be used in the form of acid or base addition salts. Acid addition salts of the free amino compounds may be prepared by methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic, acetic, trifluoroacetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids. Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids. Base addition salts included those salts that form with the carboxylate anion and include salts formed with organic and inorganic cations such as those chosen from the alkali and alkaline earth metals (for example, lithium, sodium, potassium, magnesium, barium and calcium), as well as the ammonium ion and substituted derivatives thereof (for example, dibenzylammonium, benzylammonium, 2-hydroxyethylammonium, and the like). Thus, the term “pharmaceutically acceptable salt” of the compounds described herein is intended to encompass any and all acceptable salt forms.
  • Compounds described herein may sometimes be depicted as an anionic species. One of ordinary skill in the art will recognize that the compounds exist with an equimolar ratio of cation. For instance, the compounds can exist in the fully protonated form, or in the form of a salt such as sodium, potassium, ammonium or in combination with any inorganic base as described above. When more than one anionic species is depicted, each anionic species may independently exist as either the protonated species or as the salt species.
  • With regard to stereoisomers, the compounds described herein may have one or more chiral (or asymmetric) centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers (e.g., cis or trans). Likewise, unless otherwise indicated, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included. It is therefore contemplated that various stereoisomers and mixtures thereof include “enantiomers,” which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another. Thus, the compounds may occur in any isomeric form, including racemates, racemic mixtures, and as individual enantiomers or diastereomers.
  • Furthermore, some of the crystalline forms of the compounds may exist as polymorphs, which are contemplated herein. In addition, some of the compounds may also form solvates with water or other organic solvents. Such solvates are similarly included within the scope of the compounds described herein.
  • As one of skill in the art would appreciate, any of the aforementioned compounds may incorporate radioactive isotopes. Accordingly, also contemplated is use of isotopically-labeled compounds identical to those described herein, wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into these compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to, 2H, 3H, 13C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36Cl respectively. Certain isotopically-labeled compounds, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are also useful in drug or substrate tissue distribution assays. Tritiated hydrogen (3H) and carbon-14 (14C) isotopes are particularly preferred for their ease of preparation and detectability. Substitution with heavier isotopes such as deuterium (2H) can provide certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dose requirements and, therefore, may be preferred in some circumstances. To this end, reference to an element, such as hydrogen (H) or carbon (C), is intended to encompass all isotopes of the same. Thus, reference to H encompasses 1H (protium), 2H (deuterium) and 3H (tritium), and reference to C encompasses 12C, 13C and 14C. For example, compounds of formula (I) wherein both R1 groups are 2H (deuterium) are encompassed within the scope of this invention by reference to R1 being, in one embodiment, hydrogen (H). Isotopically-labeled compounds can generally be prepared by performing procedures routinely practiced in the art.
  • “Prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein. Thus, the term “prodrug” refers to a metabolic precursor of a compound described herein that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound as described herein. Prodrugs are typically rapidly transformed in vivo to yield the parent compound described herein, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • The term “prodrug” is also meant to include any covalently bonded carriers which release the active compound as described herein in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound described herein may be prepared by modifying functional groups present in the compound described herein in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound described herein. Prodrugs include compounds described herein wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, ester and amide derivatives of hydroxy, carboxy, mercapto or amino functional groups in the compounds described herein and the like.
  • In general, the compounds used in the reactions described herein may be made according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature. “Commercially available chemicals” may be obtained from standard commercial sources including Acros Organics (Pittsburgh Pa.), Aldrich Chemical (Milwaukee Wis., including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester Pa.), Crescent Chemical Co. (Hauppauge N.Y.), Eastman Organic Chemicals, Eastman Kodak Company (Rochester N.Y.), Fisher Scientific Co. (Pittsburgh Pa.), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan Utah), ICN Biomedicals, Inc. (Costa Mesa Calif.), Key Organics (Cornwall U.K.), Lancaster Synthesis (Windham N.H.), Maybridge Chemical Co. Ltd. (Cornwall U.K.), Parish Chemical Co. (Orem Utah), Pfaltz & Bauer, Inc. (Waterbury Conn.), Polyorganix (Houston Tex.), Pierce Chemical Co. (Rockford Ill.), Riedel de Haen AG (Hanover, Germany), Spectrum Quality Product, Inc. (New Brunswick, N.J.), TCI America (Portland Oreg.), Trans World Chemicals, Inc. (Rockville Md.), and Wako Chemicals USA, Inc. (Richmond Va.).
  • Methods known to one of ordinary skill in the art may be identified through various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present disclosure, or provide references to articles that describe the preparation, include for example, “Synthetic Organic Chemistry,” John Wiley & Sons, Inc., New York; S. R. Sandler et al., “Organic Functional Group Preparations,” 2nd Ed., Academic Press, New York, 1983; H. O. House, “Modem Synthetic Reactions”, 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, “Heterocyclic Chemistry”, 2nd Ed., John Wiley & Sons, New York, 1992; J. March, “Advanced Organic Chemistry: Reactions, Mechanisms and Structure,” 4th Ed., Wiley-Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present disclosure, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G. “Organic Synthesis: Concepts, Methods, Starting Materials”, Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3-527-29074-5; Hoffman, R.V. “Organic Chemistry, An Intermediate Text” (1996) Oxford University Press, ISBN 0-19-509618-5; Larock, R. C. “Comprehensive Organic Transformations: A Guide to Functional Group Preparations” 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-19031-4; March, J. “Advanced Organic Chemistry: Reactions, Mechanisms, and Structure” 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-60180-2; Otera, J. (editor) “Modern Carbonyl Chemistry” (2000) Wiley-VCH, ISBN: 3-527-29871-1; Patai, S. “Patai's 1992 Guide to the Chemistry of Functional Groups” (1992) Interscience ISBN: 0-471-93022-9; Quin, L. D. et al. “A Guide to Organophosphorus Chemistry” (2000) Wiley-Interscience, ISBN: 0-471-31824-8; Solomons, T. W. G. “Organic Chemistry” 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-19095-0; Stowell, J. C., “Intermediate Organic Chemistry” 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471-57456-2; “Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia” (1999) John Wiley & Sons, ISBN: 3-527-29645-X, in 8 volumes; “Organic Reactions” (1942-2000) John Wiley & Sons, in over 55 volumes; and “Chemistry of Functional Groups” John Wiley & Sons, in 73 volumes.
  • Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C., may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services. A reference for the preparation and selection of pharmaceutical salts of the present disclosure is P. H. Stahl & C. G. Wermuth “Handbook of Pharmaceutical Salts,” Verlag Helvetica Chimica Acta, Zurich, 2002.
  • Compound Synthesis
  • Detailed compound synthesis methods are described herein in the Examples. A person having ordinary skill in the chemical art would be able to make a compound of Formula I, including specific compounds described herein, by these methods or similar methods or other methods practiced by a person skilled in the art. In general, starting components may be obtained from commercial sources.
  • Methods of Treatment
  • Methods are provided herein for treating or preventing (i.e., reducing the likelihood of occurrence of neurological condition, disease or disorder including but not limited to Alzheimer's Disease, Lewy Body Dementia and the cognitive deficits associated with schizophrenia; Parkinson's Disease, drug induced Parkinsonism, dyskinesias, dystonia, chorea, levodopa induced dyskinesia, cerebral palsy and progressive supranuclear palsy, and Huntington's disease, including chorea associate with Huntington's disease. While some of these diseases are considered cognitive disorders (e.g., Alzheimer's disease), and other diseases are considered neurological movement diseases/disorders, several have both cognitive and movement deficiencies or conditions associated with them (e.g., Parkinson's disease, Huntington's disease).
  • The effectiveness of a muscarinic receptor antagonist, such as a selective M4 antagonist, with respect to treating a neurological condition, disease or disorder described herein can readily be determined by a person skilled in the medical and clinical arts. One or any combination of diagnostic methods appropriate for the particular disease or disorder, which methods are well known to a person skilled in the art, including physical examination, patient self-assessment, assessment and monitoring of clinical symptoms, performance of analytical tests and methods, including clinical laboratory tests, physical tests, and exploratory surgery, for example, may be used for monitoring the health status of the subject and the effectiveness of the inhibitor. The effects of the methods of treatment described herein can be analyzed using techniques known in the art, such as comparing symptoms of patients suffering from or at risk of a particular disease or disorder that have received the pharmaceutical composition comprising an antagonist with those of patients who were not treated with the inhibitor or who received a placebo treatment.
  • As understood by a person skilled in the medical art, the terms, “treat” and “treatment,” refer to medical management of a disease, disorder, or condition of a subject (i.e., patient) (see, e.g., Stedman's Medical Dictionary). In general, an appropriate dose and treatment regimen provide the M4 antagonist in an amount sufficient to provide therapeutic and/or prophylactic benefit. Therapeutic benefit for subjects to whom the M4 antagonist compound(s) described herein are administered, includes, for example, an improved clinical outcome, wherein the object is to prevent or slow or retard (lessen) an undesired physiological change associated with the disease, or to prevent or slow or retard (lessen) the expansion or severity of such disease. As discussed herein, effectiveness of the one or more M4 antagonists may include beneficial or desired clinical results that comprise, but are not limited to, abatement, lessening, or alleviation of symptoms that result from or are associated with the disease to be treated; decreased occurrence of symptoms; improved quality of life; longer disease-free status (i.e., decreasing the likelihood or the propensity that a subject will present symptoms on the basis of which a diagnosis of a disease is made); diminishment of extent of disease; stabilized (i.e., not worsening) state of disease; delay or slowing of disease progression; amelioration or palliation of the disease state; and remission (whether partial or total), whether detectable or undetectable; and/or overall survival.
  • “Treatment” can also mean prolonging survival when compared to expected survival if a subject were not receiving treatment. Subjects in need of treatment include those who already have the disease or disorder as well as subjects prone to have or at risk of developing the disease or disorder, and those in which the disease, condition, or disorder is to be prevented (i.e., decreasing the likelihood of occurrence or recurrence of the disease or disorder).
  • A subject (i.e., patient, individual) in need of treatment with an M4 antagonist as described herein may be a human or may be a non-human primate or other animal (i.e., veterinary use) who has developed symptoms of a hyperkinetic disease or disorder or who is at risk for developing a hyperkinetic disease or disorder. Non-human animals that may be treated include mammals, for example, non-human primates (e.g., monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, rabbits), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, bovine, elephants, bears and other domestic, farm, and zoo animals.
  • Pharmaceutical Compositions
  • The present disclosure further provides for pharmaceutical compositions comprising any one of the M4 antagonist compounds described herein (a compound of Formula I, including specific compounds described herein) and a pharmaceutically acceptable excipient for use in the methods for treating hyperkinetic disorders. A pharmaceutically acceptable excipient is a physiologically and pharmaceutically suitable non-toxic and inactive material or ingredient that does not interfere with the activity of the active ingredient; an excipient also may be called a carrier. The formulation methods and excipients described herein are exemplary and are in no way limiting. Pharmaceutically acceptable excipients are well known in the pharmaceutical art and described, for example, in Rowe et al., Handbook of Pharmaceutical Excipients: A Comprehensive Guide to Uses, Properties, and Safety, 5th Ed., 2006, and in Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, Pa. (2005)). Exemplary pharmaceutically acceptable excipients include sterile saline and phosphate buffered saline at physiological pH. Preservatives, stabilizers, dyes, buffers, and the like may be provided in the pharmaceutical composition. In addition, antioxidants and suspending agents may also be used.
  • For compositions formulated as liquid solutions, acceptable carriers and/or diluents include saline and sterile water, and may optionally include antioxidants, buffers, bacteriostats and other common additives. The compositions can also be formulated as pills, capsules, granules, or tablets which contain, in addition to an M4 antagonist, diluents, dispersing and surface active agents, binders, and lubricants. One skilled in this art may further formulate the M4 antagonist in an appropriate manner, and in accordance with accepted practices, such as those disclosed in Remington, supra.
  • Methods of administration include systemic administration of an M4 antagonist described herein, preferably in the form of a pharmaceutical composition as discussed above. As used herein, systemic administration includes oral and parenteral methods of administration. For oral administration, suitable pharmaceutical compositions include powders, granules, pills, tablets, and capsules as well as liquids, syrups, suspensions, and emulsions. These compositions may also include flavorants, preservatives, suspending, thickening and emulsifying agents, and other pharmaceutically acceptable additives. For parental administration, the compounds of the present invention can be prepared in aqueous injection solutions which may contain, in addition to the M4 antagonist, buffers, antioxidants, bacteriostats, and other additives commonly employed in such solutions.
  • As described herein optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose of the M4 antagonist may depend upon the body mass, weight, blood volume, or other individual characteristics of the subject. For example, a person skilled in the medical art can consider the subject's condition, that is, stage of the disease, severity of symptoms caused by the disease, general health status, as well as age, gender, and weight, and other factors apparent to a person skilled in the medical art. In general, the amount of a compound described herein, that is present in a dose ranges from about 0.1 mg to about 2 mg per kg weight of the subject. In certain embodiments, a daily dose is about 10-150 mg. The use of the minimum dose that is sufficient to provide effective therapy is usually preferred. Subjects may generally be monitored for therapeutic effectiveness by clinical evaluation and using assays suitable for the condition being treated or prevented, which methods will be familiar to those having ordinary skill in the art and are described herein. The level of a compound that is administered to a subject may be monitored by determining the level of the compound in a biological fluid, for example, in the blood, blood fraction (e.g., plasma, serum), and/or in the urine, and/or other biological sample from the subject. Any method practiced in the art to detect the compound may be used to measure the level of compound during the course of a therapeutic regimen.
  • Pharmaceutical composition comprising an M4 antagonist may formulated for timed release (also called extended release, sustained release, controlled release, or slow release). Such compositions may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site. Sustained-release formulations may contain the compound dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Excipients for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release. The amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition to be treated or prevented.
  • The pharmaceutical compositions described herein that comprise at least one of the M4 antagonist compounds described herein may be administered to a subject in need by any one of several routes that effectively deliver an effective amount of the compound. Such administrative routes include, for example, oral, parenteral (e.g., subcutaneous, intravenous, intramuscular, intrasternal, intracavernous), enteral, rectal, intranasal, buccal, sublingual, intramuscular, and transdermal.
  • Kits with unit doses of one or more of the compounds described herein, usually in oral or injectable doses, are provided. Such kits may include a container containing the unit dose, an informational package insert describing the use and attendant benefits of the drugs in treating pathological condition of interest, and optionally an appliance or device for delivery of the composition.
  • EXAMPLES Example 1
  • Figure US20210338653A1-20211104-C00021
  • Step 1A: 5-[(3R)-3-methylpiperazin-1-yl]-2-(trifluoromethoxy)benzonitrile
  • To a solution of tert-butyl (2R)-2-methylpiperazine-1-carboxylate (5.0 g, 25 mmol, 1.0 eq) and 5-bromo-2-(trifluoromethoxy)benzonitrile (3.8 mL, 25 mmol, 1 eq) in toluene (100 mL) was added sodium tert-butoxide (7.2 g, 75 mmol, 3.0 eq), racemic 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (1.6 g, 2.5 mmol, 0.10 eq), and lastly tris(dibenzylideneacetone)dipalladium(0) (2.3 g, 2.5 mmol, 0.10 eq), and the reaction mixture heated to 100° C. overnight. The resulting dark reaction mixture was cooled, passed thru a pad of celite, and concentrated in vacuo. Silica gel column (80 g) was dry loaded and run using an increasing gradient of EtOAc (0-50%) in hexanes over 25 min. The chromatographed material was dissolved in dioxane (40 mL) and treated with a solution of 4M HCl in dioxane (10 mL). The resulting thick suspension was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate. Following removal of the resin and concentration of the filtrate, the free base of 5-[(3R)-3-methylpiperazin-1-yl]-2-(trifluoromethoxy)benzonitrile 1a (5.2 g, 18 mmol, 72% over two steps) was isolated as an orange oil.
  • In general, this reaction is completed with stirring overnight, however if necessary, additional acid equivalents and/or gentle heat (50° C.) can be used to push the reaction.
  • Other compounds made using the above synthetic scheme include:
    • 1-(3,4-difluorophenyl)piperazine 1b;
    • 3-(piperazin-1-yl)benzonitrile 1c;
    • 2-fluoro-5-(piperazin-1-yl)benzonitrile 1d;
    • 3-fluoro-5-(piperazin-1-yl)benzonitrile 1e;
    • 1-[4-(trifluoromethoxy)phenyl]piperazine 1f;
    • 1-[3-(trifluoromethoxy)phenyl]piperazine 1g;
    • 1-(3,4,5-trifluorophenyl)piperazine 1h;
    • (3R)-3-methyl-1-[4-(trifluoromethyl)phenyl]piperazine 1i;
    • 2-fluoro-5-[(3R)-3-methylpiperazin-1-yl]benzonitrile 1j;
    • 3-fluoro-5-[(3R)-3-methylpiperazin-1-yl]benzonitrile 1k;
    • (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 1l;
    • (3R)-3-methyl-1-[5-(trifluoromethyl)pyridin-2-yl]piperazine 1m;
    • (3R)-1-[3-fluoro-5-(trifluoromethyl)pyridin-2-yl]-3-methylpiperazine 1n;
    • (3R)-3-methyl-1-[5-(trifluoromethoxy)pyridin-2-yl]piperazine 1o; and
    • (3R)-3-methyl-1-[6-(trifluoromethoxy)pyridin-3-yl]piperazine 1p.
    Example 2
  • Figure US20210338653A1-20211104-C00022
  • Step 2A: 2-[(3R)-3-methylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidine
  • To a solution of tert-butyl (2R)-2-methylpiperazine-1-carboxylate (2.2 g, 11.0 mmol, 1.0 eq) and 2-chloro-5-(trifluoromethyl)pyrimidine (2.0 g, 11.0 mmol, 1.0 eq) in 1-methyl-2-pyrrolidone (NMP, 10 mL) was added N,N-diisopropylethylamine (5.7 mL, 44.0 mmol, 4.0 eq) and the reaction mixture heated to 100° C. for 1 hr. The reaction mixture was cooled, diluted heavily with EtOAc, and washed repeatedly with brine (5×). The organic layer was dried over Na2SO4 and concentrated. Silica gel column (80 g) was loaded using methylene chloride and run using an increasing gradient of EtOAc (5-90%) in hexanes over 20 min. The chromatographed material was dissolved in dioxane (25 mL) and treated with a solution of 4M HCl in dioxane (6 mL). The resulting thick white suspension was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate. Following removal of the resin and concentration of the filtrate, the free base of 2-[(3R)-3-methylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidine 2a (1.9 g, 7.6 mmol, 69% over two steps) was isolated as a white solid.
  • In general, this reaction is completed with stirring overnight, however if necessary, additional acid equivalents and/or gentle heat (50° C.) can be used to push the reaction.
  • Other compounds made using the above synthetic scheme include:
    • 4-(dimethylamino)-2-[(3R)-3-methylpiperazin-1-yl]pyrimidine-5-carbonitrile 2b;
    • 2-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]pyrimidine-5-carbonitrile 2c;
    • 2-[(3R)-3-methylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidin-4-amine 2d;
    • 2-[(3R)-3-methylpiperazin-1-yl]pyrimidine-5-carbonitrile 2e;
    • 2-[(3R,5S)-3,5-dimethylpiperazin-1-yl]pyrimidine-5-carbonitrile 2f;
    • 2-{2,5-diazabicyclo[2.2.2]octan-2-yl}pyrimidine-5-carbonitrile 2g;
    • 2-{2,6-diazaspiro[3.3]heptan-2-yl}pyrimidine-5-carbonitrile 2h;
    • 2-{3,6-diazabicyclo[3.1.1]heptan-3-yl}pyrimidine-5-carbonitrile 2i;
    • 2-{3,8-diazabicyclo[3.2.1]octan-3-yl}pyrimidine-5-carbonitrile 2j;
    • 5-chloro-2-[(3R)-3-methylpiperazin-1-yl]pyrimidine 2k;
    • 5-chloro-2-[(3R,5S)-3,5-dimethylpiperazin-1-yl]pyrimidine 2l;
    • 6-[(3R,5 S)-3,5-dimethylpiperazin-1-yl]-5-fluoropyridine-3-carbonitrile 2m; and
    • 6-[(3R,5S)-3,5-dimethylpiperazin-1-yl]pyridine-3-carbonitrile 2n.
    Example 3
  • Figure US20210338653A1-20211104-C00023
  • Step 3A: (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-cyano-4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
  • Triphosgene (2.1 g, 7.2 mmol, 0.40 eq) was dissolved in methylene chloride (50 mL) and a solution of 5-[(3R)-3-methylpiperazin-1-yl]-2-(trifluoromethoxy)benzonitrile 1a (5.2 g, 18 mmol, 1.0 eq) and N,N-diisopropylethylamine (6.0 mL, 36 mmol, 2.0 eq) in methylene chloride (50 mL) was added dropwise at room temperature. Once the addition was complete, the reaction mixture was stirred for 10 min before a solution of 2-(1-benzylpiperidin-4-yl)ethan-1-amine (4.8 g, 22 mmol, 1.2 eq) and N,N-diisopropylethylamine (6.0 mL, 36 mmol, 2.0 eq) in methylene chloride (50 mL) was added and stirred at room temperature for an additional 1 hr. Then, the reaction was diluted further with methylene chloride and washed with sat. NH4Cl followed by sat. NaHCO3. The combined organic layers were dried over Na2SO4 and concentrated in vacuo. Silica gel column (80 g) was loaded using methylene chloride and run with an increasing gradient of MeOH (0-20%) in methylene chloride over 20 min to provide (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-cyano-4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide 3-1 (5.2 g, 9.8 mmol, 54%) as an orange oil. The table below provides the observed (Obs) ion m/z ratio for 3-1 (first compound listed in Table 1) and other compounds that were made according to the procedure as described in this example.
  • TABLE 1
    Cpd. No. Compound Name Obs Ion (m/z)
    3-1 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-cyano-4- 530.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    3-2 (1S)-1-(1-benzylpiperidin-4-yl)-2-{[(2R)-4-(3-cyano-4- 522.25
    fluorophenyl)-2-methylpiperazine-1-carbonyl]amino}ethyl
    acetate
    3-3 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-(3- 494.2
    cyano-4-fluorophenyl)-N,2-dimethylpiperazine-1-carboxamide
    3-4 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 478.2
    fluorophenyl)-N,2-dimethylpiperazine-1-carboxamide
    3-5 (2R)-N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-4-[3-cyano- 546.2
    4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    3-6 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-methoxyethyl]-4-(3- 494.2
    cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
    3-7 (2R)-4-[3-cyano-4-(trifluoromethoxy)phenyl]-N-(2-{1-[(4- 656.1
    iodophenyl)methyl]piperidin-4-yl}ethyl)-2-methylpiperazine-1-
    carboxamide
    3-8 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[6- 506.2
    (trifluoromethoxy)pyridin-3-yl]piperazine-1-carboxamide
    3-9 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5- 491.2
    (trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    3-10 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-fluoro-5- 508.2
    (trifluoromethyl)pyridin-2-yl]-2-methylpiperazine-1-
    carboxamide
    3-11 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-5- 464.2
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    3-12 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5- 475.2
    trifluorophenyl)piperazine-1-carboxamide
    3-13 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5- 506.2
    (trifluoromethoxy)pyridin-2-yl]piperazine-1-carboxamide
    3-14 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5- 490.2
    (trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide
    3-15 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanopyrimidin- 448.2
    2-yl)-2-methylpiperazine-1-carboxamide
    3-16 (2R)-N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-2- 491.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    3-17 (2R)-N-{2-[1-benzyl-3-(hydroxymethyl)piperidin-4-yl]ethyl}-2- 505.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    3-18 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-(3- 480.2
    cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
    3-19 N-{2-[1-benzyl-3-(hydroxymethyl)piperidin-4-yl]ethyl}-4- 491.2
    (3,4,5-trifluorophenyl)piperazine-1-carboxamide
    3-20 (2R)-N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-2- 505.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    3-21 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-[3- 546.2
    cyano-4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-
    carboxamide
    3-22 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-2- 491.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    3-23 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyridin- 461.1
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
  • Figure US20210338653A1-20211104-C00024
  • Step 4A: (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
  • Triphosgene (1.2 g, 4.0 mmol, 0.40 eq) was dissolved in methylene chloride (20 mL) and a solution of tert-butyl (3R)-3-methylpiperazine-1-carboxylate (2.0 g, 10 mmol, 1.0 eq) and N,N-diisopropylethylamine (1.6 mL, 10 mmol, 1.0 eq) in methylene chloride (30 mL) was added dropwise at room temperature. Once complete, the reaction mixture was stirred for 10 min before a solution of 2-(1-benzylpiperidin-4-yl)ethan-1-amine (2.6 g, 12 mmol, 1.2 eq) and N,N-diisopropylethylamine (1.6 mL, 10 mmol, 1.0 eq) in methylene chloride (30 mL) was added. Stirring at room temperature, the reaction was complete within 1 hr. The reaction mixture was diluted further with methylene chloride and washed with sat. NH4Cl followed by sat. NaHCO3. The combined organic layers were dried over Na2SO4 and concentrated in vacuo. Silica gel column (40 g) was loaded using methylene chloride and run with an increasing gradient of MeOH (0-25%) in methylene chloride over 20 min. The chromatographed material was dissolved in dioxane (40 mL) and treated with a solution of 4M HCl in dioxane (5 mL). In general, this reaction is completed with stirring overnight, however if necessary, additional acid equivalents and/or gentle heat (50° C.) can be used to push the reaction. The resulting light yellow suspension was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate.
  • Following removal of the resin and concentration of the filtrate, the free base of (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide 4a (2.0 g, 5.8 mmol, 58% over two steps) was isolated as a yellow oil. (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide 4b was made according to the same procedure, but with appropriately modified starting materials.
  • Step 4B: (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
  • To a solution of (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide 4a (20 mg, 0.06 mmol, 1.0 eq) and 5-bromo-2-fluoro-benzonitrile (12 mg, 0.06 mmol, 1.0 eq) in 1:1 dioxane:toluene (1 mL) was added sodium tert-butoxide (17 mg, 0.18 mmol, 3.0 eq), racemic 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (3.7 mg, 0.006 mmol, 0.10 eq), and lastly tris(dibenzylideneacetone)-dipalladium(0) (5.5 mg, 0.006 mmol, 0.10 eq), and the reaction mixture stirred vigorously at 100° C. overnight. The resulting dark suspension was cooled, passed through an HPLC filter and concentrated in vacuo. The crude material was treated with 1.5 mL of MeOH, passed through an additional HPLC filter (leaving any precipitate behind), and submitted for directly for preparative chromatography yielding (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide 4-1. The table below provides the observed (Obs) ion m/z ratio for 4-1 (first compound listed in Table 2) and other compounds that were made according to the procedure as described in this example.
  • TABLE 2
    Cpd. No. Compound Name Obs Ion (m/z)
    4-1 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 464.2
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    4-2 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-methoxypyrimidin-2- 453.3
    yl)-2-methylpiperazine-1-carboxamide
    4-4 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3- 460.3
    methylphenyl)-2-methylpiperazine-1-carboxamide
    4-5 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[4- 489.2
    (trifluoromethyl)phenyl]piperazine-1-carboxamide
    4-6 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,2-difluoro-2H-1,3- 501.2
    benzodioxol-5-yl)-2-methylpiperazine-1-carboxamide
    4-8 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-methoxypyridin-2- 452.3
    yl)-2-methylpiperazine-1-carboxamide
    4-9 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(6- 436.3
    methylpyridin-3-yl)piperazine-1-carboxamide
    4-10 (2S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[4- 505.2
    (trifluoromethoxy)phenyl]piperazine-1-carboxamide
    4-11 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-6- 496.2
    cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    4-12 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[4- 490.2
    (trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide
    4-13 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-fluoro-4- 453.3
    methylphenyl)-2-methylpiperazine-1-carboxamide
    4-14 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanopyrimidin- 462.2
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    4-15 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3,4-difluorophenyl)-2- 457.2
    methylpiperazine-1-carboxamide
    4-16 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[3- 505.2
    (trifluoromethoxy)phenyl]piperazine-1-carboxamide
    4-17 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-4-(3,4,5- 475.2
    trifluorophenyl)piperazine-1-carboxamide
    4-18 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-4-[4- 505.2
    (trifluoromethoxy)phenyl]piperazine-1-carboxamide
    4-19 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoro-6- 454.2
    methylpyridin-2-yl)-2-methylpiperazine-1-carboxamide
    4-20 (2S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 464.2
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    4-21 (2S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-cyano-4- 530.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    4-22 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-fluoro-4- 523.2
    (trifluoromethoxy)phenyl]-3-methylpiperazine-1-carboxamide
    4-23 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyridin-3-yl)- 447.2
    2-methylpiperazine-1-carboxamide
    4-24 (2S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5- 475.2
    trifluorophenyl)piperazine-1-carboxamide
    4-25 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanophenyl)-2- 446.2
    methylpiperazine-1-carboxamide
    4-26 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-4- 478.2
    methoxypyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    4-27 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoro-4- 471.2
    methoxypyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    4-28 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-cyano-4- 491.4
    (dimethylamino)pyrimidin-2-yl]-2-methylpiperazine-1-carboxamide
    4-28 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 476.3
    methoxyphenyl)-2-methylpiperazine-1-carboxamide
    4-30 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-fluorophenyl)-2- 439.2
    methylpiperazine-1-carboxamide
    4-31 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyridin-2-yl)- 447.2
    2-methylpiperazine-1-carboxamide
    4-33 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,4-difluorophenyl)-2- 457.2
    methylpiperazine-1-carboxamide
    4-34 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[6- 490.3
    (tnfluoromethyl)pyridin-3-yl]piperazine-1-carboxamide
    4-35 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-fluoropyridin-3-yl)- 440.2
    2-methylpiperazine-1-carboxamide
    4-36 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(2,4,5- 475.2
    trifluorophenyl)piperazine-1-carboxamide
    4-37 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,3-difluorophenyl)-2- 457.2
    methylpiperazine-1-carboxamide
    4-38 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4,6-dicyanopyrimidin- 473.2
    2-yl)-2-methylpiperazine-1-carboxamide
    4-39 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-5- 476.3
    methoxyphenyl)-2-methylpiperazine-1-carboxamide
    4-40 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(quinolin-3- 472.3
    yl)piperazine-1-carboxamide
    4-41 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoropyrimidin-2- 441.3
    yl)-2-methylpiperazine-1-carboxamide
    4-43 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-2- 464.3
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    4-44 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanothiophen-2-yl)- 452.2
    2-methylpiperazine-1-carboxamide
    4-45 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,5-difluorophenyl)-2- 457.2
    methylpiperazine-1-carboxamide
    4-46 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoropyridin-2-yl)- 440.2
    2-methylpiperazine-1-carboxamide
    4-47 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4- 421.3
    phenylpiperazine-1-carboxamide
    4-48 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3,5-difluoropyridin-2- 458.2
    yl)-2-methylpiperazine-1-carboxamide
    4-49 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-fluorophenyl)-2- 439.2
    methylpiperazine-1-carboxamide
    4-50 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 522.2
    methylpiperazin-1-yl]-6-methyl-N-(propan-2-yl)pyrimidine-4-
    carboxamide
    4-51 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-methoxypyridin-2- 452.3
    yl)-2-methylpiperazine-1-carboxamide
    4-52 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-chloro-4,5- 491.2
    difluorophenyl)-2-methylpiperazine-1-carboxamide
    4-53 (2S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-fluoro-4- 523.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    4-54 (2S,6R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1- 477.3
    benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
    4-55 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-5- 466.2
    fluoropyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    4-56 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-methoxyphenyl)-3- 451.3
    methylpiperazine-1-carboxamide
    4-60 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3,5-difluoro-4- 541.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    4-61 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoropyridin-3-yl)- 440.2
    2-methylpiperazine-1-carboxamide
    4-62 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(pyridin-2- 422.2
    yl)piperazine-1-carboxamide
    4-63 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(5- 436.3
    methylpyridin-3-yl)piperazine-1-carboxamide
    4-64 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-(pyrimidin-2- 423.2
    yl)piperazine-1-carboxamide
    4-68 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyanophenyl)-2- 446.2
    methylpiperazine-1-carboxamide
    4-69 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-4- 492.2
    methoxypyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    4-70 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-fluoro-4- 469.3
    methoxyphenyl)-2-methylpiperazine-1-carboxamide
    4-71 (3S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-4-(3,4,5- 475.2
    trifluorophenyl)piperazine-1-carboxamide
    4-72 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3,5-difluorophenyl)-2- 457.25
    methylpiperazine-1-carboxamide
    4-73 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2-fluoro-4- 507.2
    (trifluoromethyl)phenyl]-2-methylpiperazine-1-carboxamide
    4-74 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3,4,5- 461.3
    trifluorophenyl)piperazine-1-carboxamide
    4-75 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4,6-dicyanopyrimidin- 473.2
    2-yl)-2-methylpiperazine-1-carboxamide
    4-76 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 475.2
    (trifluoromethyl)phenyl]piperazine-1-carboxamide
    4-77 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-(trifluoromethyl)pyridin- 476.1
    2-yl]piperazine-1-carboxamide
    4-78 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-3- 479.2
    fluoropyridin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    4-79 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[2- 491.17
    (trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide
    4-80 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin-5- 448.15
    yl)-2-methylpiperazine-1-carboxamide
  • Example 5
  • Figure US20210338653A1-20211104-C00025
  • Step 5A: (2R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
  • To a solution of (2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide 4a (0.20 g, 0.58 mmol, 1.0 eq) and 4-amino-2-chloropyrimidine-5-carbonitrile (0.90 g, 0.58 mmol, 1.0 eq) in NMP (2 mL) was added N,N-diisopropylethylamine (0.38 mL, 2.3 mmol, 4.0 eq) and the reaction mixture heated to 100° C. for 1 hr. In some cases, lower temperatures or longer reaction times were used. The reaction mixture was cooled, diluted heavily with EtOAc, and washed repeatedly with brine (3×). The organic layer was dried over Na2SO4 and concentrated. Silica gel column (24 g) was loaded using methylene chloride and run using an increasing gradient of MeOH (0-20%) in methylene chloride over 20 min to provide (2R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide 5-1 (0.14 g, 0.31 mmol, 53%) as an off-white foam. The table below provides the observed (Obs) ion m/z ratio for 5-1 (first compound listed in Table 3) and other compounds that were made according to the procedure as described in this example.
  • TABLE 3
    Cpd. Obs Ion
    No. Compound Name (m/z)
    5-1 (2R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1-benzylpiperidin- 463.2
    4-yl)ethyl]-2-methylpiperazine-1-carboxamide
    5-2 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 467.2
    methylpiperazin-1-yl]pyrimidine-5-carboxylic acid
    5-3 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4,5- 491.1
    dichloropyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    5-4 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-{5-cyano-4-[(2,2,2- 559.2
    trifluoroethyl)amino]pyrimidin-2-yl}-2,6-dimethylpiperazine-1-
    carboxamide
    5-5 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-6- 482.2
    cyanopyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    5-6 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4- 477.2
    (trifluoropyrimidin-2-yl)piperazine-1-carboxamide
    5-7 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-{5-cyano-4-[(2,2,2- 545.2
    trifluoroethyl)amino]pyrimidin-2-yl}-2-methylpiperazine-1-
    carboxamide
    5-8 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[4- 491.2
    (trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    5-9 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-5- 475.2
    fluoropyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    5-10 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-bromo-4- 537.1
    chloropyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    5-11 methyl 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 515.1
    methylpiperazin-1-yl]-6-chloropyrimidine-4-carboxylate
    5-12 methyl 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 495.2
    methylpiperazin-1-yl]-6-methylpyrimidine-4-carboxylate
    5-13 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin-2- 448.3
    yl)-2-methylpiperazine-1-carboxamide
    5-14 (2S,6R)-4-(4-amino-5-cyanopyrimidin-2-yl)-N-[2-(1- 477.3
    benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
    5-15 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-cyano-4- 491.4
    (dimethylamino)pyrimidin-2-yl]-2-methylpiperazine-1-
    carboxamide
    5-16 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-4- 478.2
    methoxypyrimidin-2-yl)-2-methylpiperazine-1-carboxamide
    5-17 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyano-4- 492.2
    methoxypyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    5-18 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-chloropyrimidin- 471.2
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    5-19 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-fluoropyrimidin- 455.25
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    5-20 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin- 462.3
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    5-21 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-cyano-4- 491.3
    (methylamino)pyrimidin-2-yl]-2,6-dimethylpiperazine-1-carboxamide
    5-22 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-chloropyrimidin-2- 457.2
    yl)-2-methylpiperazine-1-carboxamide
    5-23 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5-cyano-4- 477.2
    (methylamino)pyrimidin-2-yl]-2-methylpiperazine-1-carboxamide
    5-24 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5- 477.15
    (trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    5-25 (3S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin-2- 464.2
    yl)-3-(hydroxymethyl)piperazine-1-carboxamide
    5-26 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin-2- 464.2
    yl)-2-(hydroxymethyl)piperazine-1-carboxamide
    5-27 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5- 505.1
    (trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    5-28 (2R,6S)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1- 520.1
    benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
    5-29 (2R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1- 506.1
    benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
    5-30 4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1- 492.1
    benzylpiperidin-4-yl)ethyl]piperazine-1-carboxamide
    5-31 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-bromopyrimidin-2- 501.2
    yl)-2-methylpiperazine-1-carboxamide
    5-32 (2R)-4-(4-amino-5-chloropyrimidin-2-yl)-N-[2-(1- 472.0
    benzylpiperidin-4-yl)ethyl]-2-methylpiperazine-1-carboxamide
    5-33 4-(4-amino-5-chloropyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4- 458.0
    yl)ethyl]piperazine-1-carboxamide
    5-34 4-(4-amino-5-fluoropyrimidin-2-yl)-N-[2-(1-benzylpiperidin-4- 442.0
    yl)ethyl]piperazine-1-carboxamide
  • Figure US20210338653A1-20211104-C00026
  • Example 6 Step 6A: (2R)-2-methyl-N-[2-(piperidin-4-yl)ethyl]-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • Triphosgene (1.3 g, 4.4 mmol, 0.40 eq) was dissolved in methylene chloride (30 mL) and a solution of (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 1l (2.5 g, 11 mmol, 1.0 eq) and N,N-diisopropylethylamine (3.6 mL, 22 mmol, 2.0 eq) in methylene chloride (30 mL) was added dropwise at room temperature. Once the addition was complete, the reaction mixture was stirred for 10 min before a solution of tert-butyl 4-(2-aminoethyl)piperidine-1-carboxylate (2.9 g, 13 mmol, 1.2 eq) and N,N-diisopropylethylamine (3.6 mL, 22 mmol, 2.0 eq) in methylene chloride (30 mL) was added. Stirring at room temperature, the reaction was complete within 1 hr. The reaction mixture was diluted further with methylene chloride and washed with sat. NH4Cl followed by sat. NaHCO3. The combined organic layers were dried over Na2SO4, concentrated in vacuo, and loaded onto a silica gel column (40 g). Elution with an increasing gradient of EtOAc (0-100%) in hexanes over 25 min yielded tert-butyl 4-(2-{[(2R)-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carbonyl]amino}ethyl)piperidine-1-carboxylate 6a. This chromatographed material was dissolved in dioxane (60 mL) and treated with a solution of 4M HCl in dioxane (10 mL). After stirring overnight, the resulting light yellow suspension was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate.
  • Following removal of the resin and concentration of the filtrate, the free base of (2R)-2-methyl-N-[2-(piperidin-4-yl)ethyl]-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 6b (2.2 g, 5.8 mmol, 53% over two steps) was isolated as a brown foam. (2S,6R)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-[2-(piperidin-4-yl)ethyl]piperazine-1-carboxamide 6c was made according to the same procedure, but with appropriately modified starting materials.
  • Step 6B: (2R)—N-{2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl}-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • To NMP solutions of (2R)-2-methyl-N-[2-(piperidin-4-yl)ethyl]-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 6b (0.05 mL, 0.50 M, 1 eq) and 1H-indole-5-carbaldehyde (0.05 mL, 0.50 M, 1 eq) was added an ethanolic solution of borane-pyridine complex (0.10 mL, 0.50 M, 2 eq) followed by acetic acid (5 μL) and the mixture stirred at RT overnight. The reaction mixture was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (2R)—N-{2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl}-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 6-1. The table below provides the observed (Obs) ion m/z ratio for 6-1 (first compound listed in Table 4) and other compounds that were made according to the procedure as described in this example.
  • TABLE 4
    Cpd. Obs Ion
    No. Compound Name (m/z)
    6-1 (2R)-N-{2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl}-2- 514.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-2 (2R)-N-(2-{1-[(4-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 500.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-3 (2R)-N-{2-[1-(2,3-dihydro-1-benzofuran-7-ylmethyl)piperidin-4- 517.2
    yl]ethyl}-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-
    carboxamide
    6-4 (2R)-N-(2-{1-[(4-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 493.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-5 (2R)-N-(2-{1-[(2-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 493.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-6 (2R)-N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl}-2-methyl- 481.3
    4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-7 (2R)-2-methyl-N-{2-[1-(pyridin-2-ylmethyl)piperidin-4-yl]ethyl}- 476.2
    4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-8 (2R)-2-methyl-N-{2-[1-(1,2,3-thiadiazol-4-ylmethyl)piperidin-4- 483.2
    yl]ethyl}-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-9 (2R)-N-{2-[1-(2,3-dihydro-1,4-benzodioxin-6-ylmethyl)piperidin- 533.2
    4-yl]ethyl}-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-
    carboxamide
    6-10 (2R)-N-(2-{1-[(3-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 493.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-11 1-(5-cyanopyridin-2-yl)-N-{2-[1-(1H-indol-5-ylmethyl)piperidin- 471.3
    4-yl]ethyl}piperidine-4-carboxamide
    6-12 (2R)-2-methyl-N-(2-{1-[(2-methyl-1,3-thiazol-4- 496.2
    yl)methyl]piperidin-4-yl}ethyl)-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
    6-13 (2R)-N-[2-(1-{[2-(difluoromethoxy)phenyl]methyl}piperidin-4- 541.2
    yl)ethyl]-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-
    carboxamide
    6-14 (2R)-2-methyl-N-{2-[1-(1-phenylethyl)piperidin-4-yl]ethyl}-4- 489.2
    (3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-15 4-(benzyloxy)-N-(2-{1-[(4-fluorophenyl)methyl]piperidin-4- 447.6
    yl}ethyl)benzamide
    6-16 (2R)-N-(2-{1-[(4-carbamoylphenyl)methyl]piperidin-4-yl}ethyl)- 518.17
    2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-17 (2R)-N-(2-{1-[(2,3-difluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 511.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-18 (2R)-N-(2-{1-[(3-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 500.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-19 (2R)-2-methyl-N-(2-{1-[(5-methyl-1,3,4-oxadiazol-2- 557.2
    yl)(phenyl)methyl]piperidin-4-yl}ethyl)-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
    6-20 (2R)-2-methyl-N-[2-(1-{[3- 559.2
    (trifluoromethoxy)phenyl]methyl}piperidin-4-yl)ethyl]-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
    6-21 (2R)-2-methyl-N-[2-(1-{[2- 559.2
    (trifluoromethoxy)phenyl]methyl}piperidin-4-yl)ethyl]-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
    6-22 (2R)-2-methyl-4-(3,4,5-trifluorophenyl)-N-(2-{1-[(2,3,4- 529.1
    trifluorophenyl)methyl]piperidin-4-yl}ethyl)piperazine-1-
    carboxamide
    6-23 (2R)-N-(2-{1-[(3,5-difluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 511.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-24 (2R)-2-methyl-N-[2-(1-{[4- 559.2
    (trifluoromethoxy)phenyl]methyl}piperidin-4-yl)ethyl]-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
    6-25 (2R)-N-(2-{1-[(2,5-difluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 511.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-26 (2R)-2-methyl-N-{2-[1-(thiophen-2-ylmethyl)piperidin-4- 481.2
    yl]ethyl}-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-27 (2R)-N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-2- 491.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-28 (2R)-N-(2-{1-[(2,6-difluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 511.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-29 (2R)-N-(2-{1-[(2-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 500.2
    methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
    6-30 (2R)-N-(2-{1-[(4-cyano-2-fluorophenyl)methyl]piperidin-4- 518.1
    yl}ethyl)-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-
    carboxamide
    6-31 (2R)-2-methyl-N-{2-[1-(pyridin-4-ylmethyl)piperidin-4-yl]ethyl}- 492.2
    4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    6-32 (2R)-2-methyl-N-{2-[1-(pyridin-3-ylmethyl)piperidin-4-yl]ethyl}- 492.2
    4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    6-33 (2R)-2-methyl-N-{2-[1-(pyridin-2-ylmethyl)piperidin-4-yl]ethyl}- 492.2
    4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    6-34 (2R)-N-(2-{1-[(2-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 516.1
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-35 (2R)-2-methyl-N-(2-{1-[(2-methylphenyl)methyl]piperidin-4- 505.2
    yl}ethyl)-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-36 (2R)-N-(2-{1-[(3-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 509.1
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-37 (2R)-N-(2-{1-[(4-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 509.1
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-38 (2R)-N-(2-{1-[(2-fluorophenyl)methyl]piperidin-4-yl}ethyl)-2- 509.2
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-39 (2R)-2-methyl-N-{2-[1-(pyrimidin-5-ylmethyl)piperidin-4- 492.3
    yl]ethyl}-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-40 (2R)-N-(2-{1-[(3-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 516.1
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-41 (2R)-N-(2-{1-[(4-cyanophenyl)methyl]piperidin-4-yl}ethyl)-2- 516.1
    methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-
    carboxamide
    6-42 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(4-hydroxyphenyl)- 478.1
    methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    6-43 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-[2-(1-{[4-(dimethylamino)- 505.1
    phenyl]methyl}piperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-
    carboxamide
    6-44 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1-(2- 476.0
    phenylethyl)piperidin-4-yl]ethyl}piperazine-1-carboxamide
    6-45 (2R,6S)-N-(2-{1-[(4-cyanophenyl)methyl]piperidin-4-yl}ethyl)-4- 487.0
    (5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    6-46 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(3-fluorophenyl)- 480.1
    methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    6-47 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[1-(1H-indol-5- 501.0
    ylmethyl)-piperidin-4-yl]ethyl}-2,6-dimethylpiperazine-1-
    carboxamide
    6-48 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(4- 480.0
    fluorophenyl)methyl]-piperidin-4-yl}ethyl)-2,6-
    dimethylpiperazine-1-carboxamide
    6-49 (2R,6S)-N-(2-{1-[(4-hydroxy-3-methylphenyl)methyl]piperidin-4- 535.1
    yl}ethyl)-2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-
    yl]piperazine-1-carboxamide
    6-50 (2R,6S)-N-{2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl}-2,6- 544.1
    dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-51 (2R,6S)-N-(2-{1-[(3-fluorophenyl)methyl]piperidin-4-yl}ethyl)- 523.1
    2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-52 (2R,6S)-N-(2-{1-[(2-fluorophenyl)methyl]piperidin-4-yl}ethyl)- 523.0
    2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-53 (2R,6S)-N-[2-(1-{[4-(dimethylamino)phenyl]methyl}piperidin-4- 548.1
    yl)ethyl]-2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-
    yl]piperazine-1-carboxamide
    6-54 (2R,6S)-N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)- 521.1
    2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-55 (2R,6S)-N-(2-{1-[(4-fluorophenyl)methyl]piperidin-4-yl}ethyl)- 523.1
    2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-56 (2R,6S)-2,6-dimethyl-N-{2-[1-(2-phenylethyl)piperidin-4- 519.1
    yl]ethyl}-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-
    carboxamide
    6-57 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(4-hydroxy-3- 492.0
    methylphenyl)methyl]piperidin-4-yl}ethyl)-2,6-
    dimethylpiperazine-1-carboxamide
    6-58 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1- 464.1
    (pyrimidin-2-ylmethyl)piperidin-4-yl]ethyl}piperazine-1-
    carboxamide
    6-59 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(3-methoxyphenyl)- 492.1
    methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    6-60 (2R,6S)-N-{2-[1-(1-benzofuran-5-ylmethyl)piperidin-4-yl]ethyl}- 502.0
    4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    6-61 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1- 463.1
    (pyridin-3-ylmethyl)piperidin-4-yl]ethyl}piperazine-1-
    carboxamide
    6-62 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1- 463.1
    (pyridin-4-ylmethyl)piperidin-4-yl]ethyl}piperazine-1-
    carboxamide
    6-63 (4-{[4-(2-{[(2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl- 506.1
    piperazine-1-carbonyl]amino}ethyl)piperidin-1-
    yl]methyl}phenyl)boronic acid
    6-64 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(2-fluoropyridin-3- 481.1
    yl)methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    6-65 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(4-methoxyphenyl)- 492.1
    methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    6-66 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-(2-{1-[(1- 517.0
    methyl-1H-1,2,3-benzotriazol-5-yl)methyl]piperidin-4-
    yl}ethyl)piperazine-1-carboxamide
    6-67 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(2-methoxyphenyl)- 492.0
    methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
  • Example 7
  • Figure US20210338653A1-20211104-C00027
  • Step 7A: (2R)—N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide
  • A solution of (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 1l (0.30 g, 1.3 mmol, 1.0 eq) and triethylamine (0.34 mL, 2.6 mmol, 2.0 eq) in methylene chloride (20 mL) was prepared and cooled to 0° C. Then, 4-nitrophenyl chloroformate (0.29 g, 1.4 mmol, 1.1 eq) was added dropwise and the mixture stirred for 10 min at 0° C. before removing the ice bath and stirring for an additional 15 min. The reaction mixture was diluted with methylene chloride, washed with sat. NH4Cl, dried over Na2SO4 and concentrated in vacuo. Purification on a silica gel column (12 g) using an increasing gradient of MeOH (0-10%) in methylene chloride provided (2R)—N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 7a (0.40 g, 1.0 mmol, 78%) as a yellow oil.
  • Step 7B: (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 4-(2-aminoethyl)-1-benzylpiperidin-4-ol
  • To NMP solutions of (2R)—N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-2-methyl-4-(3,4,5-trifluorophenyl)piperazine-1-carboxamide 7a (0.05 mL, 0.50 M, 1.0 eq) and 4-(2-aminoethyl)-1-benzylpiperidin-4-ol 14b (0.05 mL, 0.50 M, 1.0 eq) was added a NMP solution of triethylamine (0.05 mL, 2.0 M, 4.0 eq) and the mixture stirred at RT overnight. The reaction was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (3R)-3-methyl-1-(3,4,5-trifluorophenyl)piperazine 4-(2-aminoethyl)-1-benzylpiperidin-4-ol 7-1. The table below provides the observed (Obs) ion m/z ratio for 7-1 (first compound listed in Table 5) and other compounds that were made according to the procedure as described in this example.
  • TABLE 5
    Cpd. Obs Ion
    No. Compound Name (m/z)
    7-1 (2R)-N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-2-methyl-4- 491.2
    (3,4,5-trifluorophenyl)piperazine-1-carboxamide
    7-2 (2R)-N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-4- 494.2
    (3-cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
    7-3 (2R)-N-[2-(4-benzylpiperazin-1-yl)ethyl]-4-[3-cyano-4- 531.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    7-4 (2R)-N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-(3- 480.3
    cyano-5-fluorophenyl)-2-methylpiperazine-1-carboxamide
    7-5 (2R)-N-{2-[(3S)-1-benzylpyrrolidin-3-yl]ethyl}-2-methyl-4- 461.2
    (3,4,5-trifluorophenyl)piperazine-1-carboxamide
    7-6 (2R)-N-[2-(4-benzylpiperazin-1-yl)ethyl]-2-methyl-4-(3,4,5- 476.2
    trifluorophenyl)piperazine-1-carboxamide
    7-7 (2R)-N-[2-(4-benzyl-4-hydroxypiperidin-1-yl)ethyl]-2-methyl-4- 491.2
    (3,4,5-trifluorophenyl)piperazine-1-carboxamide
    7-8 (2R)-N-{2-[(3S)-1-benzylpyrrolidin-3-yl]ethyl}-4-(3-cyano-5- 450.2
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    7-9 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-(difluoromethyl)- 489.2
    4-fluorophenyl]-2-methylpiperazine-1-carboxamide
    7-10 (2R)-N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-[3- 546.2
    cyano-4-(trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    7-11 (2R)-N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-4-(3-cyano- 480.2
    4-fluorophenyl)-2-methylpiperazine-1-carboxamide
    7-12 (2R)-N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-(3- 480.2
    cyano-4-fluorophenyl)-2-methylpiperazine-1-carboxamide
    7-13 (2R)-N-(2-{[(3R,5R)-1-benzyl-5-(hydroxymethyl)pyrrolidin-3- 520.2
    yl](methyl)amino}ethyl)-2-methyl-4-(3,4,5-
    trifluorophenyl)piperazine-1-carboxamide
  • Example 8
  • Figure US20210338653A1-20211104-C00028
  • Step 8A: 4-Nitrophenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate
  • A solution of 2-(1-benzylpiperidin-4-yl)ethan-1-amine (0.50 g, 2.3 mmol, 1.0 eq) in methylene chloride (10 mL) was prepared and added dropwise to a cooled methylene chloride solution (10 mL, 0° C.) of 4-nitrophenyl chloroformate (0.51 g, 2.5 mmol, 1.1 eq) and triethylamine (0.64 mL, 4.6 mmol, 2.0 eq). The reaction mixture was diluted further with methylene chloride, washed with sat. NH4Cl, dried over MgSO4 and concentrated in vacuo. Silica gel column was loaded using methylene chloride and run with an increasing gradient of MeOH (0-10%) in methylene chloride to provide 4-nitrophenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate 8a (0.51 g, 1.0 mmol, 57%) as a yellow oil. Phenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate 8b was made according to the same procedure, but with appropriately modified starting materials.
  • Step 8B: N-[2-(1-Benzylpiperidin-4-yl)ethyl]-4-[4-(trifluoromethyl)phenyl]piperidine-1-carboxamide
  • To a NMP solution of 4-nitrophenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate 8a (0.05 mL, 0.50 M, 1 eq) and 4-(4-trifluoromethylphenyl)piperidine (0.05 mL, 0.50 M, 1 eq) was added a NMP solution of triethylamine (0.05 mL, 2.0 M, 4 eq) and the mixture stirred at RT overnight. The reaction was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography which yielded N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4-(trifluoromethyl)phenyl]piperidine-1-carboxamide 8-1. The table below provides the observed (Obs) ion m/z ratio for 8-1 (first compound listed in Table 6) and other compounds that were made according to the procedure as described in this example.
  • TABLE 6
    Cpd. Obs Ion
    No. Compound Name (m/z)
    8-1 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 474.3
    (trifluoromethyl)phenyl]piperidine-1-carboxamide
    8-2 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3- 491.3
    (trifluoromethoxy)phenyl]piperazine-1-carboxamide
    8-3 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3- 474.3
    (trifluoromethyl)phenyl]piperidine-1-carboxamide
    8-4 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 440.3
    chlorophenyl)piperidine-1-carboxamide
    8-5 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3- 436.1
    methoxyphenyl)piperidine-1-carboxamide
    8-6 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2- 436.1
    methoxyphenyl)piperidine-1-carboxamide
    8-7 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-2- 466.2
    cyanophenyl)piperazine-1-carboxamide
    8-8 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 491.3
    (trifluoromethoxy)phenyl]piperazine-1-carboxamide
    8-9 1-[2-(1-benzylpiperidin-4-yl)ethyl]-3-[1-(5-bromopyridin-2- 500.2
    yl)pyrrolidin-3-yl]-3-methylurea
    8-10 4-(1-benzothiophen-3-yl)-N-[2-(1-benzylpiperidin-4- 462.4
    yl)ethyl]piperidine-1-carboxamide
    8-11 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3- 441.4
    chlorophenyl)piperazine-1-carboxamide
    8-12 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2-cyano-4- 500.2
    (trifluoromethyl)phenyl]piperazine-1-carboxamide
    8-13 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 441.3
    chlorophenyl)piperazine-1-carboxamide
    8-14 4-(1,3-benzothiazol-2-yl)-N-[2-(1-benzylpiperidin-4- 463.4
    yl)ethyl]piperidine-1-carboxamide
    8-15 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-chloro-5- 510.1
    (trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide
    8-16 1-[2-(1-benzylpiperidin-4-yl)ethyl]-3-{1-[3-chloro-5- 524.2
    (trifluoromethyl)pyridin-2-yl]pyrrolidin-3-yl}-3-methylurea
    8-17 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3- 475.3
    (trifluoromethyl)phenyl]piperazine-1-carboxamide
    8-18 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-5- 450.23
    fluorophenyl)piperazine-1-carboxamide
    8-19 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-hydroxy-4-[3- 490.4
    (trifluoromethyl)phenyl]piperidine-1-carboxamide
    8-20 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-fluorophenyl)piperidine- 424.2
    1-carboxamide
    8-21 1-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-3-{1-[4- 489.3
    (trifluoromethyl)phenyl]pyrrolidin-3-yl}urea
    8-22 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 450.3
    fluorophenyl)piperazine-1-carboxamide
    8-23 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chlorophenyl)-4- 456.2
    hydroxypiperidine-1-carboxamide
    8-24 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 436.1
    methoxyphenyl)piperidine-1-carboxamide
    8-25 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyanophenyl)piperazine- 432.3
    1-carboxamide
    8-26 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-fluorophenyl)piperidine- 424.1
    1-carboxamide
    8-27 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanophenyl)piperazine- 432.7
    1-carboxamide
    8-28 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3- 423.4
    hydroxyphenyl)piperazine-1-carboxamide
    8-29 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(pyrimidin-2-yl)piperazine- 409.3
    1-carboxamide
    8-30 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(1H-indol-3-yl)piperidine- 445.1
    1-carboxamide
    8-31 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[3-(propan-2-yl)-1,2,4- 440.2
    oxadiazol-5-yl]piperidine-1-carboxamide
    8-32 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-fluorophenyl)piperidine- 424.1
    1-carboxamide
    8-33 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-cyano-4-phenylpiperidine- 431.1
    1-carboxamide
    8-34 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2- 425.3
    fluorophenyl)piperazine-1-carboxamide
    8-35 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 425.45
    fluorophenyl)piperazine-1-carboxamide
    8-36 4-(4-acetylphenyl)-N-[2-(1-benzylpiperidin-4-yl)ethyl]piperazine- 449.2
    1-carboxamide
    8-37 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-ethylpiperazin-1- 442.4
    yl)piperidine-1-carboxamide
    8-38 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-phenylpiperidine-1- 406.3
    carboxamide
    8-39 1-[2-(1-benzylpiperidin-4-yl)ethyl]-3-[1-(4- 439.4
    fluorophenyl)piperidin-4-yl]urea
    8-40 3-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[1-(3- 446.3
    cyanophenyl)piperidin-4-yl]urea
    8-41 3-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-phenoxyphenyl)urea 430.2
    8-42 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-hydroxy-4-(2- 452.2
    methoxyphenyl)piperidine-1-carboxamide
    8-43 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2-(hydroxymethyl)-4- 451.2
    methylphenyl]piperazine-1-carboxamide
    8-44 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-oxo-1,2,3,4- 476.2
    tetrahydroquinazolin-3-yl)piperidine-1-carboxamide
    8-45 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyridin-2- 433.5
    yl)piperazine-1-carboxamide
    8-46 4-(1H-1,2,3-benzotriazol-1-yl)-N-[2-(1-benzylpiperidin-4- 447.2
    yl)ethyl]piperidine-1-carboxamide
    8-47 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-chloropyridin-2- 442.3
    yl)piperazine-1-carboxamide
    8-48 N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-(5-cyanopyrimidin-2-yl)- 460.0
    3,8-diazabicyclo[3.2.1]octane-8-carboxamide
  • Example 9
  • Figure US20210338653A1-20211104-C00029
  • Step 9A: 1-[4-(Trifluoromethoxy)phenyl]piperidine-4-carboxylic acid
  • To a solution of tert-butyl piperidine-4-carboxylate (1.6 g, 8.7 mmol, 1.0 eq) and 1-iodo-4-(trifluoromethoxy)benzene (2.5 g, 8.7 mmol, 1.0 eq) in toluene (100 mL) was added sodium tert-butoxide (2.5 g, 25 mmol, 3.0 eq), racemic 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (0.54 g, 0.87 mmol, 0.10 eq), and lastly tris(dibenzylideneacetone)-dipalladium(0) (0.80 g, 0.87 mmol, 0.10 eq), and the reaction mixture heated to 100° C. overnight. The resulting dark reaction mixture was cooled, passed thru a pad of celite, and concentrated in vacuo. Silica gel column (40 g) was dry loaded and run using an increasing gradient of EtOAc (0-50%) in hexanes over 25 min yielding tert-butyl 1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxylate 9a. The chromatographed material 9a was dissolved in 20% TFA in methylene chloride (50 mL) and heated to 60° C. overnight. Following concentration, solid material was obtained by precipitation from hexanes and ether. The resulting white solid was collected by vacuum filtration to provide 1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxylic acid 9b (1.5 g, 5.2 mmol, 60% over two steps).
  • Step 9B: N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • To a solution of acid 9b (0.30 g, 1.0 mmol, 1.0 eq) and 2-(1-benzylpiperidin-4-yl)ethan-1-amine (0.26 g, 1.2 mmol, 1.2 eq) in methylene chloride (5 mL) was added triethylamine (0.41 mL, 3.0 mmol, 3.0 eq) followed by HATU (0.46 g, 1.2 mmol, 1.2 eq) and the mixture was stirred at room temperature overnight. The reaction mixture was diluted with sat. NH4Cl and extracted with methylene chloride. The combined organic layers were dried over MgSO4 and concentrated. A silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-20%) in methylene chloride over 20 min to provide N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide 9-1 (0.39 g, 0.80 mmol, 80%). The table below provides the observed (Obs) ion m/z ratio for 9-1 (first compound listed in Table 7) and other compounds that were made according to the procedure as described in this example.
  • TABLE 7
    Cpd. Obs Ion
    No. Compound Name (m/z)
    9-1 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 490.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-2 N-(1-benzylpiperidin-4-yl)-N-cyclopropyl-4-phenylbenzamide 411.3
    9-3 N-[2-(1-benzylpiperidin-4-yl)ethyl]-N-(2-hydroxyethyl)-1-[4- 534.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-4 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-chlorophenyl)-4- 454.1
    methyl-1,3-thiazole-5-carboxamide
    9-5 (2R)-N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-4-(3- 480.2
    cyano-5-fluorophenyl)-2-methylpiperazine-1-carboxamide
    9-6 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-fluoro-4-[4- 501.2
    (trifluoromethoxy)phenyl]benzamide
    9-7 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 483.15
    (trifluoromethoxy)phenyl]benzamide
    9-8 N-[2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-1-[4- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-9 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-1-[3-fluoro-4- 524.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-10 (2R)-N-{2-[(3S)-1-benzylpyrrolidin-3-yl]ethyl}-4-[3-cyano-4- 516.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    9-11 N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-1-[3- 520.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-12 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chlorophenyl)-2- 451.2
    fluorobenzamide
    9-13 N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-1-[4- 520.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-14 methyl 4-(4-{[2-(1-benzylpiperidin-4- 457.25
    yl)ethyl]carbamoyl}phenyl)benzoate
    9-15 N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-1-(3-cyano- 465.2
    4-fluorophenyl)piperidine-4-carboxamide
    9-16 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 467.3
    (trifluoromethyl)phenyl]benzamide
    9-17 N-(2-{[(3S)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-1-[4- 505.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-18 4-chloro-3-(3-cyanoquinolin-2-yl)-N-[(3R)-1- 473.2
    cyclohexylpyrrolidin-3-yl]-N-methylbenzamide
    9-19 N-[2-(4-benzyl-4-hydroxypiperidin-1-yl)ethyl]-2-[4- 539.2
    (trifluoromethoxy)phenyl]imidazo[1,2-a]pyridine-7-carboxamide
    9-20 N-{2-[(3R)-1-benzylpyrrolidin-3-yl]ethyl}-4-chloro-3-(4-chloro- 468.1
    6-methylpyridin-3-yl)benzamide
    9-21 1-(1-benzylpiperidin-4-yl)-4-{1-[3-chloro-5- 550.2
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carbonyl}piperazine
    9-22 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-1-[3- 505.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-23 N-[1-(1-benzylpiperidin-4-yl)pyrrolidin-3-yl]-N-methyl-1-[4- 545.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-24 N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-1-(3- 479.2
    cyano-4-fluorophenyl)piperidine-4-carboxamide
    9-25 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-[4- 523.2
    (trifluoromethoxy)phenyl]imidazo[1,2-a]pyridine-7-carboxamide
    9-26 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-1-[4- 505.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-27 N-[2-(1-benzyl-3-hydroxypiperidin-4-yl)ethyl]-1-[4- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-28 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl]amino}ethyl)-1-[3-fluoro-4- 509.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-29 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4- 431.5
    cyanophenyl)piperidine-4-carboxamide
    9-30 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 424.3
    cyanophenyl)benzamide
    9-31 N-(2-{[(3S)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-1-[3- 505.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-32 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methyl-2-[4- 488.2
    (trifluoromethyl)phenyl]-1,3-thiazole-5-carboxamide
    9-33 N-[(3R)-1-benzylpyrrolidin-3-yl]-N-methyl-1-{1-[4- 545.3
    (trifluoromethoxy)phenyl]piperidine-4-carbonyl}piperidin-4-
    amine
    9-34 N-[2-(1-benzylpiperidin-4-yl)ethyl]-N-methyl-1-[4- 504.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-35 N-(2-{[1-benzyl-4-(hydroxymethyl)pyrrolidin-3- 553.3
    yl](methyl)amino}ethyl)-1-[3-fluoro-4-
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-36 3-(1-benzylpiperidin-4-yl)-N-{1-[4- 490.2
    (trifluoromethoxy)phenyl]piperidin-4-yl}propanamide
    9-37 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-bromopyrimidin-2- 488.1
    yl)piperidine-4-carboxamide
    9-38 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-4- 414.2
    phenylbenzamide
    9-39 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl](methyl)amino}ethyl)-4-[4- 498.3
    (trifluoromethoxy)phenyl]benzamide
    9-40 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-fluoro-4-(4- 431.25
    methylphenyl)benzamide
    9-41 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 417.3
    fluorophenyl)benzamide
    9-42 1-benzyl-1-methyl-4-[2-(N-methyl-1-{1-[4- 518.3
    (trifluoromethoxy)phenyl]piperidin-4-
    yl}formamido)ethyl]piperidin-1-ium
    9-43 2-[4-(benzyloxy)phenyl]-N-[2-(1-benzylpiperidin-4- 443.3
    yl)ethyl]acetamide
    9-44 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-1-(3-cyano-4- 465.2
    fluorophenyl)piperidine-4-carboxamide
    9-45 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,6-dioxo-1,3-dipropyl- 557.3
    2,3,6,9-tetrahydro-1H-purin-8-yl)benzamide
    9-46 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-1-[3- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-47 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyridin-2- 432.3
    yl)piperidine-4-carboxamide
    9-48 N-(1-benzylpiperidin-4-yl)-N-cyclopropyl-2-[4- 535.2
    (trifluoromethoxy)phenyl]imidazo[1,2-a]pyridine-7-carboxamide
    9-49 N-{2-[(3S)-1-benzylpyrrolidin-3-yl]ethyl}-1-[4- 476.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-50 3-(1-benzylpiperidin-4-yl)-N-{1-[3- 490.2
    (trifluoromethoxy)phenyl]piperidin-4-yl}propanamide
    9-51 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-methoxyphenyl)-7- 484.25
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    9-52 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-methylpyridin-2- 421.3
    yl)piperidine-4-carboxamide
    9-53 N-(1-benzylpiperidin-4-yl)-N-cyclopropyl-4-(thiophen-2- 417.2
    yl)benzamide
    9-54 N-(2-{[(3R)-1-benzylpyrrolidin-3-yl]amino}ethyl)-1-[3- 491.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-55 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(thiophen-2-yl)benzamide 405.2
    9-56 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4- 423.1
    [(trifluoromethyl)sulfanyl]benzamide
    9-57 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(6-fluoropyridin-3-yl)-4- 438.1
    methyl-1,3-thiazole-5-carboxamide
    9-58 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-chloro-5 549.2
    (trifluoromethyl)pyridin-2-yl]-N-cyclopropylpiperidine-4-
    carboxamide
    9-59 N-[2-(1-benzylpiperidin-4-yl)ethyl]-5-(4-methoxyphenyl)-7- 538.2
    (trifluoromethyl)pyrazolo[1,5-a]pyrimidine-2-carboxamide
    9-60 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-chloro-3-(3- 509.2
    cyanoquinolin-2-yl)benzamide
    9-61 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,4-dichlorophenyl)-5- 471
    methyl-1H-imidazole-4-carboxamide
    9-62 N-{2-[1-benzyl-3-(hydroxymethyl)piperidin-4-yl]ethyl}-1-[4- 520.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-63 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-chloro-5 509.2
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carboxamide
    9-64 N-{2-[1-benzyl-4-(hydroxymethyl)piperidin-4-yl]ethyl}-1-[3- 538.2
    fluoro-4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-65 N-[(1-benzylpiperidin-4-yl)methyl]-1-[3-chloro-5- 495.1
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carboxamide
    9-66 N-{2-[(3R)-1-benzylpyrrolidin-3-yl]ethyl}-1-[4- 476.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-67 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-chloro-3-(4-chloro-6- 482.2
    methylpyridin-3-yl)benzamide
    9-68 N-[2-(4-benzylpiperidin-1-yl)ethyl]-2-[4- 523.1
    (trifluoromethoxy)phenyl]imidazo[1,2-a]pyridine-7-carboxamide
    9-69 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4- 415.05
    hydroxyphenyl)benzamide
    9-70 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4- 483.15
    (trifluoromethoxy)phenyl]benzamide
    9-71 N-[2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-1-[4- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-72 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-1-[3-fluoro-4- 524.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    9-73 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyano-4- 449.3
    fluorophenyl)piperidine-4-carboxamide
    9-74 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyridin-2-yl)-2- 429.1
    methyl-1H-imidazole-4-carboxamide
    9-75 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyridin-2-yl)-1H- 415.1
    imidazole-4-carboxamide
  • Example 10
  • Figure US20210338653A1-20211104-C00030
  • Step 10A: N-[2-(1-benzylpiperidin-4-yl)ethyl]piperidine-4-carboxamide
  • To a solution of 1-[(tert-butoxy)carbonyl]piperidine-4-carboxylic acid (2.0 g, 8.7 mmol, 1.0 eq) and 2-(1-benzylpiperidin-4-yl)ethan-1-amine (2.2 g, 9.6 mmol, 1.1 eq) in methylene chloride (20 mL) was added triethylamine (4.3 mL, 26.1 mmol, 3.0 eq) followed by HATU (4.0 g, 10.4 mmol, 1.2 eq) and the reaction stirred at room temperature overnight. The reaction mixture was diluted with saturated NH4Cl and extracted with methylene chloride. The combined organic layers were dried over MgSO4 and concentrated. A silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-10%) in methylene chloride over 20 min yielding tert-butyl 4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}piperidine-1-carboxylate 10a. A portion of the chromatographed 10a (1.0 g, 2.3 mmol) was dissolved in 20% TFA in methylene chloride (5 mL) and heated to 50° C. overnight. The reaction mixture was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate. Following removal of the resin and concentration of the filtrate, the free base of N-[2-(1-benzylpiperidin-4-yl)ethyl]piperidine-4-carboxamide 10b (0.62 g, 1.9 mmol, 83%) was isolated as a yellow oil.
  • (3R,4R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methylpiperidine-4-carboxamide 10c was synthesized following the same overall procedure.
  • Step 10B: N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[5-(trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide
  • To a solution of 10b (20 mg, 0.06 mmol, 1.0 eq) and 2-bromo-5-(trifluoromethoxy)pyridine (14 mg, 0.06 mmol, 1.0 eq) in toluene (1 mL) was added sodium tert-butoxide (17 mg, 0.18 mmol, 3.0 eq), racemic 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (3.7 mg, 0.006 mmol, 0.10 eq), and lastly tris(dibenzylideneacetone)-dipalladium(0) (5.5 mg, 0.006 mmol, 0.10 eq), and the reaction mixture stirred vigorously at 100° C. overnight. The resulting dark suspension was cooled, passed through an HPLC filter and concentrated. Then, the crude material was treated with 1.5 mL of MeOH, passed through an additional HPLC filter (leaving any precipitate behind), and submitted for directly for preparative chromatography yielding N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[5-(trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide 10-1. The table below provides the observed (Obs) ion m/z ratio for 10-1 (first compound listed in Table 8) and other compounds that were made according to the procedure as described in this example.
  • TABLE 8
    Cpd. Obs Ion
    No. Compound Name (m/z)
    10-1 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[5 491.2
    (trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide
    10-2 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyano-4- 461.3
    methoxyphenyl)piperidine-4-carboxamide
    10-3 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-phenylpiperidine-4- 406.3
    carboxamide
    10-4 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methyl-1-[2- 504.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-5 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2,2-difluoro-2H-1,3- 486.3
    benzodioxol-5-yl)piperidine-4-carboxamide
    10-6 N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[4- 504.25
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-7 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3,4,5- 460.3
    trifluorophenyl)piperidine-4-carboxamide
    10-8 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-fluoro-4- 508.5
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-9 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 474.3
    (trifluoromethyl)phenyl]piperidine-4-carboxamide
    10-10 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-methoxy-4- 520.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-11 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-fluoro-3- 508.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-12 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2-fluoro-4- 523.2
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    10-13 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-cyano-3- 515.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-14 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyano-5- 449.3
    fluorophenyl)piperidine-4-carboxamide
    10-15 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyridin-3- 432.3
    yl)piperidine-4-carboxamide
    10-16 N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[2- 504.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-17 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-fluoro-1-[2- 508.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-18 N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[3- 504.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-19 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3- 490.4
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-20 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-1-[3-cyano-4- 531.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-21 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3,4- 442.2
    difluorophenyl)piperidine-4-carboxamide
    10-22 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3,5- 442.3
    difluorophenyl)piperidine-4-carboxamide
    10-23 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-cyano-5- 515.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-24 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[5-(trifluoromethyl)pyridin- 475.3
    2-yl]piperidine-4-carboxamide
    10-25 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methyl-1-[3- 504.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-26 2-(1-benzylpiperidin-4-yl)-N-({1-[3- 490.2
    (trifluoromethoxy)phenyl]piperidin-4-yl}methyl)acetamide
    10-27 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(6-chloropyridin-3- 441.2
    yl)piperidine-4-carboxamide
    10-28 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-methoxypyridin-2- 437.2
    yl)piperidine-4-carboxamide
    10-29 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-methoxy-5- 520.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-30 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 476.3
    (trifluoromethyl)pyrimidin-2-yl]piperidine-4-carboxamide
    10-31 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methyl-1-[4- 504.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-32 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-(trifluoromethyl)pyridin- 475.2
    2-yl]piperidine-4-carboxamide
    10-33 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4-methoxy-3- 520.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-34 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[6- 491.2
    (trifluoromethoxy)pyridin-3-yl]piperidine-4-carboxamide
    10-35 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-fluorophenyl)piperidine- 424.3
    4-carboxamide
    10-36 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-cyano-4- 531.2
    (trifluoromethoxy)phenyl]-4-hydroxypiperidine-4-carboxamide
    10-37 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[2-fluoro-4- 508.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-38 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methoxy-1-[4- 520.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-39 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-fluoro-4- 454.4
    methoxyphenyl)piperidine-4-carboxamide
    10-40 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-hydroxy-1-(3,4,5- 476.3
    trifluorophenyl)piperidine-4-carboxamide
    10-41 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-fluoro-4- 524.2
    (trifluoromethoxy)phenyl]-4-hydroxypiperidine-4-carboxamide
    10-42 N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[4- 504.25
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-43 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3,4,5- 460.3
    trifluorophenyl)piperidine-4-carboxamide
    10-44 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-fluoro-4- 508.5
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-45 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2,2-difluoro-2H-1,3- 486.3
    benzodioxol-4-yl)piperidine-4-carboxamide
    10-46 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-cyano-5- 531.2
    (trifluoromethoxy)phenyl]-4-hydroxypiperidine-4-carboxamide
    10-47 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methoxy-1-[4- 505.2
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carboxamide
    10-48 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methoxy-1-[5- 505.2
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carboxamide
    10-49 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3- 476.15
    (trifluoromethoxy)phenyl]pyrrolidine-3-carboxamide
    10-50 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-cyano-2- 449.3
    fluorophenyl)piperidine-4-carboxamide
    10-51 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyano-4-fluorophenyl)-4- 465.25
    hydroxypiperidine-4-carboxamide
    10-52 (3S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3- 476.2
    (trifluoromethoxy)phenyl]pyrrolidine-3-carboxamide
    10-53 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3- 436.3
    methoxyphenyl)piperidine-4-carboxamide
    10-54 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyano-5- 461.6
    methoxyphenyl)piperidine-4-carboxamide
    10-55 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-fluoro-1-[4- 508.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-56 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-methyl-4- 504.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-57 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-cyano-4- 515.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-58 N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-1-(3-cyano-4- 465.2
    fluorophenyl)piperidine-4-carboxamide
    10-59 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2-fluorophenyl)piperidine- 424.3
    4-carboxamide
    10-60 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4- 436.35
    methoxyphenyl)piperidine-4-carboxamide
    10-61 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-hydroxy-1-[5- 507.2
    (trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide
    10-62 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-hydroxy-1-[3- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-63 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methoxy-1-[5- 521.2
    (trifluoromethoxy)pyridin-2-yl]piperidine-4-carboxamide
    10-64 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2,5-difluorophenyl)-4- 458.2
    hydroxypiperidine-4-carboxamide
    10-65 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3- 484.3
    methanesulfonylphenyl)piperidine-4-carboxamide
    10-66 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-cyanopyridin-2- 432.3
    yl)piperidine-4-carboxamide
    10-67 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2-cyanophenyl)piperidine- 431.25
    4-carboxamide
    10-68 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[6-(trifluoromethyl)pyridin- 475.3
    3-yl]piperidine-4-carboxamide
    10-69 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(2-cyano-4- 449.4
    fluorophenyl)piperidine-4-carboxamide
    10-70 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[2- 490.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-71 (3S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 476.2
    (trifluoromethoxy)phenyl]pyrrolidine-3-carboxamide
    10-72 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 476.25
    (trifluoromethoxy)phenyl]pyrrolidine-3-carboxamide
    10-73 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-fluoropyridin-2- 425.3
    yl)piperidine-4-carboxamide
    10-74 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4,6-dimethylpyrimidin-2- 436.4
    yl)piperidine-4-carboxamide
    10-75 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-methoxy-1-[2- 520.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    10-76 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[4- 472.3
    (difluoromethoxy)phenyl]piperidine-4-carboxamide
    10-77 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(3-cyanophenyl)piperidine- 431.3
    4-carboxamide
    10-78 N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(4-cyano-3- 449.4
    fluorophenyl)piperidine-4-carboxamide
    10-79 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyridin-2-yl)- 446.2
    3-methylpiperidine-4-carboxamide
    10-80 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[5- 489.2
    (trifluoromethyl)pyridin-2-yl]piperidine-4-carboxamide
    10-81 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-[3-fluoro-5- 507.15
    (trifluoromethyl)pyridin-2-yl]-3-methylpiperidine-4-carboxamide
  • Example 11
  • Figure US20210338653A1-20211104-C00031
  • Step 11A: N-[2-(piperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • To a solution of 1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxylic acid 9b (0.30 g, 1.0 mmol, 1.0 eq) and tert-butyl 4-(2-aminoethyl)piperidine-1-carboxylate (0.27 g, 1.2 mmol, 1.2 eq) in methylene chloride (5 mL) was added triethylamine (0.41 mL, 3.0 mmol, 3.0 eq) followed by HATU (0.46 g, 1.2 mmol, 1.2 eq) and the reaction was stirred at room temperature overnight. The reaction mixture was diluted with sat. NH4Cl and extracted with methylene chloride. The combined organic layers were dried over MgSO4 and concentrated. A silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-5%) in methylene chloride over 20 min yielding tert-butyl 4-[2-({1-[4-(trifluoromethoxy)phenyl]piperidin-4-yl}formamido)ethyl]-piperidine-1-carboxylate 11a. The chromatographed 11a was dissolved in 20% TFA in methylene chloride (5 mL) and heated to 50° C. overnight. The reaction mixture was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate. Following removal of the resin and concentration of the filtrate, the free base of N-[2-(piperidin-4-yl)ethyl]-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide 11b was isolated as a foam.
  • Step 11B: N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • To NMP solutions of 11b (0.05 mL, 0.50 M, 1 eq) and 4-hydroxybenzaldehyde (0.05 mL, 0.50 M, 1 eq) was added an ethanolic solution of borane-pyridine complex (0.10 mL, 0.50 M, 2 eq) followed by acetic acid (5 μL) and the mixture stirred at RT overnight. The reaction was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide 11-1. The table below provides the observed (Obs) ion m/z ratio for 11-1 (first compound listed in Table 9) and other compounds that were made according to the procedure as described in this example.
  • TABLE 9
    Obs Ion
    Cpd. No. Compound Name (m/z)
    11-1 N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-1-[4- 506.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-2 N-(2-{1-[(2-fluoropyridin-3-yl)methyl]piperidin-4-yl}ethyl)-4- 434.3
    phenoxybenzamide
    11-3 4-(benzyloxy)-N-(2-{1-[(2-hydroxyphenyl)methyl]piperidin-4- 445.2
    yl}ethyl)benzamide
    11-4 N-(2-{1-[(2-methyl-1,3-thiazol-4-yl)methyl]piperidin-4-yl}ethyl)- 511.2
    1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-5 N-(2-{1-[(2-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-4- 431.2
    phenoxybenzamide
    11-6 methyl 2-phenyl-2-{4-[2-({l-[4- 548.2
    (trifluoromethoxy)phenyl]piperidin-4-
    yl}formamido)ethyl]piperidin-1-yl}acetate
    11-7 4-(benzyloxy)-N-{2-[1-(2,3-dihydro-1,4-benzodioxin-5- 487.2
    ylmethyl)piperidin-4-yl]ethyl}benzamide
    11-8 1-(5-cyanopyridin-2-yl)-N-{2-[1-(cyclohexylmethyl)piperidin-4- 438.2
    yl]ethyl}piperidine-4-carboxamide
    11-9 4-(benzyloxy)-N-(2-{1-[(2-hydroxy-3- 475.2
    methoxyphenyl)methyl]piperidin-4-yl}ethyl)benzamide
    11-10 N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-4- 498.2
    phenoxybenzamide
    11-11 N-{2-[1-(1,2,3-thiadiazol-4-ylmethyl)piperidin-4-yl]ethyl}-1-[4- 520.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-12 N-{2-[1-(2-hydroxy-1-phenylethyl)piperidin-4-yl]ethyl}-1-[4- 448.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-13 N-(2-{1-[(3-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-4- 431
    phenoxybenzamide
    11-14 4-(benzyloxy)-N-{2-[1-(1-phenylethyl)piperidin-4- 443.3
    yl]ethyl}benzamide
    11-15 N-(2-{1-[(1-methyl-1H-pyrrol-2-yl)methyl]piperidin-4-yl}ethyl)- 493.3
    1-[4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-16 1-(5-cyanopyridin-2-yl)-N-{2-[1-(2,3-dihydro-1,4-benzodioxin-5- 490.2
    ylmethyl)piperidin-4-yl]ethyl}piperidine-4-carboxamide
    11-17 1-(5-cyanopyridin-2-yl)-N-(2-{1-[(4- 448.3
    hydroxyphenyl)methyl]piperidin-4-yl}ethyl)piperidine-4-
    carboxamide
    11-18 N-{2-[1-(2,3-dihydro-1,4-benzodioxin-5-ylmethyl)piperidin-4- 473.3
    yl]ethyl}-4-phenoxybenzamide
    11-19 N-{2-[1-(3-methylbutyl)piperidin-4-yl]ethyl}-1-[4- 430.1
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-20 4-(benzyloxy)-N-{2-[1-(pyridin-3-ylmethyl)piperidin-4- 433.4
    yl]ethyl}benzamide
    11-21 N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl}-1-[4- 496.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-22 N-{2-[1-(1H-indol-5-ylmethyl)piperidin-4-yl]ethyl}-1-[4- 529.2
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-23 N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]-2-hydroxyethyl}-1-[4- 512.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-24 4-(benzyloxy)-N-(2-{1-[(3-fluorophenyl)methyl]piperidin-4- 446.9
    yl}ethyl)benzamide
    11-25 1-(5-cyanopyridin-2-yl)-N-(2-{1-[(2- 448.3
    hydroxyphenyl)methyl]piperidin-4-yl}ethyl)piperidine-4-
    carboxamide
    11-26 1-(5-cyanopyridin-2-yl)-N-(2-{1-[(2- 447.5
    hydroxyphenyl)methyl]piperidin-4-yl}ethyl)piperidine-4-
    carboxamide
    11-27 N-{2-[1-(2,2-dimethylpropyl)piperidin-4-yl]ethyl}-1-[4- 435.3
    (trifluoromethoxy)phenyl]piperidine-4-carboxamide
    11-28 4-phenoxy-N-{2-[1-(3-phenylpropyl)piperidin-4- 431.2
    yl]ethyl}benzamide
    11-29 N-{2-[1-(2,3-dihydro-1-benzofuran-7-ylmethyl)piperidin-4- 532.2
    yl]ethyl}-1-[4-(trifluoromethoxy)phenyl]piperidine-4-
    carboxamide
    11-30 N-(2-{l-[(6-methoxypyridin-2-yl)methyl]piperidin-4-yl}ethyl)-1- 521.1
    [4-(trifluoromethoxy)phenyl]piperidine-4-carboxamide
  • Example 12
  • Figure US20210338653A1-20211104-C00032
  • Step 12A: 2-(1-Benzylpiperidin-4-yl)ethyl chloroformate
  • A solution of 2-(1-benzylpiperidin-4-yl)ethan-1-ol (0.50 g, 2.3 mmol, 1.0 eq) and triphosgene (0.81 g, 2.7 mmol, 1.2 eq) in anhydrous THE (7 mL) was prepared and cooled to 0° C. Then, pyridine (0.27 g, 3.4 mmol, 1.5 eq) was added dropwise upon which a precipitate formed. After stirring for 20 min, the resulting suspension was filtered and concentrated to provide 2-(1-benzylpiperidin-4-yl)ethyl chloroformate 12a (0.40 g, 1.4 mmol, 62%) as an oil.
  • Step 12B: 2-(1-benzylpiperidin-4-yl)ethyl 4-[4-(trifluoromethoxy)phenyl]piperazine-1-carboxylate
  • To a solution of 1-[4-(trifluoromethoxy)phenyl]piperazine if (0.10 g, 0.41 mmol, 1.0 eq) and 12a (0.13 g, 0.45 mmol, 1.1 eq) in methylene chloride (3 mL) was added triethylamine (0.17 mL, 1.2 mmol, 3.0 eq) and the reaction mixture stirred at RT. After 10 min, the reaction mixture was diluted with methylene chloride, washed with sat. NH4Cl, dried over MgSO4 and concentrated. A silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-15%) in methylene chloride over 20 min to provide 2-(1-benzylpiperidin-4-yl)ethyl 4-[4-(trifluoromethoxy)phenyl]piperazine-1-carboxylate 12-1 (0.15 g, 0.31 mmol, 75%) as a foam. The table below provides the observed (Obs) ion m/z ratio for 12-1 (first compound listed in Table 10) and other compounds that were made according to the procedure as described in this example.
  • TABLE 10
    Obs Ion
    Cpd. No. Compound Name (m/z)
    12-1 2-(1-benzylpiperidin-4-yl)ethyl 4-[4- 492.2
    (trifluoromethoxy)phenyl]piperazine-1-carboxylate
    12-2 2-(1-benzylpiperidin-4-yl)ethyl 4-(3-cyanophenyl)piperazine-1- 433.25
    carboxylate
    12-3 2-(1-benzylpiperidin-4-yl)ethyl 4-(4-fluorophenyl)piperazine-1- 426.3
    carboxylate
    12-4 2-(1-benzylpiperidin-4-yl)ethyl 4-[3- 492.7
    (trifluoromethoxy)phenyl]piperazine-1-carboxylate
    12-5 2-(1-benzylpiperidin-4-yl)ethyl N-[1-(3-cyanophenyl)piperidin- 447.3
    4-yl]carbamate
    12-6 2-(1-benzylpiperidin-4-yl)ethyl N-[1-(4-fluorophenyl)piperidin- 440.3
    4-yl]carbamate
    12-7 2-(1-benzylpiperidin-4-yl)ethyl 4-hydroxy-4-phenylpiperidine-1- 423.1
    carboxylate
    12-8 2-(1-benzylpiperidin-4-yl)ethyl 4-phenylpiperidine-1-carboxylate 407.2
    12-9 2-(1-benzylpiperidin-4-yl)ethyl 4-(3-cyano-5- 451.2
    fluorophenyl)piperazine-1-carboxylate
    12-10 2-(1-benzylpiperidin-4-yl)ethyl 4-(3-cyano-4- 451.3
    fluorophenyl)piperazine-1-carboxylate
    12-11 2-(1-benzylpiperidin-4-yl)ethyl N-{1-[4- 506.2
    (trifluoromethoxy)phenyl]piperidin-4-yl}carbamate
    12-12 2-(1-benzylpiperidin-4-yl)ethyl 4-(3,4,5- 462.2
    trifluorophenyl)piperazine-1-carboxylate
  • Example 13
  • Figure US20210338653A1-20211104-C00033
  • Step 13A: Ethyl (4E)-1-benzyl-4-(cyanomethylidene)piperidine-3-carboxylate
  • To a solution of diethyl cyanomethylphosphonate (0.5 g, 2.8 mmol, 1.2 eq) in anhydrous THF (5 mL) was added K2CO3 (0.4 g, 2.8 mmol, 1.2 eq) and the mixture was stirred at room temp for 15 min, then heated to reflux for 20 min. The mixture was cooled and ethyl 1-benzyl-4-oxopiperidine-3-carboxylate (0.6 g, 2.3 mmol, 1.0 eq) was added and refluxed for 12 hours. The mixture was cooled, diluted with EtOAc (10 mL) and washed with sat. NaHCO3. The organic layer was dried over MgSO4, filtered and concentrated. The crude product was purified by via ISCO chromatography with an increasing gradient from 0% to 60% EtOAc in hexanes. Two peaks were combined as they will merge on reduction to a racemic mixture to give ethyl (4E)-1-benzyl-4-(cyanomethylidene)piperidine-3-carboxylate 13a (0.4 g, 1.4 mmol, 50% yield).
  • Step 13B: [4-(2-aminoethyl)-1-benzylpiperidin-3-yl]methanol
  • (4E)-1-benzyl-4-(cyanomethylidene)piperidine-3-carboxylate (0.4 g, 1.4 mmol, 1.0 eq) was dissolved in anhydrous THF (10 mL) and LAH (0.14 g, 4.23 mmol, 3.0 eq) was added and the mixture stirred for one hour. The reaction was quenched with H2O (0.5 mL) and diluted with EtOAc and filtered. The organic layer was concentrated to give [4-(2-aminoethyl)-1-benzylpiperidin-3-yl]methanol 13b (0.35 g, 1.4 mmol, 100% yield).
  • Example 14
  • Figure US20210338653A1-20211104-C00034
  • Step 14A: 2-(1-benzyl-4-hydroxypiperidin-4-yl)acetonitrile
  • n-BuLi (2 M in pentane, 7.3 mL, 1.1 eq) was added dropwise to solution of MeCN (0.76 mL) in THF (6 mL) at −78° C. and stirred 20 minutes. Next, a solution of 1-benzylpiperidine-4-one (2.5 g, 13.2 mmol, 1.0 eq) in 4 mL THF was added dropwise at −78° C. The reaction mixture was warmed to room temperature slowly overnight. The mixture was diluted with EtOAc and extracted from water. The crude product was purified via ISCO silica chromatography eluting with a gradient of 0% to 50% (80-18-2 CHCl3/MeOH/NH3) in DCM to give 2-(1-benzyl-4-hydroxypiperidin-4-yl)acetonitrile 14a (2.6 g, 11.4 mmol) in a 86% yield.
  • Step 14B: 4-(2-aminoethyl)-1-benzylpiperidin-4-ol
  • 2-(1-Benzyl-4-hydroxypiperidin-4-yl)acetonitrile 14a (2.6 g, 11.4 mmol, 1.0 eq) was dissolved in anhydrous THF (30 mL) and LAH (1.1 g, 28 mmol, 2.5 eq) was added. The reaction mixture was stirred at room temp for 30 min then slowly quenched with water and excess EtOAc. The reaction was filtered through celite and concentrated to give 4-(2-aminoethyl)-1-benzylpiperidin-4-ol 14b (1.9 g, 7.9 mmol) in a 72% yield.
  • Example 15
  • Figure US20210338653A1-20211104-C00035
  • Step 15A: Ethyl 1-benzyl-4-(cyanomethyl)piperidine-4-carboxylate
  • Ethyl 1-benzylpiperidine-4-carboxylate (2.0 g, 8.1 mmol, 1.0 eq) was dissolved in anhydrous THF (20 mL) and cooled to −78° C. Next, LDA (2.0M in THF, 8.1 mL, 2.0 eq) was added dropwise and stirred for one hour at −78° C. A solution of bromoacetonitrile (1.9 g, 16.2 mmol, 2.0 eq) in THF (10 mL) was added dropwise at 78° C. and stirred at this temperature for 3 hours then warmed to room temperature. The reaction mixture was diluted with EtOAc and washed with sat. NH4Cl and brine. The organic layer was dried over MgSO4, filtered and concentrated. The crude product was purified via ISCO silica chromatography eluting with an increasing gradient of EtOAc (0% to 100%) in hexanes to give ethyl 1-benzyl-4-(cyanomethyl)piperidine-4-carboxylate 15a (1.3 g, 4.5 mmol) in a 56% yield.
  • Step 15B: [4-(2-Aminoethyl)-1-benzylpiperidin-4-yl]methanol
  • Ethyl 1-benzyl-4-(cyanomethyl)piperidine-4-carboxylate 15a (0.9 g, 3.1 mmol, 1.0 eq) was dissolved in anhydrous THF (10 mL) and LAH (0.21 g, 6.3 mmol, 2.0 eq) was added. The mixture was stirred at room temp for 30 min then slowly quenched with water and excess EtOAc. The reaction was filtered through celite and concentrated to give [4-(2-aminoethyl)-1-benzylpiperidin-4-yl]methanol 15b (0.77 g, 3.1 mmol) in a quantitative yield.
  • Example 16
  • Figure US20210338653A1-20211104-C00036
  • Step 16A: Benzyl 4-(oxiran-2-yl)piperidine-1-carboxylate
  • Trimethylsulfoxonium iodide (13.5 g, 60.7 mmol, 1.5 eq) was added in two portions to a solution of NaH (1.5 g, 60.7 mmol, 1.5 eq) in anhydrous DMSO (15 mL). This mixture was stirred for one hour at room temp. Then a solution of benzyl 4-formylpiperidine-1-carboxylate (10.0 g, 40.4 mmol, 1.0 eq) in anhydrous DMSO (20 mL) was added and the mixture stirred two hours. The reaction was poured into water and extracted twice with Et2O. The organic layer was dried over MgSO4, filtered, and concentrated. The crude product was purified on ISCO with 0% to 70% EtOAC in hexanes to give benzyl 4-(oxiran-2-yl)piperidine-1-carboxylate 16a as a clear oil (6.8 g, 26.1 mmol) in a 65% yield.
  • Step 16B: Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate
  • (S,S)-(+)-N,N′-Bis(3,5-di-tert-butylsalicylidene)-1,2-cyclohexanediaminocobalt(II) [88264-84-8] (0.40 g, 0.65 mmol, 0.02 eq) was dissolved in toluene (10 mL) and acetic acid (0.07 mL, 1.3 mmol, 0.04 eq) was added and the mixture stirred for one hour. The mixture was then concentrated and dried on a high vacuum to give a solid. Benzyl 4-(oxiran-2-yl)piperidine-1-carboxylate 16a (6.8 g, 26.1 mmol, 1.0 eq) was dissolved in anhydrous THF (15 mL) and the catalyst dissolved in minimal anhydrous THF was added and cooled to 0° C. and water (0.26 mL, 14.6 mmol, 0.6 eq) was added dropwise. The reaction slowly warmed to room temperature and stirred overnight. LCMS showed slow reaction so additional water (0.05 mL, 2.8 mmol, 0.1 eq) was added and the reaction was stirred overnight. The reaction mixture was concentrated and purified by ISCO column chromatography eluting with a gradient of 0% to 70% EtOAc in hexanes. Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate 16b (3.4 g, 12.9 mmol) was determined to be 99% ee by chiral chromatography compared to the racemic mixture.
  • Step 16C: Benzyl 4-[(1S)-2-azido-1-hydroxyethyl]piperidine-1-carboxylate
  • Benzyl 4-[(2S)-oxiran-2-yl]piperidine-1-carboxylate 16b (3.4 g, 12.9 mmol, 1.0 eq) was dissolved in EtOH (9 mL) and water (1 mL) and sodium azide (1.7 g, 25.8 mmol, 2.0 eq) and ammonium chloride (1.4 g, 25.8 mmol, 2.0 eq) were added. The reaction mixture was heated to 55° C. and stirred overnight. EtOH was removed in vacuo and the reaction mixture was diluted with DCM and extracted from sat. NaHCO3. The organic layer was dried over MgSO4, filtered and concentrated to give benzyl 4-[(1S)-2-azido-1-hydroxyethyl]piperidine-1-carboxylate 16c (3.9 g, 12.7 mmol) in 98% yield.
  • Step 16D: Benzyl 4-[(1S)-2-{[(tert-butoxy)carbonyl]amino}-1-hydroxyethyl]-piperidine-1-carboxylate
  • Benzyl 4-[(1S)-2-azido-1-hydroxyethyl]piperidine-1-carboxylate 16c (3.9 g, 12.7 mmol, 1.0 eq) was dissolved in anhydrous MeOH (100 mL) then dichloronickel hexahydrate (3.0 g, 12.7 mmol, 1.0 eq) and NaBH4 (0.96 g, 25.4 mmol, 2.0 eq) were added and the mixture stirred for one hour. MeOH was removed in vacuo and the product was redissolved in DCM, filtered through celite and concentrated. The crude product was then redissolved in DCM (100 mL) and triethylamine (5.3 mL, 38.1 mmol, 3.0 eq) and di-tert-butyl dicarbonate (5.5 g, 25.4 mmol, 3.0 eq) were added and the reaction was stirred overnight. An additional 0.2 equiv of TEA and di-tert-butyl dicarbonate were added along with MeOH (10 mL) for solubility and reaction stirred for one hour. Solvent was removed in vacuo and crude product redissolved in DCM and washed with sat. NH4Cl. The organic layer was dried over MgSO4, filtered and concentrated. The crude product was purified by ISCO column chromatography elution with a 0% to 100% of EtOAc in hexanes to afford benzyl 4-[(1S)-2-{[(tert-butoxy)carbonyl]amino}-1-hydroxyethyl]piperidine-1-carboxylate 16d (3.98 g, 10.5 mmol) in a 83% yield.
  • Step 16E: (1S)-2-amino-1-(1-benzylpiperidin-4-yl)ethan-1-ol
  • Benzyl 4-[(1S)-2-{[(tert-butoxy)carbonyl]amino}-1-hydroxyethyl]piperidine-1-carboxylate 16d (3.98 g, 10.5 mmol, 1.1 eq) was dissolved in MeOH and 10% Pd/C was added. The sealed vessel was purged with H2 and the reaction was stirred one hour. The reaction was filtered through celite and concentrated to afford the crude amine which was used without further purification. The crude amine (2.4 g, 9.8 mmol, 1.0 eq) was dissolved in EtOH (75 mL) and acetic acid (0.55 mL, 10.5 mmol, 1.1 eq) and benzaldehyde (1.6 mL, 15.8 mmol, 1.6 eq) were added followed by sodium cyanoborohydride (1.0 g, 15.8 mmol, 1.6 eq) and the reaction was stirred for 2 hours Additional benzaldehyde (1.0 mL, 8.0 mmol) was added and reaction complete after one hour of stirring. The reaction mixture was quenched with water and MeOH was removed in vacuo. The reaction mixture was redissolved in DCM and extracted with sat. NaHCO3 (2×). The organic layer was dried over MgSO4 and filtered and concentrated. The crude product was purified by ISCO column chromatography eluting with a gradient of MeOH from 0% to 30% in DCM to yield the benzyl protected amine (3.1 g, 9.3 mmol) in a 95% yield. Next, the benzylamine (3.1 g, 9.3 mmol, 1.0 eq) was dissolved in DCM (50 mL) and TFA (10 mL) was added and the reaction mixture was stirred for one hour. The reaction mixture was concentrated and redissolved in MeOH and MP-carbonate resin was added until the solution was basic. The reaction was filtered and concentrated to afford (1S)-2-amino-1-(1-benzylpiperidin-4-yl)ethan-1-ol 16e (2.1 g, 9.0 mmol) in a 97% yield.
  • (1R)-2-Amino-1-(1-benzylpiperidin-4-yl)ethan-1-ol 16f was synthesized following the same overall procedure but using (R,R)-(±)-N,N′-Bis(3,5-di-tert-butylsalicylidene)-1,2-cyclohexanediaminocobalt(II) in Step 16B.
  • Example 17
  • Figure US20210338653A1-20211104-C00037
  • Step 17A: Ethyl 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylate
  • A solution of 3-bromo-1H-pyrazol-5-amine (4.2 g, 26 mmol, 1.0 eq) and ethyl (3Z)-3-[(dimethylamino)methylidene]-4-oxopentanoate (4.9 g, 26 mmol, 1.0 eq) in ethanol (300 mL) was prepared and heated to reflux for 2 hrs. The reaction mixture was concentrated. Silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-5%) in methylene chloride over 20 min to provide ethyl 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylate 17a (6.0 g, 21 mmol, 81%) as a white solid.
  • Step 17B: 7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxylic acid
  • To a solution of ethyl 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylate 17a (2.0 g, 7.0 mmol, 1.0 eq) in anhydrous dioxane (45 mL) was added [3-(trifluoromethyl)phenyl]boronic acid (2.0 g, 10 mmol, 1.5 eq) followed by aq. K2CO3 (7 mL, 22.5 mmol, 3.2 eq) and the resulting solution purged with nitrogen for 10 min. Then, [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.52 g, 0.70 mmol, 0.1 eq) was added and the reaction heated to 90° C. overnight. The resulting dark reaction mixture was cooled, diluted heavily with EtOAc, and washed with brine. The organic layer was washed again with brine, dried over Na2SO4, and concentrated. Silica gel column (80 g) was loaded using methylene chloride and run using an increasing gradient of EtOAc (5-95%) in hexanes over 20 min. The chromatographed material was then suspended in MeOH (50 mL) and treated with an aq. solution of 1 M LiOH (5 mL). After stirring at RT overnight, the dark orange suspension was concentrated and the resulting solid re-dissolved in water. Then, the suspension was made acidic with careful addition of 6 M HCl. The precipitate was collected and dried to provide 7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxylic acid 17b (1.6 g, 4.9 mmol, 70% over two steps) as a tan solid.
  • Step 17C: N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • To a solution of 7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo-[1,5-a]pyrimidine-6-carboxylic acid 17b (1.6 g, 4.9 mmol, 1.0 eq) and 2-(1-benzylpiperidin-4-yl)ethan-1-amine (1.1 g, 4.9 mmol, 1.0 eq) in NMP (20 mL) was added triethylamine (2.7 mL, 20 mmol, 4.0 eq) followed by HATU (1.9 g, 4.9 mmol, 1.0 eq) and the reaction stirred at room temperature overnight. The resulting dark reaction mixture was diluted heavily with EtOAc and washed repeatedly with brine. During the course of the extraction, a large amount of orange precipitate formed; thus after evaporating all organic solvent, the aqueous layers were left to sit overnight. Then, all precipitate was collected by vacuum filtration and washed with water. Silica gel column (120 g) was dry loaded and run using MeOH (0-20%) in DCM over 25 min to provide N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide 17-1 (1.5 g, 2.9 mmol, 59%) as an off-white solid. The table below provides the observed (Obs) ion m/z ratio for 17-1 (first compound listed in Table 11) and other compounds that were made according to the procedure as described in this example.
  • TABLE 11
    Obs Ion
    Cpd. No. Compound Name (m/z)
    17-1 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[3- 522.2
    (trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
    17-2 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2- 454.2
    phenylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    17-3 N-[2-(1-benzyl-4-hydroxypiperidin-4-yl)ethyl]-7-methyl-2-[3- 538.14
    (trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
    17-4 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-chlorophenyl)-7- 488.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    17-5 N-[(2S)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-7-methyl-2- 538.1
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    17-6 N-[(2R)-2-(1-benzylpiperidin-4-yl)-2-hydroxyethyl]-7-methyl-2- 538.1
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    17-7 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-(thiophen-2- 460.3
    yl)pyrazolo[1,5-a]pyrimidine-6-carboxamide
    17-8 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[4- 522.2
    (trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Example 18
  • Figure US20210338653A1-20211104-C00038
  • Step 18A: N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
  • To a solution of ethyl 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylate 17a (1.0 g, 3.6 mmol, 1.0 eq) in THE/water (4:1) was added solid NaOH (0.21 g, 5.3 mmol, 1.5 eq). After stirring at RT overnight, the organic solvent was concentrated and the remaining aqueous phase made acidic with careful addition of 6 M HCl. The resulting precipitate was collected and dried to provide 2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxylic acid 18a. A portion of this material (0.70 g, 2.7 mmol, 1.0 eq) together with 2-(1-benzylpiperidin-4-yl)ethan-1-amine (0.90 g, 4.1 mmol, 1.5 eq) in methylene chloride/DMF 2:1 (15 mL) was treated with triethylamine (1.5 mL, 11 mmol, 4.0 eq) and HATU (1.4 g, 3.6 mmol, 1.0 eq) and stirred at RT overnight. The reaction mixture was diluted with methylene chloride, washed with sat. NH4Cl, dried over MgSO4 and concentrated. Silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-20%) in methylene chloride over 20 min to provide N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide 18b (1.1 g, 2.6 mmol, 94%).
  • Step 18B: N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,4-difluorophenyl)-7-methyl-pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • To an NMP solution of N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-bromo-7-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide 18b (0.20 mL, 0.13 M, 1.0 eq) was added a spatula tip of (2,4-difluorophenyl)boronic acid followed by aq. K3PO4 (0.05 mL, 1.5 M, 2.9 eq). Then, a generous spatula tip of PS—Pd(PPh3) was added and the reaction mixture heated to 90° C. In general, this reaction and others of its kind are complete within 2 hrs, however additional PS—Pd(PPh3) and/or longer reaction times can be used to push the coupling. The resulting dark suspension was cooled, passed through an HPLC filter, diluted to 1 mL using MeOH, and submitted for directly for preparative chromatography to yield N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,4-difluorophenyl)-7-methyl-pyrazolo[1,5-a]pyrimidine-6-carboxamide 18-1. The table below provides the observed (Obs) ion m/z ratio for 18-1 (first compound listed in Table 12) and other compounds that were made according to the procedure as described in this example.
  • TABLE 12
    Obs Ion
    Cpd. No. Compound Name (m/z)
    18-1 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,4-difluorophenyl)-7- 490.15
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-2 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-(pyridin-2- 456.0
    yl)pyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-3 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-cyanophenyl)-7- 479.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-4 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-fluorophenyl)-7- 472.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-5 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(3,5-difluorophenyl)-7- 490.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-6 N-[2-(1-benzylpiperidin-4-yl)ethyl]2-(3-fluorophenyl)-7- 472.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-7 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,3-difluorophenyl)-7- 490.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-8 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(3-cyanophenyl)-7- 479.1
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-9 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2-methoxyphenyl)-7- 484.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-10 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2,5-difluorophenyl)-7- 490.15
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-11 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[2- 522.1
    (trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    18-12 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(4-cyano-3-fluorophenyl)- 497.2
    7-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-13 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[4- 538.2
    (trifluoromethoxy)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    18-14 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2-methoxypyridin-3-yl)-7- 485.3
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-15 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(6-fluoropyridin-3-yl)-7- 473.1
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-16 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-[3- 538.1
    (trifluoromethoxy)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    18-17 N-[2-(1-benzylpiperidin-4-yl)ethyl]-7-methyl-2-(3,4,5- 508.1
    trifluorophenyl)pyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-18 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2-fluoropyridin-3-yl)-7- 473.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    18-19 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(6-methoxy-4- 499.2
    methylpyridin-3-yl)-7-methylpyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    18-20 N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-(2-fluoropyridin-4-yl)-7- 473.2
    methylpyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Example 19
  • Figure US20210338653A1-20211104-C00039
  • Step 19A: 7-methyl-N-[2-(piperidin-4-yl)ethyl]-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • To a solution of 7-methyl-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxylic acid 17b (0.09 g, 0.27 mmol, 1.0 eq) and tert-butyl 4-(2-aminoethyl)piperidine-1-carboxylate (0.07 g, 0.32 mmol, 1.2 eq) in methylene chloride was added triethylamine (0.11 mL, 0.81 mmol, 3.0 eq) followed by HATU (0.12 g, 0.32 mmol, 1.2 eq) and the reaction stirred at room temperature overnight. The reaction mixture diluted with sat. NH4Cl and extracted with methylene chloride. The combined organic layers were dried over MgSO4 and concentrated. Silica gel column was loaded using methylene chloride and run using an increasing gradient of MeOH (0-10%) in methylene chloride yielding tert-butyl 4-[2-({7-methyl-2-[3-(trifluoromethyl)phenyl]-pyrazolo[1,5-a]pyrimidin-6-yl}formamido)ethyl]piperidine-1-carboxylate 19a. The chromatographed 19a was dissolved in 20% TFA in methylene chloride and stirred at RT overnight. The reaction mixture was concentrated, dissolved in MeOH, and made basic with the addition of MP-carbonate. Following removal of the resin and concentration of the filtrate, the free base of 7-methyl-N-[2-(piperidin-4-yl)ethyl]-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide 19b (0.08 g, 0.19 mmol, 70% over two steps) was isolated as an oil.
  • Step 19B: 7-methyl-N-{2-[1-(pyridin-3-ylmethyl)piperidin-4-yl]ethyl}-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide
  • To NMP solutions of 7-methyl-N-[2-(piperidin-4-yl)ethyl]-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide 19b (0.05 mL, 0.50 M, 1 eq) and pyridine-3-carbaldehyde (0.05 mL, 0.50 M, 1 eq) was added an ethanolic solution of borane-pyridine complex (0.10 mL, 0.50 M, 2 eq) followed by acetic acid (5 μL) and the mixture stirred at RT overnight. The reaction was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding 7-methyl-N-{2-[1-(pyridin-3-ylmethyl)piperidin-4-yl]ethyl}-2-[3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-carboxamide 19-1. The table below provides the observed (Obs) ion m/z ratio for 19-1 (first compound listed in Table 13) and other compounds that were made according to the procedure as described in this example.
  • TABLE 13
    Obs Ion
    Cpd. No. Compound Name (m/z)
    19-1 7-methyl-N-{2-[1-(pyridin-3-ylmethyl)piperidin-4-yl]ethyl}-2- 523.1
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    19-2 N-{2-[1-(2,3-dihydro-1,4-benzodioxin-5-ylmethyl)piperidin-4- 512.2
    yl]ethyl}-7-methyl-2-phenylpyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    19-3 N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl}-7-methyl-2- 528.2
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    19-4 N-(2-{1-[(2-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-7- 470.1
    methyl-2-phenylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    19-5 7-methyl-N-{2-[1-(pyridin-4-ylmethyl)piperidin-4-yl]ethyl}-2- 523.2
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    19-6 7-methyl-N-{2-[1-(pyridin-2-ylmethyl)piperidin-4-yl]ethyl}-2- 523.2
    [3-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrimidine-6-
    carboxamide
    19-7 N-(2-{1-[(3-methoxyphenyl)methyl]piperidin-4-yl}ethyl)-7- 484.3
    methyl-2-phenylpyrazolo[1,5-a]pyrimidine-6-carboxamide
    19-8 N-(2-{1-[(4-hydroxyphenyl)methyl]piperidin-4-yl}ethyl)-7- 470.2
    methyl-2-phenylpyrazolo[1,5-a]pyrimidine-6-carboxamide
  • Example 20
  • Figure US20210338653A1-20211104-C00040
  • Step 20A: (3R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyrimidin-2-yl)-3-methylpiperidine-4-carboxamide
  • To NMP solutions of (3R,4R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methylpiperidine-4-carboxamide 10c (0.05 mL, 0.50 M, 1 eq) and 2-chloro-5-cyanopyrimidine (0.025 mmol, 1 eq) was added a NMP solution of trimethylamine (0.100 mL, 1.0 M, 4 eq) and the mixture stirred at 100° C. overnight. The reaction was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (3R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyrimidin-2-yl)-3-methylpiperidine-4-carboxamide 20-1. The table below provides the observed (Obs) ion m/z ratio for 20-1 (first compound listed in Table 13) and other compounds that were made according to the procedure as described in this example.
  • TABLE 14
    Obs Ion
    Cpd. No. Compound Name (m/z)
    20-1 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-cyanopyrimidin-2- 447.2
    yl)-3-methylpiperidine-4-carboxamide
    20-2 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-3-methyl-1-[5- 490.14
    (trifluoromethyl)pyrimidin-2-yl]piperidine-4-carboxamide
    20-3 (3R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-1-(5-chloropyrimidin-2- 456.1
    yl)-3-methylpiperidine-4-carboxamide
  • Example 21
  • Figure US20210338653A1-20211104-C00041
  • Step 21A: (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl}-2,6-dimethylpiperazine-1-carboxamide
  • To NMP solutions of (2S,6R)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-[2-(piperidin-4-yl)ethyl]piperazine-1-carboxamide 6c (0.20 mL, 0.12 M, 1.0 eq) and N,N-diisopropylethylamine (0.05 mL, 0.5 M, 4.0 eq) was added (bromomethyl)cyclohexane (4.4 mg, 0.025 mmol, 1.0 eq) and the reaction mixture heated at 50° C. overnight. The reaction mixture was diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[1-(cyclohexylmethyl)piperidin-4-yl]ethyl}-2,6-dimethylpiperazine-1-carboxamide 21-1. The table below provides the observed (Obs) ion m/z ratio for 21-1 (first compound listed in Table 15) and other compounds that were made according to the procedure as described in this example.
  • TABLE 15
    Obs Ion
    Cpd. No. Compound Name (m/z)
    21-1 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[1- 468.1
    (cyclohexylmethyl)piperidin-4-yl]ethyl}-2,6-
    dimethylpiperazine-1-carboxamide
    21-2 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-(2-{1-[α,α- 469.1
    2H-benzyl]piperidin-4-yl}ethyl)piperazine-1-carboxamide
    21-3 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1- 463.1
    (pyridin-2-ylmethyl)piperidin-4-yl]ethyl}piperazine-1-
    carboxamide
    21-4 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[l-(2,3-dihydro-1H- 488.1
    inden-2-yl)piperidin-4-yl]ethyl}-2,6-dimethylpiperazine-1-
    carboxamide
    21-5 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-{2-[1- 440.4
    (cyclobutylmethyl)piperidin-4-yl]ethyl}-2,6-dimethylpiperazine-
    1-carboxamide
    21-6 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1-(2- 428.1
    methylpropyl)piperidin-4-yl]ethyl}piperazine-1-carboxamide
    21-7 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-(2-{1-[2- 472.1
    (trimethylsilyl)ethyl]piperidin-4-yl}ethyl)piperazine-1-
    carboxamide
    21-8 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1-(3- 442.1
    methylbutyl)piperidin-4-yl]ethyl}piperazine-1-carboxamide
    21-9 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-{2-[1-(oxan- 470.1
    2-ylmethyl)piperidin-4-yl]ethyl}piperazine-1-carboxamide
    21-10 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-(2-{1-[(3- 456.1
    methyloxetan-3-yl)methyl]piperidin-4-yl}ethyl)piperazine-1-
    carboxamide
    21-11 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-N-(2-{1-[(1,1-dioxo-1-thian- 518.1
    3-yl)methyl]piperidin-4-yl}ethyl)-2,6-dimethylpiperazine-1-
    carboxamide
    21-12 (2R,6S)-N-[2-(1-{bicyclo[1.1.1]pentan-1-ylmethyl}piperidin-4- 452.1
    yl)ethyl]-4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-
    carboxamide
    21-13 (2R,6S)-N-{2-[1-(cuban-1-ylmethyl)piperidin-4-yl]ethyl}-4-(5- 488.1
    cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide
    21-14 (2R)-2-methyl-N-(2-{1-[α,α-2H-benzyl]piperidin-4-yl}ethyl)-4- 493.1
    [5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    21-15 (2R,6S)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethyl-N-(2-{1-[α,α- 464.2
    2H-benzyl]piperidin-4-yl}ethyl)piperazine-1-carboxamide
    21-16 (2R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]- 507.3
    2-methyl-N-(2-{1-[α,α-
    2H-benzyl]piperidin-4-yl}ethyl)piperazine-1-carboxamide
    21-17 (2R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-2-methyl-N- 512.3
    (2-{1-[α,α-2H-benzyl]piperidin-4-yl}ethyl)piperazine-1-
    carboxamide
  • Example 22
  • Figure US20210338653A1-20211104-C00042
  • Step 22A: (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide
  • To a solid mixture of (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide 4b (15 mg, 0.04 mmol, 1.0 eq), 5-bromo-2-(trifluoromethyl)pyrimidine (9.4 mg, 0.04 mmol, 1.0 eq), sodium tert-butoxide (11 mg, 0.12 mmol, 3.0 eq), and lastly bis(tri-tert-butylphosphine)palladium(0) (3.1 mg, 0.006 mmol, 0.15 eq) was added dioxane (1 mL) and reaction mixture stirred vigorously at 50° C. overnight. The resulting dark suspension was cooled, passed through an HPLC filter and concentrated in vacuo. The crude material was treated with 1.5 mL of MeOH, passed through an additional HPLC filter (leaving any precipitate behind), and purified by preparative chromatography yielding (2R,6S)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2-(trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide 22-1. The table below provides the observed (Obs) ion m/z ratio for 22-1 (first compound listed in Table 16) and other compounds that were made according to the procedure as described in this example. For some less reactive halides, methanesulfanato(2-di-t-butylphosphino-2′,4′,6′-tri-1-propyl-1,1′-biphenyl)(2′ amino-1,1′-biphenyl-2-yl) palladium (II) was used in place of bis(tri-tert-butylphosphine)palladium(0). These reactions were carried out at 100° C. for 1-2 hrs.
  • TABLE 16
    Obs Ion
    Cpd. No. Compound Name (m/z)
    22-1 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2- 505.1
    (trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide
    22-2 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyanophenyl)- 460.1
    2,6-dimethylpiperazine-1-carboxamide
    22-3 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-2- 478.1
    fluorophenyl)-2,6-dimethylpiperazine-1-carboxamide
    22-4 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3- 478.1
    fluorophenyl)-2,6-dimethylpiperazine-1-carboxamide
    22-5 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3,5- 472.1
    difluoropyridin-2-y1)-2,6-dimethylpiperazine-1-carboxamide
    22-6 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4- 489.2
    (2,4,5-trifluorophenyl)piperazine-1-carboxamide
    22-7 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-chloro-4,5- 505.1
    difluorophenyl)-2,6-dimethylpiperazine-1-carboxamide
    22-8 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-chloro-5- 488.0
    fluoropyridin-3-yl)-2,6-dimethylpiperazine-1-carboxamide
    22-9 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3,5- 496.1
    difluorophenyl)-2,6-dimethylpiperazine-1-carboxamide
    22-10 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-2,5- 482.1
    difluorophenyl)-2-methylpiperazine-1-carboxamide
    22-11 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-chloropyridin-3- 456.1
    yl)-2-methylpiperazine-1-carboxamide
    22-12 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3,5- 482.1
    difluorophenyl)-2-methylpiperazine-1-carboxamide
    22-13 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-3,5- 491.1
    difluorophenyl)-2-methylpiperazine-1-carboxamide
    22-14 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-chloro-3- 473.1
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    22-15 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3- 464.1
    fluorophenyl)-2-methylpiperazine-1-carboxamide
    22-16 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-methoxypyridin- 452.1
    4-yl)-2-methylpiperazine-1-carboxamide
    22-17 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4-cyano-3- 530.1
    (trifluoromethoxy)phenyl]-2-methylpiperazine-1-carboxamide
    22-18 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-fluoropyridin-4- 440.1
    yl)-2-methylpiperazine-1-carboxamide
    22-19 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4- 423.1
    (pyrimidin-5-yl)piperazine-1-carboxamide
    22-20 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2- 453.0
    methoxypyrimidin-5-yl)-2-methylpiperazine-1-carboxamide
    22-21 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[2- 466.3
    (dimethylamino)pyrimidin-5-yl]-2-methylpiperazine-1-
    carboxamide
    22-22 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5- 491.1
    (trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide
    22-23 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5- 505.1
    (trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide
    22-24 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-4- 490.1
    methoxyphenyl)-2,6-dimethylpiperazine-1-carboxamide
    22-25 N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[5- 476.9
    (trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide
    22-26 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4-cyano-3- 489.1
    (dimethylamino)phenyl]-2-methylpiperazine-1-carboxamide
    22-27 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrazin-2- 448.1
    yl)-2-methylpiperazine-1-carboxamide
    22-28 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3- 476.0
    methoxyphenyl)-2-methylpiperazine-1-carboxamide
    22-29 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[2- 490.0
    (trifluoromethyl)pyridin-4-yl]piperazine-1-carboxamide
    22-30 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2,6-difluoropyridin- 458.1
    4-yl)-2-methylpiperazine-1-carboxamide
    22-31 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-[4-cyano-3- 514.1
    (trifluoromethyl)phenyl]-2-methylpiperazine-1-carboxamide
    22-32 (2R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(4-cyano-3-fluoro-5- 494.1
    methoxyphenyl)-2-methylpiperazine-1-carboxamide
  • Example 23
  • Figure US20210338653A1-20211104-C00043
  • Step 23A: (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin-5-yl)-2,6-dimethylpiperazine-1-carboxamide
  • To a solid mixture of (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide 4b (30 mg, 0.08 mmol, 1.0 eq), 5-bromopyrimidine-2-carbonitrile (22 mg, 0.12 mmol, 1.5 eq) and cesium carbonate (39 mg, 0.12 mmol, 1.5 eq) was added NMP (1 mL) and reaction mixture stirred at 45° C. over the weekend. The resulting suspension was cooled, passed through an HPLC filter diluting to 1 mL with MeOH and submitted for directly for preparative chromatography yielding (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin-5-yl)-2,6-dimethylpiperazine-1-carboxamide 23-1. The table below provides the observed (Obs) ion m/z ratio for 23-1 (first compound listed in Table 17) and other compounds that were made according to the procedure as described in this example.
  • TABLE 17
    Obs Ion
    Cpd. No. Compound Name (m/z)
    23-1 (2S,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(2-cyanopyrimidin- 462.2
    5-yl)-2,6-dimethylpiperazine-1-carboxamide
    23-2 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrazin-2- 462.1
    yl)-2,6-dimethylpiperazine-1-carboxamide
    23-3 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(6-cyanopyridazin- 462.1
    3-yl)-2,6-dimethylpiperazine-1-carboxamide
  • Example 24
  • Figure US20210338653A1-20211104-C00044
  • Step 24A: 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]pyrimidine-5-carbonitrile
  • To a suspension of (2R,6R)-2,6-dimethylpiperazine dihydrochloride (0.250 g, 1.34 mmol, 1.0 eq) and 2-chloropyrimidine-5-carbonitrile (0.187 g, 1.34 mmol, 1.0 eq) in acetonitrile (5 mL) was added triethylamine (0.93 mL, 6.7 mmol, 5.0 eq) and reaction mixture stirred at room temperature overnight. The resulting suspension was filtered to remove triethylamine hydrochloride and concentrated to yield 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]pyrimidine-5-carbonitrile 24a as an orange solid. The crude material was carried to Example 26 without further purification.
  • Other compounds made using the above synthetic scheme include:
    • 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifuoromethyl)pyrimidin-4-amine 24b;
    • 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidine 24c; and 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidine
    • 2-[(3S,5S)-3,5-dimethylpiperazin-1-yl]pyrimidine-5-carbonitrile 24d.
    Example 25
  • Figure US20210338653A1-20211104-C00045
  • Step 25A: 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrazine
  • To a solid mixture of (2R,6R)-2,6-dimethylpiperazine dihydrochloride (0.08 g, 0.44 mmol, 1.0 eq), sodium tert-butoxide (0.21 g, 2.2 mmol, 5.0 eq) and bis(tri-tert-butylphosphine)palladium(0) (34 mg, 0.07 mmol, 0.15 eq) was added dioxane (4 mL) followed by 2-bromo-5-(trifluoromethyl)pyrazine (0.10 g, 0.44 mmol, 1.0 eq) and the reaction mixture stirred at 50° C. overnight. The resulting suspension was filtered thru a pad of celite using EtOAc and concentrated. Silica gel column (24 g) was loaded using methylene chloride and run using an increasing gradient of MeOH (0-20%) in methylene chloride over 25 min. Following concentration of the product eluents, 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrazine 25a (0.09 g, 0.33 mmol, 75%) was isolated as a yellow oil. The purified material was carried to Example 26.
  • 5-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-2-(trifluoromethyl)pyrimidine 25b was made in a similar fashion.
  • Example 26
  • Figure US20210338653A1-20211104-C00046
  • Step 26A: (2R,6R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
  • To a solution of phenyl N-[2-(1-benzylpiperidin-4-yl)ethyl]carbamate 8b (15 mg, 0.04 mmol, 1.0 eq) and crude 2-[(3R,5R)-3,5-dimethylpiperazin-1-yl]-5-(trifluoromethyl)pyrimidin-4-amine 24b (22 mg, 0.08 mmol, 2.0 eq) in NMP (0.50 mL) was added triethylamine (0.02 mL, 0.16 mmol, 4.0 eq) and reaction mixture stirred at 100° C. overnight. The reaction mixture was filtered and diluted to a total volume of 1 mL using MeOH and submitted directly for preparative chromatography yielding
  • (2R,6R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide 26-1. The table below provides the observed (Obs) ion m/z ratio for 26-1 (first compound listed in Table 18) and other compounds that were made according to the procedure as described in this example.
  • TABLE 18
    Obs Ion
    Cpd. No. Compound Name (m/z)
    26-1 (2R,6R)-4-[4-amino-5-(trifluoromethyl)pyrimidin-2-yl]-N-[2-(1- 520.1
    benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide
    26-2 (2R,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin- 462.1
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    26-3 (2S,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(5-cyanopyrimidin- 462.1
    2-yl)-2,6-dimethylpiperazine-1-carboxamide
    26-4 (2R,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5- 505.1
    (trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide
    26-5 (2R,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5- 505.0
    (trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide
    26-6 (2R,6R)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[2- 505.05
    (trifluoromethyl)pyrimidin-5-yl]piperazine-1-carboxamide
  • Example 27
  • Figure US20210338653A1-20211104-C00047
  • Step 27A: 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5-dimethylpiperazin-1-yl]-N-cyclopropylpyrimidine-5-carboxamide
  • To a 0.5 M NMP solution of (2S,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethylpiperazine-1-carboxamide 4b (1.0 mL, 0.50 mmol, 1.0 eq) and triethylamine (0.28 mL, 2.0 mmol, 4 eq) was added 2-chloropyrimidine-5-carboxylic acid (79 mg, 0.50 mmol, 1.0 eq) and the reaction mixture stirred at 50° C. overnight. Then, a 75 μL aliquot was treated with NMP solutions of cyclopropylamine (0.10 mL, 0.5 M, 1.3 eq), triethylamine (0.10 mL, 2.0 M, 5.2 eq) and HATU (0.10 mL, 0.5 M, 1.3 eq) and again stirred at 50° C. overnight. The reaction mixture was passed through an HPLC filter diluting to 1 mL with MeOH and purified by preparative chromatography yielding 2-[(3R,5 S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5-dimethylpiperazin-1-yl]-N-cyclopropylpyrimidine-5-carboxamide 27-1. The table below provides the observed (Obs) ion m/z ratio for 27-1 (first compound listed in Table 19) and other compounds that were made according to the procedure as described in this example.
  • TABLE 19
    Obs Ion
    Cpd. No. Compound Name (m/z)
    27-1 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 520.1
    dimethylpiperazin-1-yl]-N-cyclopropylpyrimidine-5-carboxamide
    27-2 (2R,6S)-N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5- 534.1
    (pyrrolidine-1-carbonyl)pyrimidin-2-yl]piperazine-1-carboxamide
    27-3 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 536.2
    dimethylpiperazin-1-yl]-N-methyl-N-(propan-2-yl)pyrimidine-5-
    carboxamide
    27-4 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 520.2
    dimethylpiperazin-1-yl]-N-(prop-2-en-1-yl)pyrimidine-5-
    carboxamide
    27-5 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 534.2
    dimethylpiperazin-1-yl]-N-(cyclopropylmethyl)pyrimidine-5-
    carboxamide
    27-6 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 534.1
    dimethylpiperazin-1-yl]-N-cyclobutylpyrimidine-5-carboxamide
    27-7 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 526.1
    dimethylpiperazin-1-yl]-N-(2-fluoroethyl)pyrimidine-5-
    carboxamide
    27-8 2-[(3R,5S)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3,5- 536.2
    dimethylpiperazin-1-yl]-N-(butan-2-yl)pyrimidine-5-carboxamide
    27-9 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 508.3
    methylpiperazin-1-yl]-N-ethyl-N-methylpyrimidine-5-carboxamide
    27-10 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 480.3
    methylpiperazin-1-yl]-N-methylpyrimidine-5-carboxamide
    27-11 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 536.3
    methylpiperazin-1-yl]-6-methyl-N-(2-methylpropyl)pyrimidine-4-
    carboxamide
    27-12 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 522.3
    methylpiperazin-1-yl]-N-(2-methylpropyl)pyrimidine-5-
    carboxamide
    27-13 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 522.2
    methylpiperazin-1-yl]-6-methyl-N-(propan-2-yl)pyrimidine-4-
    carboxamide
    27-14 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 520.3
    methylpiperazin-1-yl]-N-cyclopropyl-6-methylpyrimidine-4-
    carboxamide
    27-15 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 508.3
    methylpiperazin-1-yl]-N-(propan-2-yl)pyrimidine-5-carboxamide
    27-16 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 494.2
    methylpiperazin-1-yl]-N-ethylpyrimidine-5-carboxamide
    27-17 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 494.2
    methylpiperazin-1-yl]-N,N-dimethylpyrimidine-5-carboxamide
    27-18 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 522.3
    methylpiperazin-1-yl]-N-ethyl-N,6-dimethylpyrimidine-4-
    carboxamide
    27-19 2-[(3R)-4-{[2-(1-benzylpiperidin-4-yl)ethyl]carbamoyl}-3- 508.3
    methylpiperazin-1-yl]-N-ethyl-6-methylpyrimidine-4-carboxamide
  • Biology Examples Binding Assay
  • Binding affinity (Ki) of compounds was measured by inhibition of radioligand binding to membranes from CHO cells expressing human M1, M2, M3, M4 and M5 receptors. Membranes were prepared by nitrogen cavitation and differential centrifugation as previously described (Hoare et al., Mol. Pharmacol. 2003 March; 63(3): 751-65). The radioligand employed was tritiated N-methylscopolamine, used at a concentration of 1.5 nM. A dose-response of twelve concentrations of compound was used, ranging from 10 μM to 32 pM. The assay buffer was 50 mM HEPES, 100 mM NaCl, 5 mM MgCl2, 1 mM ethylenediaminetetraacetic acid, pH-adjusted to pH 7.4. Membranes, radioligand and compound were incubated together for 90 minutes at 37° C., in a total volume of 150 μl in a 96-well plate. Receptor-bound radioligand was then collected by harvesting the assay over glass fiber filters pretreated with polyethylenimine to trap the cell membranes, using rapid vacuum filtration. Harvesting and radioactivity counting was conducted as previously described (see, e.g., Hoare et al., Mot. Pharmacol. 2003 63(3):751-65); Erratum at Mol. Pharmacol. 2005 July; 68(1): 260).
  • Binding affinities of all the exemplified compounds, which are described in the examples and listed in the tables above, are less than 1 μM against the M4 receptor. More specifically, specificity for the M4 receptor for each of the compounds listed in Table 20 is as follows: (1) “+” means the compound had a Ki against the M4 receptor of less than 1 μM (1,000 nM) but greater or equal to 100 nM; (2) “++” means the compound had a Ki against the M4 receptor of less than 100 nM but greater or equal to 10 nM; and (3) “+++” means that the compound had a Ki against the M4 receptor of less than 10 nM.
  • TABLE 20
    Cpd. No. Ki
    3-1  +++
    3-2  ++
    3-3  +
    3-4  +
    3-5  +++
    3-6  ++
    3-7  ++
    3-8  +++
    3-9  +++
    3-10 +++
    3-11 +++
    3-12 +++
    3-13 +++
    3-14 +++
    3-15 ++
    3-16 +++
    3-17 +++
    3-18 +++
    3-19 +++
    3-20 ++
    3-21 +++
    3-22 +++
    3-23 +++
    4-1  +++
    4-2  +
    4-4  +
    4-5  +++
    4-6  +++
    4-8  +
    4-9  +
    4-10 +++
    4-11 +++
    4-12 +++
    4-13 +++
    4-14 +++
    4-15 +++
    4-16 +++
    4-17 +++
    4-18 +++
    4-19 +++
    4-20 +++
    4-21 +++
    4-22 +++
    4-23 +
    4-24 ++
    4-25 ++
    4-26 ++
    4-27 ++
    4-28 ++
    4-28 ++
    4-30 ++
    4-31 ++
    4-33 ++
    4-34 ++
    4-35 ++
    4-36 ++
    4-37 ++
    4-38 ++
    4-39 ++
    4-40 ++
    4-41 ++
    4-43 ++
    4-44 ++
    4-45 ++
    4-46 ++
    4-47 ++
    4-48 ++
    4-49 +
    4-51 +
    4-52 +++
    4-53 +++
    4-54 +++
    4-55 +
    4-56 +
    4-60 +++
    4-61 +
    4-62 +
    4-63 +
    4-64 +
    4-68 +++
    4-69 ++
    4-70 ++
    4-71 +++
    4-72 +++
    4-73 +++
    4-74 ++
    4-75 ++
    4-76 +++
    4-77 +++
    4-78 ++
    4-79 +++
    4-80 +++
    5-1  +++
    5-2  +
    5-3  +
    5-4  ++
    5-5  +++
    5-6  ++
    5-7  ++
    5-8  +++
    5-9  +
    5-10 +
    5-11 +
    5-12 +
    5-13 ++
    5-14 +++
    5-15 ++
    5-16 ++
    5-17 +++
    5-18 +++
    5-19 ++
    5-20 +++
    5-21 +
    5-22 +++
    5-23 +++
    5-24 +++
    5-25 +
    5-26 +
    5-27 +++
    5-28 +++
    5-29 +++
    5-30 +++
    5-31 +++
    5-32 +++
    5-33 ++
    5-34 +
    6-1  +++
    6-2  +
    6-3  +++
    6-4  +++
    6-5  ++
    6-6  ++
    6-7  ++
    6-8  ++
    6-9  ++
    6-10 ++
    6-11 ++
    6-12 ++
    6-13 ++
    6-14 ++
    6-15 ++
    6-16 ++
    6-17 ++
    6-18 ++
    6-19 +
    6-20 +
    6-21 +
    6-22 +
    6-23 +
    6-24 +
    6-25 +
    6-26 +++
    6-27 +++
    6-28 +
    6-29 +
    6-30 +
    6-31 +
    6-32 ++
    6-33 +
    6-34 +
    6-35 +++
    6-36 +++
    6-37 +++
    6-38 ++
    6-39 +
    6-40 +++
    6-41 ++
    6-42 +++
    6-43 ++
    6-44 +
    6-45 +
    6-46 +++
    6-47 +++
    6-48 +++
    6-49 +
    6-50 +++
    6-51 ++
    6-52 ++
    6-53 ++
    6-54 ++
    6-55 ++
    6-56 ++
    6-57 ++
    6-58 +
    6-59 ++
    6-60 ++
    6-61 +
    6-62 +
    6-63 +
    6-64 +
    6-65 ++
    6-66 +
    6-67 ++
    7-1  +++
    7-2  ++
    7-3  ++
    7-4  ++
    7-5  +
    7-6  +
    7-7  +
    7-8  +
    7-9  +++
    7-10 +++
    7-11 +++
    7-12 +++
    7-13 +
    8-1  +++
    8-2  ++
    8-3  ++
    8-4  ++
    8-5  +
    8-6  +
    8-7  +
    8-8  ++
    8-9  ++
    8-10 ++
    8-11 ++
    8-12 ++
    8-13 ++
    8-14 ++
    8-15 ++
    8-16 ++
    8-17 ++
    8-18 ++
    8-19 ++
    8-20 +
    8-21 +
    8-22 +
    8-23 +
    8-24 +
    8-25 +
    8-26 +
    8-27 +
    8-28 +
    8-29 +
    8-30 +
    8-31 +
    8-32 +
    8-33 +
    8-34 +
    8-35 +
    8-36 +
    8-37 +
    8-38 +
    8-39 +
    8-40 +
    8-41 +
    8-42 +
    8-43 +
    8-44 +
    8-45 +
    8-46 +
    8-47 +
    8-48 +++
    9-1  +++
    9-2  +
    9-3  +
    9-4  +
    9-5  +++
    9-6  +++
    9-7  +++
    9-8  +++
    9-9  +++
    9-10 +++
    9-11 ++
    9-12 ++
    9-13 ++
    9-14 ++
    9-15 ++
    9-16 ++
    9-17 ++
    9-18 +
    9-19 +
    9-20 +
    9-21 ++
    9-22 ++
    9-23 ++
    9-24 ++
    9-25 ++
    9-26 ++
    9-27 ++
    9-28 ++
    9-29 ++
    9-30 ++
    9-31 ++
    9-32 ++
    9-33 ++
    9-34 +
    9-35 +
    9-36 +
    9-37 +
    9-38 +
    9-39 ++
    9-40 ++
    9-41 ++
    9-42 +
    9-43 +
    9-44 ++
    9-45 ++
    9-46 ++
    9-47 +
    9-48 +
    9-49 +
    9-50 +
    9-51 +
    9-52 +
    9-53 +
    9-54 +
    9-55 +
    9-56 +
    9-57 +
    9-58 +
    9-59 +
    9-60 +
    9-61 +
    9-62 +++
    9-63 +++
    9-64 +++
    9-65 +
    9-66 +
    9-67 +
    9-68 +
    9-69 +
    9-70 +++
    9-71 +++
    9-72 +++
    9-73 +++
    9-74 +
    9-75 +
    10-1  ++
    10-2  +
    10-3  +
    10-4  +
    10-5  +++
    10-6  +++
    10-7  +++
    10-8  +++
    10-9  +++
    10-10  +++
    10-11  +++
    10-12  +++
    10-13  +++
    10-14  +++
    10-15  +
    10-16  +
    10-17  +
    10-18  +++
    10-19  +++
    10-20  +++
    10-21  +++
    10-22  +++
    10-23  +++
    10-24  ++
    10-25  ++
    10-26  +
    10-27  +
    10-28  +
    10-29  ++
    10-30  ++
    10-31  ++
    10-32  ++
    10-33  ++
    10-34  ++
    10-35  ++
    10-36  ++
    10-37  ++
    10-38  ++
    10-39  ++
    10-40  ++
    10-41  ++
    10-42  +++
    10-43  +++
    10-44  +++
    10-45  ++
    10-46  ++
    10-47  ++
    10-48  +
    10-49  +
    10-50  +
    10-51  +
    10-52  +
    10-53  +
    10-54  +++
    10-55  ++
    10-56  +++
    10-57  +++
    10-58  +++
    10-59  +
    10-60  +
    10-61  +
    10-62  +
    10-63  +
    10-64  +
    10-65  +
    10-66  +
    10-67  +
    10-68  +
    10-69  +
    10-70  +
    10-71  +
    10-72  +
    10-73  +
    10-74  +
    10-75  +
    10-76  ++
    10-77  ++
    10-78  ++
    10-79  +++
    10-80  +++
    10-81  +++
    11-1  ++
    11-2  ++
    11-3  ++
    11-4  +
    11-5  +
    11-6  ++
    11-7  ++
    11-8  +
    11-9  +
    11-10  +
    11-11  +
    11-12  +
    11-13  +
    11-14  +
    11-15  +
    11-16  +
    11-17  +
    11-18  +
    11-19  +
    11-20  +
    11-21  +
    11-22  +
    11-23  +
    11-24  +
    11-25  +
    11-26  +
    11-27  +
    11-28  +
    11-29  +
    11-30  +
    12-1  ++
    12-2  +
    12-3  +
    12-4  ++
    12-5  +
    12-6  +
    12-7  +
    12-8  +
    12-9  +
    12-10  +
    12-11  ++
    12-12  ++
    17-1  +++
    17-2  +
    17-3  +++
    17-4  ++
    17-5  +++
    17-6  +++
    17-7  +
    17-8  ++
    18-1  +++
    18-2  +
    18-3  +
    18-4  +++
    18-5  +++
    18-6  ++
    18-7  ++
    18-8  ++
    18-9  ++
    18-10  ++
    18-11  ++
    18-12  ++
    18-13  ++
    18-14  ++
    18-15  +
    18-16  +++
    18-17  +++
    18-18  +
    18-19  +
    18-20  ++
    19-1  +
    19-2  ++
    19-3  +
    19-4  +
    19-5  +
    19-6  +
    19-7  +
    19-8  +
    20-1  ++
    20-2  +++
    20-3  +++
    21-1  ++
    21-2  +++
    21-3  +
    21-4  ++
    21-5  +
    21-6  +
    21-7  +++
    21-8  +
    21-9  +
    21-10  +
    21-11  +
    21-12  +
    21-13  ++
    21-14  +++
    21-15  +++
    21-16  +++
    21-17  +++
    22-1  ++
    22-2  +++
    22-3  +++
    22-4  +++
    22-5  +++
    22-6  ++
    22-7  +++
    22-8  +++
    22-9  +++
    22-10  +++
    22-11  ++
    22-12  +++
    22-13  +++
    22-14  +++
    22-15  +++
    22-16  ++
    22-17  +++
    22-18  ++
    22-19  ++
    22-20  NT
    22-21  NT
    22-22  +++
    22-23  +++
    22-24  +++
    22-25  +++
    22-26  ++
    22-27  ++
    22-28  ++
    22-29  ++
    22-30  +++
    22-31  +++
    22-32  +++
    23-1  +
    23-2  ++
    23-3  ++
    26-1  +++
    26-2  +++
    26-3  +
    26-4  +++
    26-5  +++
    26-6  +++
    27-1  ++
    27-2  +
    27-3  +
    27-4  +
    27-5  +
    27-6  +
    27-7  +
    27-8  +
    27-9  +
    27-10  +
    27-11  ++
    27-12  ++
    27-13  +
    27-14  +
    27-15  +
    27-16  +
    27-17  +
    27-18  +
    27-19  +
  • For the compounds of Table 20 above with Ki values against the M4 receptor of less than 10 nM (i.e., the “+++” compounds), their selectivity over the M1, M2, M3 and M5 receptors are set forth in Table 21 below. In Table 21, activity is expressed as follows: (1) “+++” means the compound had a Ki against the noted receptor of less than 10 nM; (2) “++” means the compound had a Ki against the noted receptor of less than 100 nM but greater than or equal to 10 nM; (3) “+” means the compound had a Ki against the noted receptor of less than 1 μM (1,000 nM) but greater or equal to 100 nM; and (4) “−” means the compound had a Ki against the noted receptor of 1 μM (1,000 nM) or greater or that activity was not detected against the noted receptor. (“NT” in Table 21 means that the compound was not tested against the muscarinic receptor noted.)
  • TABLE 21
    Cpd No M1 M2 M3 M5
    3-1  + ++
    3-5  +
    3-8  + ++ +
    3-9  + ++ +
    3-10 + ++ +
    3-11 +
    3-12 + ++
    3-13 + +
    3-14 + ++
    3-16
    3-17 +
    3-18
    3-19 +
    3-21 +
    3-22 +
    3-23 +
    4-1  + +
    4-5  + ++
    4-6  + ++ NT
    4-10 + ++ + +
    4-11 + ++
    4-12 + ++ +
    4-13 +
    4-14 + NT
    4-15 + +
    4-16 + + NT
    4-17 +
    4-18 +
    4-19 + +
    4-20 + ++
    4-21 + ++ +
    4-22 + +
    4-52 + ++ +
    4-53 + ++ + +
    4-54 + +
    4-60 + ++ + +
    4-68 + +
    4-71 + +
    4-72 + + NT
    4-73 + ++ +
    4-76 +
    4-77 + ++
    4-79 + ++
    4-80 + +
    5-1  +
    5-5  + +
    5-8  ++ +
    5-14 + +
    5-17 + + +
    5-18 ++
    5-20 +
    5-22 + ++
    5-23 + ++
    5-24 + ++
    5-27 + +
    5-28 + +
    5-29 +
    5-30 +
    5-31 ++ +++ ++ +
    5-32 ++
    6-1  + +++ + +
    6-3  + ++ +
    6-4  + +
    6-26 + ++
    6-27 + ++ +
    6-35 + ++ +
    6-36 + +
    6-37 + ++
    6-40 + ++ +
    6-42 ++ +
    6-46 +++
    6-47 + +++ + +
    6-48 ++
    6-50 + ++ +
    7-1 
    7-9  + +
    7-10 +
    7-11
    7-12
    8-1  + + + +
    8-48 ++ +++ + +
    9-1  +
    9-5 
    9-6  + ++ + +
    9-7  + +
    9-8  +
    9-9  + +
    9-10 + + +
    9-62 + +
    9-63 ++ +++ ++ +
    9-64 + +
    9-70 + +
    9-71 +
    9-72 + +
    9-73 +
    10-5  + ++
    10-6  + +
    10-7  +
    10-8  +
    10-9  +
    10-10  + +
    10-11  + +
    10-12  + ++ + +
    10-13  + +
    10-14  +
    10-18  +
    10-19  + +
    10-20  +
    10-21  +
    10-22  +
    10-23  + ++
    10-42  + +
    10-43  +
    10-44  +
    10-54  + ++
    10-56  + ++ +
    10-57  + ++
    10-58 
    10-79  + ++ +
    10-80  ++ ++ + +
    10-81  ++ ++ ++ +
    17-1  +
    17-3  + +
    17-5  + ++
    17-6  +
    18-1  ++
    18-4  + +
    18-5  +
    18-16  +
    18-17  +
    20-2  ++ ++ ++ +
    20-3  + + +
    21-2  +
    21-7  ++ +++ ++ +
    21-14  + ++ +
    21-15  +
    21-16  ++ +++ ++ +
    21-17  ++ +++ ++ +
    22-2  +
    22-3  +
    22-4  ++
    22-5  + ++ +
    22-7  + ++ + +
    22-8  + ++
    22-9  + ++
    22-10  +
    22-12  ++ +
    22-13  + ++ + +
    22-14  + ++ + +
    22-15  + ++ +
    22-17  + ++ +
    22-22  + ++
    22-23  + +++ + +
    22-24  + ++
    22-25  ++
    22-30  + ++
    22-31  ++ ++ +
    22-32  + + +
    26-1  + ++
    26-2  +
    26-4  +
    26-6  ++ ++ + +
  • Functional Assay
  • Functional antagonism of acetylcholine responses were evaluated using a 35S-GTPγS binding assay. Acetylcholine binding to the muscarinic receptors activates G-proteins. Activation of G-proteins can be determined by their binding of the radiolabeled GTP analogue 35S-GTPγS. In the assay, acetylcholine stimulates the binding of 35S-GTPγS to G-proteins associated with cell membranes, and the incorporated 35S-GTPγS can be collected by harvesting the membranes. Antagonist activity of the compounds was determined as the IC50 for inhibition of the acetylcholine response. The assay buffer used was 50 mM HEPES, 100 mM NaCl, 5 mM MgCl2, 1 mM ethylenediaminetetraacetic acid, pH-adjusted to pH 7.4. Acetylcholine, compound (a dose-response of twelve concentrations ranging from 10 μM to 32 pM) and membranes from CHO cells expressing M4 or M2 receptors were incubated together in 150 μl buffer for 30 minutes at 30° C. in a 96-well plate. 35S-GTPγS was then added, to a final concentration of 0.2 nM and a final volume of 175 μl. Twenty minutes later, membranes were harvested by rapid vacuum filtration onto non-treated glass fiber filters, as previously described (see, e.g., Hoare et al., Mol. Pharmacol. 2003 63(3):751-65); Erratum at Mol. Pharmacol. 2005 July; 68(1): 260). The concentration of acetylcholine used was that which stimulated 80% of the maximal response (3 μM for the M4 receptor, 1 μM for M2). Many of the compounds described above have been evaluated in the functional assay.
  • Electrophysiology Assay
  • Adult (>8 weeks) female Lister hooded rats (Harlan, UK) were killed by decapitation and the brain was removed and placed into ice-cold oxygenated sucrose Krebs' medium containing (mM): sucrose 202, KCl 2, KH2PO4 1.25, MgSO4 10, CaCl2 0.5, NaHCO3 26, glucose 10. The brain was hemisected along the midline and 300 μM parasagittal slices were prepared with an oscillating microtome (Integraslice; Campden Instruments Ltd., Loughborough, UK). Slices were then transferred to a recovery chamber at room temperature containing oxygenated Krebs' solution (mM): NaCl 124, KCl 2, KH2PO4 1.25, MgSO4 1, CaCl2 2, NaHCO3 26, glucose 10. Following at least 1 hour of recovery, individual slices were transferred to an interface recording chamber where they were perfused with Krebs' solution (33° C.). Extracellular field potential recordings were made with an Axoprobe 1A amplifier (Axon Instruments Ltd., USA) via a Krebs'-filled glass micropipette (resistance 2-5 M12) positioned in the stratum radiatum of the CAI, digitized (10 kHz) via a CED1401 interface and stored on a computer with Spike2 software (Cambridge Electronic Design Ltd., Cambridge, UK). Field excitatory postsynaptic potential (fEPSP) responses were evoked (pair of 0.02 ms pulses, separated by 40 ms; applied every 10 s; adjusted to approximately 60% of the maximal spike-free response) by a bipolar stimulating electrode positioned in the stratum radiatum near the CA3-CA1 border.
  • The cholinergic agonist carbachol (aza-acetylcholine, resistant to degradation by acetylcholinesterase) was used to stimulate muscarinic receptors. The M1 muscarinic receptor was blocked using 5 μM VU0255035, a selective M1 antagonist. The resulting inhibitory signal was primarily M4-mediated, based on its sensitivity to the M4 activator VU010010. The effect of M4 antagonists on this M4-mediated inhibition of fEPSPs was measured by adding M4 compound 20 minutes prior to application of carbachol.
  • 6-OHDA Surgical Lesion and Behavioral Testing Procedures
  • 6-OHDA Lesion protocol: Male Sprague-Dawley rats were anesthetized with isoflurane and placed into the stereotaxic frame. Thirty minutes prior the injection of 6-OHDA, rats received desipramine (15 mg/kg, i.p.) to prevent the entry of the toxin into the noradrenergic cells. A unilateral lesion was induced by injections of 6-OHDA (8 μg/4 μl/site/rat; flow rate 1 μl/min; dissolved in 0.9% NaCl with 0.02% ascorbic acid) or vehicle into the left and right medial forebrain bundle at the following coordinates: AP −4.4. mm; L ±1.2 mm; V −7.8 mm relative to Bregma (Paxinos and Watson, 2007). The rats were allowed to recover for 14 days and were then tested for locomotor activity induced by novelty (placing the rat in a new cage, 30 min) and for contraversive (contralateral) rotational behavior induced by apomorphine (0.2 mg/kg, s.c.).
  • Experimental animal selection criteria: Only the rats with activity higher than 5 turns/min following apomorphine treatment were enrolled in the study; rats not fulfilling the criteria were excluded from the study (typically 20%). Turning activity was then recorded for each group once per week for four consecutive weeks.
  • Locomotor Activity
  • Young adult male, Sprague-Dawley rats (240-250 g) were purchased from Charles River Laboratories and assessed in the Open Field (Kinder Scientific, CA) task during the light hours of a 12:12 L:D cycle and were tested under bright light conditions. Animals were allowed to acclimate to the facility for at least one week prior to use. On the day of testing, animals were acclimated to the test room for at least 1 hour and were then treated orally with the Neurocrine compound and placed into the test chamber 30 minutes later. Animals were allowed to freely ambulate for 60 minutes. Measurements taken included, but were not limited to, total horizontal and vertical beam breaks.
  • The various embodiments described above can be combined to provide further embodiments. All U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications, and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments.
  • The disclosures of U.S. provisional patent application Ser. No. 62/252,179, filed Nov. 6, 2015, and U.S. provisional patent application Ser. No. 62/275,708, filed Jun. 1, 2016, are incorporated herein by reference in their entirety.
  • These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.

Claims (3)

1-61. (canceled)
62. A pharmaceutical composition comprising a compound selected from:
(2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide;
(2S,6R)—N-(2-(1-benzylpiperidin-4-yl)ethyl)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide;
(2R,6R)—N-(2-(1-benzylpiperidin-4-yl)ethyl)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide;
(2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide;
(2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide; and
(2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-5-fluorophenyl)-2-methylpiperazine-1-carboxamide;
or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
63. A method for treating a neurological disease or disorder in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound selected from:
(2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide;
(2S,6R)—N-(2-(1-benzylpiperidin-4-yl)ethyl)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide;
(2R,6R)—N-(2-(1-benzylpiperidin-4-yl)ethyl)-4-(5-cyanopyrimidin-2-yl)-2,6-dimethylpiperazine-1-carboxamide;
(2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2-methyl-4-[5-(trifluoromethyl)pyrimidin-2-yl]piperazine-1-carboxamide;
(2R,6R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-2,6-dimethyl-4-[5-(trifluoromethyl)pyrazin-2-yl]piperazine-1-carboxamide; and
(2R)—N-[2-(1-benzylpiperidin-4-yl)ethyl]-4-(3-cyano-5-fluorophenyl)-2-methylpiperazine-1-carboxamide;
or a pharmaceutically acceptable salt thereof, wherein the neurological disease or disorder is selected from the group consisting of Alzheimer's Disease, Lewy body dementia, a cognitive deficit associated with schizophrenia, Parkinson's disease, drug induced Parkinsonism, dyskinesia, dystonia, chorea, cerebral palsy, progressive supranuclear palsy, and Huntington's disease.
US17/239,726 2015-11-06 2021-04-26 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases Abandoned US20210338653A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/239,726 US20210338653A1 (en) 2015-11-06 2021-04-26 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201562252179P 2015-11-06 2015-11-06
US201662275708P 2016-01-06 2016-01-06
PCT/US2016/060659 WO2017079641A1 (en) 2015-11-06 2016-11-04 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases
US201815773915A 2018-05-04 2018-05-04
US17/239,726 US20210338653A1 (en) 2015-11-06 2021-04-26 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/773,915 Continuation US11033539B2 (en) 2015-11-06 2016-11-04 Compounds of formulas (VII), (VIII), (IX), (XI), (XII), (XIII), and (XIV) as muscarinic receptor 4(M4) antagonists for treating neurological diseases
PCT/US2016/060659 Continuation WO2017079641A1 (en) 2015-11-06 2016-11-04 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases

Publications (1)

Publication Number Publication Date
US20210338653A1 true US20210338653A1 (en) 2021-11-04

Family

ID=57349137

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/773,915 Active 2037-07-22 US11033539B2 (en) 2015-11-06 2016-11-04 Compounds of formulas (VII), (VIII), (IX), (XI), (XII), (XIII), and (XIV) as muscarinic receptor 4(M4) antagonists for treating neurological diseases
US17/239,726 Abandoned US20210338653A1 (en) 2015-11-06 2021-04-26 N-[2-(1 -benzylpiperidin-4-yl)ethyl]-4-(pyrazin-2-yl)-piperazine-1 -carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/773,915 Active 2037-07-22 US11033539B2 (en) 2015-11-06 2016-11-04 Compounds of formulas (VII), (VIII), (IX), (XI), (XII), (XIII), and (XIV) as muscarinic receptor 4(M4) antagonists for treating neurological diseases

Country Status (22)

Country Link
US (2) US11033539B2 (en)
EP (1) EP3371164B1 (en)
JP (2) JP6917989B2 (en)
KR (1) KR20180073685A (en)
CN (1) CN108349936B (en)
AU (1) AU2016348524B2 (en)
BR (1) BR112018008630A2 (en)
CA (1) CA3001873A1 (en)
CL (1) CL2018001217A1 (en)
CO (1) CO2018004800A2 (en)
ES (1) ES2915266T3 (en)
HK (1) HK1253029A1 (en)
IL (1) IL258862B (en)
MA (1) MA43168A (en)
MX (1) MX2018005215A (en)
MY (1) MY194461A (en)
PE (1) PE20181010A1 (en)
PH (1) PH12018500940A1 (en)
SA (1) SA518391518B1 (en)
SG (1) SG11201803757UA (en)
TN (1) TN2018000130A1 (en)
WO (1) WO2017079641A1 (en)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MY194461A (en) * 2015-11-06 2022-11-30 Neurocrine Biosciences Inc N-[2-(1-benzylpiperidin-4-yl)4-(pyrazin-2-yl)-piperazine-1-carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases
PE20181885A1 (en) 2015-11-06 2018-12-07 Hoffmann La Roche DERIVATIVES OF INDOLIN-2-ONA
AU2018313802A1 (en) 2017-08-08 2020-02-20 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
US11149022B2 (en) 2017-10-17 2021-10-19 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CA3079617A1 (en) 2017-10-20 2019-04-25 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor m4
AU2018360581A1 (en) 2017-10-31 2020-06-11 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
US11325896B2 (en) 2017-12-20 2022-05-10 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CA3090130A1 (en) * 2018-02-02 2019-08-08 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor m4
ES2910071T3 (en) 2018-03-08 2022-05-11 Incyte Corp Aminopyrazine diol compounds as PI3K-Y inhibitors
WO2020010003A1 (en) 2018-07-02 2020-01-09 Incyte Corporation AMINOPYRAZINE DERIVATIVES AS PI3K-γ INHIBITORS
WO2020048827A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 1, 3, 9-triazaspiro[5.5] undecan-2-one compounds
WO2020048828A1 (en) 2018-09-03 2020-03-12 Bayer Pharma Aktiengesellschaft 5-heteroaryl-3,9-diazaspiro[5.5]undecane compounds
WO2020048826A1 (en) 2018-09-03 2020-03-12 Bayer Aktiengesellschaft 5-substituted 1-oxa-3,9-diazaspiro[5.5]undecan-2-one compounds
US20210155629A1 (en) * 2018-09-04 2021-05-27 Pipeline Therapeutics, Inc. Muscarinic acetylcholine m1 receptor antagonists
EP4031540A1 (en) * 2019-09-17 2022-07-27 Bial-R&D Investments, S.A. Substituted, saturated and unsaturated n-heterocyclic carboxamides and related compounds for their use in the treatment of medical disorders
WO2021113478A1 (en) 2019-12-06 2021-06-10 Neurocrine Biosciences, Inc. Muscarinic receptor 4 antagonists and methods of use
CN111072551A (en) * 2019-12-30 2020-04-28 上海睿瓦科技有限公司 Method for preparing piperidine amine by catalytic hydrogenation one-step method
AR121260A1 (en) * 2020-02-05 2022-05-04 Neurocrine Biosciences Inc MUSCARINIC RECEPTOR 4 ANTAGONISTS AND METHODS OF USE
WO2022156708A1 (en) * 2021-01-20 2022-07-28 Jacobio Pharmaceuticals Co., Ltd. Parp7 enzyme inhibitor
KR20240019340A (en) * 2021-06-11 2024-02-14 뉴로내슨트, 아이엔씨. Methods and compositions for lipid preparations of lipophilic small molecule therapy of heterocyclic type
TW202321222A (en) * 2021-07-30 2023-06-01 美商紐羅克里生物科學有限公司 Muscarinic receptor 4 antagonists and methods of use

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5700801A (en) 1994-12-23 1997-12-23 Karl Thomae, Gmbh Piperazine derivatives, pharmaceutical compositions containing these compounds, their use and processes for preparing them
CN100418951C (en) * 1997-11-18 2008-09-17 帝人医药株式会社 Cycloamine derivative and its use as medicine
WO2000069432A1 (en) * 1999-05-18 2000-11-23 Teijin Limited Remedies or preventives for diseases in association with chemokines
AU5786300A (en) * 1999-09-22 2001-04-24 Schering Corporation Muscarinic antagonists
AU778173B2 (en) * 1999-12-08 2004-11-18 Teijin Limited Cycloamine CCR5 receptor antagonists
US6410566B1 (en) * 2000-05-16 2002-06-25 Teijin Limited Cyclic amine derivatives and their use as drugs
JPWO2003062234A1 (en) 2002-01-23 2005-05-19 山之内製薬株式会社 Quinoxaline compounds
US8673924B2 (en) 2002-09-04 2014-03-18 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as cyclin dependent kinase inhibitors
CA2509711A1 (en) 2002-12-13 2004-07-01 Smithkline Beecham Corporation Piperidine derivatives as ccr5 antagonists
TW200536830A (en) 2004-02-06 2005-11-16 Chugai Pharmaceutical Co Ltd 1-(2H)-isoquinolone derivative
WO2007075629A2 (en) * 2005-12-21 2007-07-05 Schering Corporation Phenoxypiperidines and analogs thereof useful as histamine h3 antagonists
WO2007130383A2 (en) * 2006-04-28 2007-11-15 Northwestern University Compositions and treatments using pyridazine compounds and secretases
EP1997805A1 (en) * 2007-06-01 2008-12-03 Commissariat à l'Energie Atomique Compounds with antiparasitic activity, applications thereof to the treatment of infectious diseases caused by apicomplexans
JP2013028538A (en) * 2009-11-13 2013-02-07 Dainippon Sumitomo Pharma Co Ltd Novel amide derivative
CN103946214B (en) 2011-11-25 2016-08-17 内尔维阿诺医学科学有限公司 3-phenyl-isoquinolin-1 (2H)-one derivant as PARP-1 inhibitor
US20150259649A1 (en) 2012-11-08 2015-09-17 Emory University Cellular compositions used to restore stem cell or progenitor cell function and methods related thereto
BR112015011094A2 (en) 2012-11-20 2017-07-11 Hoffmann La Roche Substituted 1,6-naphthyridines
SG11201506385WA (en) 2013-03-18 2015-10-29 Genoscience Pharma Quinolines derivatives as novel anticancer agents
US9598398B2 (en) * 2013-04-04 2017-03-21 Takeda Pharmaceutical Company Limited Heterocyclic compound
CA2922469A1 (en) * 2013-09-11 2015-03-19 Institute Of Cancer Research: Royal Cancer Hospital (The) 3-aryl-5-substituted-isoquinolin-1-one compounds and their therapeutic use
BR112016021626A2 (en) * 2014-03-20 2018-05-15 Samumed, Llc 5-substituted indazol-3-carboxamides and their preparation and use
MY194461A (en) * 2015-11-06 2022-11-30 Neurocrine Biosciences Inc N-[2-(1-benzylpiperidin-4-yl)4-(pyrazin-2-yl)-piperazine-1-carboxamide derivatives and related compounds as muscarinic receptor 4 (m4) antagonists for treating neurological diseases

Also Published As

Publication number Publication date
CL2018001217A1 (en) 2018-08-03
KR20180073685A (en) 2018-07-02
TN2018000130A1 (en) 2019-10-04
HK1253029A1 (en) 2019-06-06
JP6917989B2 (en) 2021-08-11
PH12018500940A1 (en) 2018-12-17
JP2021050243A (en) 2021-04-01
CN108349936B (en) 2021-10-01
MA43168A (en) 2018-09-12
EP3371164A1 (en) 2018-09-12
AU2016348524A1 (en) 2018-05-17
CA3001873A1 (en) 2017-05-11
US11033539B2 (en) 2021-06-15
CO2018004800A2 (en) 2018-10-31
AU2016348524B2 (en) 2021-01-28
IL258862A (en) 2018-06-28
JP2018531981A (en) 2018-11-01
SA518391518B1 (en) 2021-06-13
SG11201803757UA (en) 2018-06-28
US20180325887A1 (en) 2018-11-15
MY194461A (en) 2022-11-30
IL258862B (en) 2021-02-28
ES2915266T3 (en) 2022-06-21
BR112018008630A2 (en) 2018-10-30
MX2018005215A (en) 2018-08-01
CN108349936A (en) 2018-07-31
WO2017079641A1 (en) 2017-05-11
PE20181010A1 (en) 2018-06-26
EP3371164B1 (en) 2022-03-16

Similar Documents

Publication Publication Date Title
US11033539B2 (en) Compounds of formulas (VII), (VIII), (IX), (XI), (XII), (XIII), and (XIV) as muscarinic receptor 4(M4) antagonists for treating neurological diseases
US11053242B2 (en) [9,10-dimethoxy-3-(2-methylpropyl)-1H,2H,3H,4H,6H,7H,11BH-pyrido-[2,1-a]isoquinolin-2-yl]methanol and compounds, compositions and methods relating thereto
US8969565B2 (en) Imidazo[1,2-b]pyridazine-based compounds, compositions comprising them, and methods of their use
AU2010207507B2 (en) Amino-heterocyclic compounds used as PDE9 inhibitors
US8993756B2 (en) Pyrrolopyrimidines as janus kinase inhibitors
US8309578B2 (en) Bicyclic pyrazole and isoxazole derivatives as antitumor and antineurodegenerative agents
US11247965B2 (en) Hepatitis B capsid assembly modulators
US11142521B2 (en) Substituted anilines as CCR(4) antagonists
NL2000380C2 (en) 3-aminocyclopentane carboxamides as modulators of chemokine receptors.
US11390623B2 (en) 6,7,8,9-tetrahydro-5H-pyrido[2,3-d]azepine dopamine D3 ligands
EA039638B1 (en) Muscarinic receptor 4 antagonists and methods of using same
WO2023064218A1 (en) Tyro3 inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: NEUROCRINE BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARRIOTT, NICOLE;PAGANO, NICHOLAS;REEL/FRAME:056570/0655

Effective date: 20161026

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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