US20080108618A1 - Compounds with activity on muscarinic receptors - Google Patents

Compounds with activity on muscarinic receptors Download PDF

Info

Publication number
US20080108618A1
US20080108618A1 US11/933,177 US93317707A US2008108618A1 US 20080108618 A1 US20080108618 A1 US 20080108618A1 US 93317707 A US93317707 A US 93317707A US 2008108618 A1 US2008108618 A1 US 2008108618A1
Authority
US
United States
Prior art keywords
alkyl
compound
alkoxy
butyl
composition
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
US11/933,177
Inventor
Mark Brann
Terri Messier
Erika Currier
Kate Duggento
Tracy Spalding
Mikael Friberg
Niels Skjaerbaek
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.)
Individual
Original Assignee
Individual
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
Priority claimed from PCT/US1999/007057 external-priority patent/WO1999050247A1/en
Application filed by Individual filed Critical Individual
Priority to US11/933,177 priority Critical patent/US20080108618A1/en
Publication of US20080108618A1 publication Critical patent/US20080108618A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/10Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms
    • C07D295/104Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/108Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • 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
    • C07D211/20Heterocyclic 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 singly bound oxygen or sulphur 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
    • C07D211/08Heterocyclic 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
    • C07D211/20Heterocyclic 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 singly bound oxygen or sulphur atoms
    • C07D211/22Heterocyclic 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 singly bound oxygen or sulphur atoms by oxygen 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
    • C07D211/08Heterocyclic 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
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/44Radicals substituted by doubly-bound oxygen, sulfur, or nitrogen atoms, or by two such atoms singly-bound to the same carbon atom
    • C07D213/46Oxygen atoms
    • C07D213/50Ketonic radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings
    • C07D265/321,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings with oxygen atoms directly attached to ring carbon atoms

Definitions

  • the present invention relates to novel compounds which are selective for muscarinic acetylcholine receptor subtypes, as well as to methods for activating muscarinic receptors and for treating or alleviating diseases in which modification of muscarinic receptor activity is beneficial.
  • Muscarinic acetylcholine receptors play a central role in the central nervous system for higher cognitive functions, as well as in the peripheral parasympathetic nervous system. Cloning has established the presence of five distinct muscarinic receptor subtypes (termed m1-m5) (cf. T. I. Bonner et al, Science 237, 1987, pp. 527-532; T. I. Bonner et al., Neuron 1, 1988, pp. 403-410).
  • m1 is the predominant subtype in the cerebral cortex and is believed to be involved in the control of cognitive functions
  • m2 is predominant in heart and is believed to be involved in the control of heart rate
  • m3 is believed to be involved in gastrointestinal and urinary tract stimulation as well as sweating and salivation
  • m4 is present in brain
  • m5 is present in brain and may be involved certain functions of the central nervous system associated with the dopaminergic system.
  • muscarinic compounds have a profound effect on cognitive functions, e.g. learning and memory. This would suggest a potential utility of muscarinic agonists in the improvement of cognitive functions in diseases characterized by cognitive impairment, both age-related (such as Alzheimer's disease or other dementias) and not age-related (such as attention deficit hyperactivity disorder).
  • m1 receptor subtype is the more abundant one in the cerebral cortex, basal ganglia and hippocampus where it accounts for 35-60% of all muscarinic receptor binding sites (cf. A. Levey, Proc. Natl. Acad. Sci. USA 93, 1996, pp. 13541-13546).
  • the m1 (and possibly m4) subtype plays a major role as a postsynaptic muscarinic receptor (located on cholinoceptive neurons in the neocortex and hippocampus) in various cognitive and motor functions and is likely to be a major contributor to the m1 responses measured in these regions of the brain.
  • acetylcholine esterase (ACHE) inhibitors such as 9-amino-1,2,3,4-tetrahydroacridine (tacrine) results in an increase of acetylcholine in the brain which indirectly causes stimulation of muscarinic receptors.
  • Tacrine treatment has resulted in a moderate and temporary cognitive improvement in Alzheimer's patients (cf. Kasa et al., supra).
  • tacrine has been found to have cholinergic side effects due to the peripheral acetylcholine stimulation. These include abdominal cramps, nausea, vomiting, diarrhea, anorexia, weight loss, myopathy and depression. Gastrointestinal side effects have been observed in about a third of the patients treated.
  • Tacrine has also been found to cause significant hepatotoxicity, elevated liver transaminases having been observed in about 30% of the patients (cf. P. Taylor, “Anticholinergic Agents”, Chapter 8 in Goodman and Gilman: The Pharmacological Basis of Therapeutics, 9 th Ed., 1996, pp. 161-176).
  • the adverse effects of tacrine have severely limited its clinical utility.
  • Another AChE inhibitor, (R,S)-1-benzyl-4-[5,6-dimethoxy-1-indanon-2-yl]methylpiperidine.HCl (donepezil) has recently been approved for the treatment of symptoms of mild to moderate Alzheimer's disease (cf. P. Kasa et al, supra). No hepatic damage has been observed for this compound but it has gastrointestinal effects similar to those of tacrine, probably due to stimulation of the m3 receptor caused by elevated parasympathetic tone.
  • muscarinic agonists (believed to be m1 selective) hitherto suggested for the treatment of Alzheimer's disease, such as arecoline, have not shown greater efficacy in clinical trials than ACHE inhibitors (cf. S. V. P. Jones et al., supra).
  • arecoline was found to have not so much cognitive enhancing effects as effects on behavioral changes often observed in Alzheimer's disease patients, such as a significant increase in motor activity, significant uplifting of mood, and significant decrease in anergia.
  • m1 agonists have later been found to be weak partial agonists selective for the m2 and/or m3 receptor subtypes (H. B syndromener-Osborne et al., J. Med. Chem. 38, 1995, pp. 2188-2195).
  • m2 subtype selectivity is presumed to be responsible for the cardiovascular effects observed for these agonists, e.g. tachycardia and bradycardia, and m3 activity is believed to account for the adverse gastrointestinal effects of the agonists.
  • m2 and/or m3 activity is therefore a significant drawback for the muscarinic agonists proposed until now for the treatment of Alzheimer's disease, severely limiting the doses of the drugs which it has been possible to administer to patients who may therefore have received sub-optimal doses.
  • the lack of subtype selectivity and low potency of the currently tested cholinergic compounds appear to favor the negative peripheral side effects and have limited cognitive effects because of weak and/or opposing actions in the brain. It would therefore be of great advantage to develop compounds which have an improved selectivity for the m1 subtype, but which have little or no activity on the m2 and m3 subtypes.
  • the present invention provides compounds with muscarinic agonist activity of the general formula (I):
  • X 1 , X 2 , X 3 , X 4 and X 5 are selected from C, N and O;
  • k 0 or 1
  • t 0, 1 or 2;
  • R 1 is straight or branched-chain C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkylidene, C 1-8 alkoxy, C 1-8 heteroalkyl, C 1-8 aminoalkyl, C 1-8 haloalkyl, C 1-8 alkoxycarbonyl, C 1-8 hydroxyalkoxy, C 1-8 hydroxyalkyl, —SH, C 1-8 alkylthio, —O—CH 2 —C 5-6 aryl, —C(O)—C 5-6 aryl substituted with C 1-3 alkyl or halo; C 5-6 aryl or C 5-6 cycloalkyl optionally comprising 1 or more heteroatoms selected from N, S and O; —C(O)NR 3 R 4 , —NR 3 R 4 , —NR 3 C(O)NR 4 R 5 , —CR 3 R 4 , —OC(O)R 3 , —(O)(CH 2
  • A is C 5-12 aryl or C 5-7 cycloalkyl, each optionally comprising 1 or more heteroatoms selected from N, S and O;
  • R 2 is H, amino, hydroxyl, halo, or straight or branched-chain C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 heteroalkyl, C 1-6 aminoalkyl, C 1-6 haloalkyl, C 1-6 alkylthio, C 1-6 alkoxycarbonyl, —CN, —CF 3 , —OR 3 , —COR 3 , NO 2 , —NHR 3 , —NHC(O)R 3 , —C(O)NR 3 R 4 , —NR 3 R 4 , —NR 3 C(O)NR 4 R 5 , —OC(O)R 3 , —C(O)R 3 R 4 , —O(CH 2 ) q NR 3 , —CNR 3 R 4 or —(CH 2 ) q NR 3 R 4 ; where q is an integer from 1 to 6;
  • n 0, 1, 2, 3 or 4, the groups R 2 , when n>1, being the same or different;
  • p is 0 or an integer from 1 to 5;
  • Y is O, S, CHOH, —NHC(O)—, —C(O)NH—, —C(O)—, —OC(O)—, NR 7 or —CH ⁇ N—, and R 7 is H or C 1-4 alkyl; or absent; and
  • Z is CR 8 R 9 wherein R 8 and R 9 are independently selected from H, and straight or branched chain C 1-8 alkyl; or
  • the present invention further provides pharmaceutical compositions comprising an effective amount of a compound of formula (I).
  • compositions comprising a compound of formula (I).
  • the present invention provides a method of treating the symptoms of a disease or condition associated with increased intraocular pressure, such as, for example, glaucoma, where the method comprises administering a therapeutically effective amount of a composition comprising a muscarinic receptor agonist, preferably an m1 selective agonist, preferably selected from the compounds of formula (I).
  • FIG. 1 is a graph showing raw data from one 96-well microtiter plate of screening of 35,000 small organic molecules in the assay described in Example XVI.
  • FIG. 2 is a graph showing data comparing the profile of the reference antagonist atropine with m1 muscarinic receptor transfected cells stimulated with either carbachol (open triangles) or compound A (Example I) (closed triangles).
  • the present invention provides compounds preferably showing a relatively high selectivity towards the m1 receptor subtype relative to other muscarinic subtypes which may have a beneficial effect in the treatment of cognitive impairment such as Alzheimer's disease or other conditions associated with age-related cognitive decline while avoiding the adverse affects of the drugs hitherto suggested for this purpose.
  • Compounds exhibiting this property have surprisingly been isolated by screening against m1-m5 receptor subtypes.
  • the present invention provides compounds of formula (I), wherein
  • X 1 , X 2 , X 3 , X 4 and X 5 are C; or one of X 1 , X 2 , X 3 , X 4 or X 5 is O or N and the others are C;
  • k 0 or 1
  • t 1;
  • R 1 is straight or branched-chain C 1-8 alkyl, C 2-8 alkenyl, C 2-8 alkynyl, C 1-8 alkylidene, C 1-8 alkoxy, C 1-8 aminoalkyl, C 1-8 haloalkyl, C 1-8 alkoxycarbonyl, —C(O)NR 3 R 4 , —NR 3 R 4 , —NR 3 C(O)NR 4 R 5 , —OC(O)R 3 , or —(CH 2 ) s NR 3 R 4 ; where R 3 , R 4 and R 5 are the same or different, each independently being selected from H and C 1-6 alkyl; and skis an integer from 1 to 8;
  • n 1, 2 or 3;
  • A is phenyl or naphthyl
  • R 2 is straight or branched-chain C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 aminoalkyl, C 1-6 haloalkyl, C 1-6 alkoxycarbonyl, —CN, —CF 3 , —OH, —COR 3 , —NHR 3 , —NHC(O)R 3 , —C(O)NR 3 R 4 , —NR 3 R 4 , —NR 3 C(O)NR 4 R 5 , —OC(O)R 3 , or —(CH 2 ) q NR 3 R 4 ; where q is an integer from 1 to 6; or
  • A is aryl comprising 1 or more heteroatoms selected from N, S and O;
  • R 2 is H, halo, straight or branched-chain C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 heteroalkyl, C 1-6 aminoalkyl, C 1-6 haloalkyl, C 1-6 alkoxycarbonyl, —CN, —CF 3 , OH, —COR 3 , —NHR 3 , —NHC(O)R 3 , —C(O)NR 3 R 4 , —NR 3 R 4 , —NR 3 C(O)NR 4 R 5 , —OC(O)R 3 or —(CH 2 ) q NR 3 R 4 ; or
  • the compound is of the formula (II):
  • Preferred subgeneric embodiments of compounds of formula (II) include compounds of formulas (IIa) and (IIb):
  • t is 1 and Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—.
  • X 3 is C.
  • R 1 is alkyl, where preferably R 2 is alkyl, aminoalkyl, alkoxy or hydroxyl.
  • p is 3.
  • R 1 is C 2-8 alkyl and R 2 is methyl, hydroxyl or alkoxy.
  • n is 1 or 2; Y is —C(O)— or O and t is 1.
  • R 2 is halo.
  • X 3 may also be N, where according to one embodiment, R 1 is alkyl or alkoxy; or R 1 is benzyl or phenyl; where R 2 is alkyl or alkoxy.
  • X 3 is O, where t may be, for example, 0.
  • R 2 is alkyl or alkoxy; or R 2 is halo.
  • Particular embodiments of the invention include:
  • the present invention further provides a method of agonizing a muscarinic receptor comprising contacting the receptor with an effective amount of a compound of formula (I), inclusive of all compounds within the scope of formula (I) (i.e., including 4-n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine; 2-[3-(3-n-Butylpiperidine)propanesulfanyl]toluene; and 4-Propyloxy-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine).
  • a compound of formula (I) inclusive of all compounds within the scope of formula (I) (i.e., including 4-n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[
  • compositions comprising an effective amount of a compound of formula (I), inclusive of all compounds within the scope of formula (I)(i.e., including -n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine; 2-[3-(3-n-Butylpiperidine)propane-sulfanyl]toluene; and 4-Propyloxy-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine).
  • the present invention further also provides methods of treating the symptoms of a disease or condition associated with reduced levels of acetylcholine, the method comprising administering a therapeutically effective amount of a composition described herein.
  • diseases or conditions include neurogenerative disease, cognitive impairment, age-related cognitive decline or dementia.
  • the present invention further provides a method of treating the symptoms of a disease or condition associated with increased intraocular pressure, the method comprising administering a therapeutically effective amount of a muscarinic receptor agonist.
  • Glaucoma is a disease in which an abnormality is observed in the circulation-control mechanism of the aqueous humor filling up the anterior chamber, i.e., the space formed between the cornea and the lens. This leads to an increase in the volume of the aqueous humor and an increase in intraocular pressure, consequently leading to the visual field defect and even to loss of eyesight due to the compulsion and contraction of the papillae of the optic nerve.
  • the method comprises administering a therapeutically effective amount of a muscarinic receptor agonist comprising m1 receptor agonist activity, and more preferably m1 selective agonist activity.
  • a muscarinic receptor agonist comprising m1 receptor agonist activity, and more preferably m1 selective agonist activity.
  • any known m1 receptor selective agonist may be used in practicing the invention, preferably the compounds of the present invention as described herein are used.
  • agonists available in the art include xanomeline (3-[4-(hexyloxy)-1,2,5-thiadiazol-3-yl]-1,2,5,6-tetrahydro-1-methylpyridine) (Novo Nordisk); talsaclidine (3-(2-propynyloxy)-1-azabicyclo[2.2.2]octane)(Boehringer Ingelheim); Z-1-azabicyclo[2.2.1]heptane-3-one, O-[3-(3-methoxyphenyl)-2-propynyl]oxime (Parke-Davis PD-151832); sabcomeline (R-(Z)- ⁇ -(methoxyimino)-1-azabicyclo-[2,2,2]octane-3-acetonitrile HCl)(SmithKline Beecham); 2,8-dimethyl-3-methylene-1-oxa-8-azaspiro[4.5]decane (Yamanouchi
  • the compounds of the present invention preferably show selective agonist activity towards the m1 receptor.
  • an agonist is defined as a compound that increases the activity of the m1 muscarinic receptor when it contacts the receptor.
  • selectivity means that an agonist causes a relatively greater increase in the activity of the m1 receptor than of the m3 receptor subtype.
  • the agonist causes at least about a 10 fold greater increase in activity of the m1 receptor than of the m3 receptor.
  • the m1 selective agonist also causes a relatively greater increase, preferably at least about a 10 fold greater increase, in the activity of an m1 receptor than of an m2; preferably than of an m4; and preferably than of an m5.
  • selectivity is defined as a property of a muscarinic m1 agonist whereby an amount of agonist effective to increase the activity of the m1 receptor causes little or no increase in the activity of the m3 and m5 subtypes, and preferably the m2 and m4 subtypes.
  • alkyl means a straight or branched-chain alkane group with 1-6 carbon atoms in the chain, for instance methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, etc.
  • heteroalkyl is intended to indicate an alkane group containing 1 or 2 heteroatoms selected from O, S or N.
  • alkenyl means a straight or branched-chain alkene group with 2-6 carbon atoms in the chain; the term “alkynyl” is intended to indicate a straight or branched-chain alkyne group with 2-6 carbon atoms in the chain.
  • aryl and “cycloalkyl” preferably refer to mono- and bicyclic ring structures comprising 5 to 12 carbon atoms, more preferably monocyclic rings comprising 5 to 6 carbon atoms. Where such rings comprise one or more heteroatoms, selected from N, S and O, (i.e., heterocyclic rings) such rings comprise a total of 5 to 12 atoms, more preferably 5 to 6 atoms.
  • Heterocyclic rings include, but are not limited to, furyl, pyrrolyl, pyrazolyl, thienyl, imidazolyl, isoxazolyl, oxazolyl, thiazolyl, isothiazolyl, pyridyl, piperidinyl, piperazinyl, pyridazinyl, pyrimidinyl, pyrazinyl, morpholinyl, oxadiazolyl, thiadiazolyl, imidazolinyl, imidazolidinyl and the like.
  • the ring may be substituted by one or more of the groups included in the definition of R 2 above.
  • substituents C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, C 1-6 alkoxy, C 1-6 heteroalkyl, C 1-6 aminoalkyl, C 1-6 haloalkyl or C 1-6 alkoxycarbonyl may, if present, be substituted by one or more of hydroxyl, C 1-4 alkoxy, halogen, cyano, amino or nitro.
  • halogen or “halo” includes chlorine, fluorine, iodine and bromine.
  • the starting compound having formula (X) may be prepared by general methods of organic synthesis.
  • general methods of preparing compounds of formula (X) reference is made to Fuller, R. W. et al., J. Med. Chem. 14:322-325 (1971); Foye, W. O., et al., J. Pharm. Sci. 68:591-595 (1979); Bossier, J. R. et al., Chem. Abstr. 66:46195h and 67:21527a (1967); Aldous, F. A. B., J. Med. Chem. 17:1100-1111 (1974); Fuller, R. W. et al., J. Pharm. Pharmacol.
  • radiolabelled derivatives having formula (XX) may be prepared by, for example, using a tritiated reducing agent to form the reductive amination or by utilizing a 14 C-labelled starting material.
  • the compound having the formula (XXII) may be reduced with, for example, AlH 3 , diborane:methyl sulfide or other standard carbonyl reducing reagents to produce the ligand having the formula (XXX).
  • the receptor ligands having formula (XXXII) may be prepared by nucleophilic displacement of an electrophile (E) by the amino derivative (XXXI).
  • electrophiles which may be used for this purpose include halides such as I, Cl, Br, tosylate or mesylate.
  • this compound may be prepared from oxidation of an sec. alcohol with for example pyridinium chlorocromate or N-chlorosuccinimide or CrO 3 —H 2 SO 4 or nickel peroxide or metal (Al, K) or DCC-DMSO.
  • this compound may be prepared by alkylation of an alcohol with arylhalides under for example Cu catalysis.
  • this compound may be prepared by alkylation of a thiol with arylhalides under for example Cu catalysis.
  • this compound may be prepared by reduction of the corresponding ketone by catalytic hydrogenation or by the use of NaBH 4 or by the use of LiAlH 4 .
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g. sodium or potassium salts; alkaline earth metal salts, e.g. calcium or magnesium salts; and salts formed with suitable organic ligands, e.g.
  • salts examples include the acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium, carbonate, chloride, clavulanate, citrate, dihydrochloride, fumarate, gluconate, glutamate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, nitrate, N-methylglucamine ammonium salt, oleate, oxalate, phosphate/diphosphate, salicylate, stearate, sulfate, succinate, tannate, tartrate, tosylate, triethiodide and valerate salt.
  • the present invention includes within its scope prodrugs of the compounds of this invention.
  • prodrugs are inactive derivatives of the compounds of this invention which are readily convertible in vivo into the required compound.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
  • the compounds according to the invention may exist as a racemate or as enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention. Furthermore, some of the crystalline forms for compounds of the present invention may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are also included in the scope of this invention.
  • Such isomers may be separated by conventional techniques such as preparative chiral chromatography.
  • the compounds may be prepared in racemic form, or individual enantiomers may be prepared either by stereoselective synthesis or by resolution.
  • the compounds may, for example, be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as ( ⁇ )-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base.
  • the compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary.
  • any of the processes for preparation of the compounds of the present invention it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry , ed. J. F. W. McOmie, Plenum Press, 1973; and T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis , John Wiley & Sons, 1991.
  • the protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • Compounds of the present invention may be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever specific pharmacological modification of the activity of muscarinic receptors is required.
  • compositions comprising one or more compounds of the invention together with a pharmaceutically acceptable diluent or excipient.
  • compositions are in unit dosage forms such as tablets, pills, capsules (including sustained-release or delayed-release formulations), powders, granules, elixirs, tinctures, syrups and emulsions, sterile parenteral solutions or suspensions, aerosol or liquid sprays, drops, ampoules, auto-injector devices or suppositories; for oral, parenteral (e.g.
  • compositions may be in sustained-release form suitable for once-weekly or once-monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection.
  • suitable topical formulations for administration e.g. eye or skin or mucosa.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents, flavoring agents and coloring agents can also be incorporated into the mixture.
  • suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or beta-lactose, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include, without limitation, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • the active ingredient is mixed with a suitable pharmaceutical excipient, e.g. such as the ones described above, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof.
  • a suitable pharmaceutical excipient e.g. such as the ones described above
  • other pharmaceutical diluents e.g. water
  • the tablets or pills of the present composition may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner core containing the active compound and an outer layer as a coating surrounding the core.
  • the outer coating may be an enteric layer which serves to resist disintegration in the stomach and permits the inner core to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with conventional materials such as shellac, cetyl alcohol and cellulose acetate.
  • the liquid forms in which the present compositions may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical carriers.
  • Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, gelatin, methylcellulose or polyvinyl-pyrrolidone. Other dispersing agents which may be employed include glycerin and the like.
  • sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.
  • the compositions can also be formulated as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration
  • the present invention also relates to a method of alleviating or treating a disease or condition in which modification of muscarinic receptor activity, in particular m1 receptor activity, has a beneficial effect by administering a therapeutically effective amount of a compound of the present invention to a subject in need of such treatment.
  • diseases or conditions may, for instance arise from inadequate stimulation or activation of muscarinic receptors.
  • compounds which are selective for a particular muscarinic receptor subtype, in particular m1 the problems with adverse side effects observed with the known muscarinic drugs, such as tachycardia or bradycardia or gastrointestinal effects, may substantially be avoided.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease being treated.
  • compounds of general formula I exhibit subtype selectivity for the muscarinic m1 receptor subtype.
  • the compounds exhibit selectivity for the muscarinic m1 receptor subtype compared to other human G-protein coupled receptors tested including serotonin, histamine, dopamine or adrenergic receptors.
  • selectivity is that these compounds may be effective in the treatment or amelioration of a number of diseases and disorders of the central nervous system without the undesirable side effects previously observed with non-selective compounds.
  • muscarinic m1 receptor subtype selectivity makes them potentially very useful in treating a number of diseases and disorders characterized by cognitive impairment such as attention deficit disorder, or neurodegenerative diseases, e.g. Alzheimer's disease, other forms of age-related cognitive decline, e.g. senile dementia, or dementia-related symptoms such as decreased motor activity, mood changes, anergia, apathy, restlessness and aggressive behavior. It is currently believed that the muscarinic m1 receptor may also be involved in control of intraocular pressure, and that muscarinic m1 agonists may therefore be used to treat or alleviate ocular diseases such as glaucoma.
  • cognitive impairment such as attention deficit disorder, or neurodegenerative diseases, e.g. Alzheimer's disease, other forms of age-related cognitive decline, e.g. senile dementia, or dementia-related symptoms such as decreased motor activity, mood changes, anergia, apathy, restlessness and aggressive behavior.
  • muscarinic m1 receptor may also
  • compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses two, three or four times daily.
  • compounds for the present invention may be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to persons skilled in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed.
  • a physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the disease or disorder which is being treated.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 100 mg per adult human per day.
  • the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a unit dose typically contains from about 0.001 mg to about 50 mg of the active ingredient, preferably from about 1 mg to about 10 mg of active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0001 mg/kg to about 25 mg/kg of body weight per day.
  • the range is from about 0.001 to 10 mg/kg of body weight per day, and especially from about 0.001 mg/kg to 1 mg/kg of body weight per day.
  • the compounds may be administered on a regimen of 1 to 4 times per day.
  • Compounds according to the present invention may be used alone at appropriate dosages defined by routine testing in order to obtain optimal pharmacological effect on a muscarinic receptor, in particular the muscarinic m1 receptor subtype, while minimizing any potential toxic or otherwise unwanted effects.
  • co-administration or sequential administration of other agents which improve the effect of the compound may, in some cases, be desirable.
  • the pharmacological properties and the selectivity of the compounds of this invention for specific muscarinic receptor subtypes may be demonstrated by a number of different assay methods using recombinant receptor subtypes, preferably of the human receptors if these are available, e.g. conventional second messenger or binding assays.
  • a particularly convenient functional assay system is the receptor selection and amplification assay disclosed in U.S. Pat. No. 5,707,798 describing a method of screening for bioactive compounds by utilizing the ability of cells transfected with receptor DNA, e.g. coding for the different muscarinic subtypes, to amplify in the presence of a ligand of the receptor. Cell amplification is detected as increased levels of a marker also expressed by the cells.
  • reaction mixture was quenched by addition of H 2 SO 4 (10 mL, 2 M) and stirred at rt for 12 hours followed by addition of NaOH (10 mL, 2 M).
  • Addition of THF (15 mL) was followed by extraction with ethyl acetate (3 ⁇ 50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), dried (MgSO 4 ) and evaporated to dryness to produce 0.43 g of crude 9.
  • reaction mixture was quenched by addition of H 2 SO 4 (10 mL, 2 M) and left stirring overnight at room temperature, followed by addition of NaOH (20 mL, 2 M) until basic conditions.
  • the reaction mixture was extracted with ethyl acetate (3 ⁇ 50 mL)) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the combined organic phases were dried (MgSO 4 ) and evaporated to dryness to produce 0.60 g of crude 18.
  • the crude product was subjected to CC [eluent: Tol:EtOAc (1:1)] to give pure 18 (0.32 g, 34%); LC-MS [M+H] + 331 (cald. 331.5).
  • reaction mixture was quenched by addition of H 2 SO 4 (7 mL, 2 M) and stirred at rt for 3 hours, followed by addition of NaOH (20 mL, 2 M) until basic conditions.
  • the reaction mixture was extracted with ethyl acetate (3 ⁇ 50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO 4 ) and evaporated to dryness to produce 0.69 g of crude 19.
  • the crude product was subjected to CC [eluent: CH 2 Cl 2 MeOH (99:1)] to give pure 19 (0.40 g, 64%); LC-MS [M+H] + 315 (cald. 315.5).
  • reaction mixture was quenched by addition of H 2 SO 4 (8 mL, 2 M) and stirred at rt for 4 hours, then the reaction mixture was basified by addition of NaOH (20 mL, 2 M).
  • Addition of THF (20 mL) was followed by extraction with ethyl acetate (3 ⁇ 50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO 4 ) and evaporated to dryness to produce 0.61 g of crude 20.
  • the crude product was subjected to CC [eluent: CH 2 Cl 2 :MeOH (99:1)] to give pure 20 (0.21 g, 35%); LC-MS [M+H] + 315 (cald. 315.5).
  • reaction mixture was extracted with ethyl acetate (3 ⁇ 50 mL)) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the combined organic phases were dried (MgSO 4 ) and evaporated to dryness to produce 1.28 g of crude 26.
  • the crude product was subjected to CC [eluent: Tol:EtOAc (1:1)] to give pure 26 (0.51 g, 51%); LC-MS [M+H] + 393 (cald. 393.7).
  • the reaction mixture was basified by addition of NaOH (2 mL, 2.0 M) and filtered through celite.
  • the aqueous phase was extracted with ethyl acetate (3 ⁇ 50 mL) and the combined organic phases were washed with brine (10 mL) and NaOH (10 mL, 2 M), dried (MgSO 4 ) and evaporated to dryness to produce 0.42 g of crude 27.
  • the crude product was subjected to CC [eluent: CH 2 Cl 2 :MeOH (99:1)] to give pure 27 (0.21 g, 58%); LC-MS [M+H] + 303 (cald. 303.2).
  • NIH 3T3 cells available from the American Type Culture Collection as ATCC CRL 1658
  • NIH 3T3 cells were grown at 37° C. in a humidified atmosphere (5% CO 2 ) incubator in Dulbecco's modified Eagle's medium (DMEM) supplemented with 4.5 g/l glucose, 4 mM glutamine, 50 units/ml penicillin, 50 units/ml streptomycin (available from Advanced Biotechnologies, Inc., Gaithersburg, Md.) and 10% calf serum (available from Sigma, St. Louis, Mich.).
  • DMEM Dulbecco's modified Eagle's medium
  • the cells were treated with trypsin-EDTA, spun down and plated at 2 ⁇ 10 6 per 15 cm dish in 20 ml of DMEM containing 10% calf serum.
  • the m1-m5 muscarinic receptor subtypes were cloned substantially as described by Bonner et al., Science 237, 1987, p. 527, and Bonner et al., Neuron 1, 1988, p. 403.
  • the cells were co-transfected with DNA encoding a chimera between the Gq protein and the five carboxy-terminal amino acids of the Gi protein (the Gq-i5 construct is described by Conklin et al., Nature 363, 1993, p. 274).
  • the cells were transfected using the Superfect transfection reagent (available from Qiagen, Valencia, Calif.) in accordance with the manufacturer's instructions.
  • 60 ⁇ l Superfect was added to the DNA and mixed thoroughly by pipetting up and down several times. The mixture was incubated at room temperature for 10-15 minutes.
  • the media were aspirated, and 12 ml fresh DMEM containing 10% calf serum and 50 units/ml penicillin streptomycin was added to the plates.
  • the DNA-Superfect solution was mixed once more with a pipette and added to the plate which was swirled to distribute the DNA mixture evenly over the surface.
  • the cells were incubated overnight at 37° C. and 5% CO 2 .
  • the media were aspirated and the plates were rinsed once with a 15 ml volume of Hank's Buffered Saline. The plates were swirled to ensure thorough rinsing. 20 ml fresh DMEM supplemented with 10% calf serum and 50 units/ml penicillin/streptomycin was added to the plates. The cells were incubated for 24-48 hours until the plates were 100% confluent.
  • DMEM containing 2% Cyto-SF3 was heated at 37° C. in a water bath under sterile conditions. Sterile working stock solutions of test compounds to be assayed were prepared by diluting the compounds in DMEM to 8 ⁇ the final concentration for testing. The compound (carbachol) to be included in the assay as a positive control was also diluted in DMEM to 8 ⁇ the final concentration. 50 ⁇ l of the DMEM containing 2% Cyto-SF3 was added to each well of 96-well microtiter plates under sterile conditions.
  • HBS Hank's Buffered Saline
  • the cells were centrifuged at 1000 rpm for 5-10 min. in an IEC Centra CL2 centrifuge (produced by Sorvall). Afterwards, the medium was carefully aspirated so as not to dislodge the cells.
  • the cell pellet was suspended in 1600 ⁇ l DMEM containing 10% calf serum and 50 units/ml penicillin and 50 units/ml streptomycin, after which 20 ml DMEM supplemented with 2% Cyto-SF3 was added. 50 ⁇ l aliquots of this cell suspension was added to the wells of the 96-well microtiter plates prepared above (except for the plate control wells). The plates were then incubated for 4 days at 37° C. and 5% CO 2 .
  • FIG. 1 illustrates data from one 96-well plate from the screen. On this plate, two compounds were active at one or more of the transfected receptors. In the total screen, four related compounds were identified that showed activity. To determine which of the receptors were activated in the screen, the compounds were tested as described above against each of the receptors transfected into separate cell cultures. Compound A only activated the m1 receptor subtype, at which it was a potent partial agonist, inducing a lower maximal response than the reference compound carbachol.
  • the four compounds were found to selectively activate the m1 receptor with no significant activity at the m2, m3, m4 or m5 muscarinic receptors.
  • the most active compound, compound A was not an antagonist of carbachol-induced responses of the five muscarinic receptor subtypes.
  • Compound A was further tested for agonist activity against several other receptors at the ⁇ -adrenergic receptor subtypes 1D, 1B, 1A, 2A, 2B and 2C, the histamine H1 and the serotonin 5-HT1A and 5-HT2A subtypes. The compound showed no significant activity in these assays. In antagonist experiments, compound A did not inhibit responses of the ⁇ -adrenergic receptor subtypes 2A, 2B or 2C, or the serotonin receptor subtypes 5-HT1A or 5-HT2A. As illustrated in FIG. 2 , the responses induced by compound A were blocked by the muscarinic antagonist atropine with the same potency as were responses induced by the muscarinic agonist carbachol.
  • R-SAT assays (see U.S. Pat. No. 5,707,798, incorporated herein by reference) were carried out where cells transfected with m1, m3 or m5 receptors were exposed to seven compounds at 1.5 ⁇ M concentration. Cellular response is expressed as a percentage of the maximum response of the cells (defined as response to 10 ⁇ M carbachol). The results are presented in the following table.
  • the compounds are selective agonists of the m1 receptor.

Abstract

Compounds and methods are provided for the alleviation or treatment of diseases or conditions in which modification of muscarinic m1 receptor activity has a beneficial effect. In the method, a therapeutically effective amount of a selective muscarinic m1 agonist compound is administered to a patient in need of such treatment.

Description

    FIELD OF THE INVENTION
  • The present invention relates to novel compounds which are selective for muscarinic acetylcholine receptor subtypes, as well as to methods for activating muscarinic receptors and for treating or alleviating diseases in which modification of muscarinic receptor activity is beneficial.
  • BACKGROUND OF THE INVENTION
  • Muscarinic acetylcholine receptors play a central role in the central nervous system for higher cognitive functions, as well as in the peripheral parasympathetic nervous system. Cloning has established the presence of five distinct muscarinic receptor subtypes (termed m1-m5) (cf. T. I. Bonner et al, Science 237, 1987, pp. 527-532; T. I. Bonner et al., Neuron 1, 1988, pp. 403-410). It has been found that m1 is the predominant subtype in the cerebral cortex and is believed to be involved in the control of cognitive functions, m2 is predominant in heart and is believed to be involved in the control of heart rate, m3 is believed to be involved in gastrointestinal and urinary tract stimulation as well as sweating and salivation, m4 is present in brain, and m5 is present in brain and may be involved certain functions of the central nervous system associated with the dopaminergic system.
  • Animal studies of various muscarinic ligands (S. Iversen, Life Sciences 60 (Nos. 13/14), 1997, pp. 1145-1152) have shown that muscarinic compounds have a profound effect on cognitive functions, e.g. learning and memory. This would suggest a potential utility of muscarinic agonists in the improvement of cognitive functions in diseases characterized by cognitive impairment, both age-related (such as Alzheimer's disease or other dementias) and not age-related (such as attention deficit hyperactivity disorder).
  • Based on the presence of muscarinic receptor subtypes in various tissues, it would appear that the m1 receptor subtype is the more abundant one in the cerebral cortex, basal ganglia and hippocampus where it accounts for 35-60% of all muscarinic receptor binding sites (cf. A. Levey, Proc. Natl. Acad. Sci. USA 93, 1996, pp. 13541-13546). It has been proposed that the m1 (and possibly m4) subtype plays a major role as a postsynaptic muscarinic receptor (located on cholinoceptive neurons in the neocortex and hippocampus) in various cognitive and motor functions and is likely to be a major contributor to the m1 responses measured in these regions of the brain.
  • It has previously been found that conditions associated with cognitive impairment, such as Alzheimer's disease, are accompanied by selective loss of acetylcholine in the brain. This is believed to be the result of degeneration of cholinergic neurons in the basal forebrain which innervate areas of the association cortex and hippocampus involved in higher processes (cf. S. Iversen, supra). This finding would suggest that such conditions may be treated or at least ameliorated with drugs that augment the cholinergic function in the affected areas of the brain.
  • Treatment with acetylcholine esterase (ACHE) inhibitors such as 9-amino-1,2,3,4-tetrahydroacridine (tacrine) results in an increase of acetylcholine in the brain which indirectly causes stimulation of muscarinic receptors. Tacrine treatment has resulted in a moderate and temporary cognitive improvement in Alzheimer's patients (cf. Kasa et al., supra). On the other hand, tacrine has been found to have cholinergic side effects due to the peripheral acetylcholine stimulation. These include abdominal cramps, nausea, vomiting, diarrhea, anorexia, weight loss, myopathy and depression. Gastrointestinal side effects have been observed in about a third of the patients treated. Tacrine has also been found to cause significant hepatotoxicity, elevated liver transaminases having been observed in about 30% of the patients (cf. P. Taylor, “Anticholinergic Agents”, Chapter 8 in Goodman and Gilman: The Pharmacological Basis of Therapeutics, 9th Ed., 1996, pp. 161-176). The adverse effects of tacrine have severely limited its clinical utility. Another AChE inhibitor, (R,S)-1-benzyl-4-[5,6-dimethoxy-1-indanon-2-yl]methylpiperidine.HCl (donepezil), has recently been approved for the treatment of symptoms of mild to moderate Alzheimer's disease (cf. P. Kasa et al, supra). No hepatic damage has been observed for this compound but it has gastrointestinal effects similar to those of tacrine, probably due to stimulation of the m3 receptor caused by elevated parasympathetic tone.
  • It has previously been suggested that, since the muscarinic m1 receptors in the prefrontal cortex and hippocampus appear to be intact, it may be possible to remedy or at least ameliorate the loss of acetylcholine in Alzheimer's disease patients by administration of drugs acting as agonists on those muscarinic receptors (cf. J. H. Brown and P. Taylor, “Muscarinic Receptor Agonists and Antagonists”, Chapter 7 in Goodman and Gilman: The Pharmacological Basis of Therapeutics, 9th Ed., 1996, p. 147).
  • The muscarinic agonists (believed to be m1 selective) hitherto suggested for the treatment of Alzheimer's disease, such as arecoline, have not shown greater efficacy in clinical trials than ACHE inhibitors (cf. S. V. P. Jones et al., supra). In one study (cf. T. Sunderland et al., Brain Res. Rev. 13, 1988, pp. 371-389), arecoline was found to have not so much cognitive enhancing effects as effects on behavioral changes often observed in Alzheimer's disease patients, such as a significant increase in motor activity, significant uplifting of mood, and significant decrease in anergia. However, presumed m1 agonists have later been found to be weak partial agonists selective for the m2 and/or m3 receptor subtypes (H. Bräuner-Osborne et al., J. Med. Chem. 38, 1995, pp. 2188-2195). As indicated above, m2 subtype selectivity is presumed to be responsible for the cardiovascular effects observed for these agonists, e.g. tachycardia and bradycardia, and m3 activity is believed to account for the adverse gastrointestinal effects of the agonists.
  • m2 and/or m3 activity is therefore a significant drawback for the muscarinic agonists proposed until now for the treatment of Alzheimer's disease, severely limiting the doses of the drugs which it has been possible to administer to patients who may therefore have received sub-optimal doses. Furthermore, the lack of subtype selectivity and low potency of the currently tested cholinergic compounds appear to favor the negative peripheral side effects and have limited cognitive effects because of weak and/or opposing actions in the brain. It would therefore be of great advantage to develop compounds which have an improved selectivity for the m1 subtype, but which have little or no activity on the m2 and m3 subtypes.
  • SUMMARY OF THE INVENTION
  • The present invention provides compounds with muscarinic agonist activity of the general formula (I):
    Figure US20080108618A1-20080508-C00001
  • wherein
  • X1, X2, X3, X4 and X5 are selected from C, N and O;
  • k is 0 or 1;
  • t is 0, 1 or 2;
  • R1 is straight or branched-chain C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 heteroalkyl, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, C1-8 hydroxyalkoxy, C1-8 hydroxyalkyl, —SH, C1-8 alkylthio, —O—CH2—C5-6 aryl, —C(O)—C5-6 aryl substituted with C1-3 alkyl or halo; C5-6 aryl or C5-6 cycloalkyl optionally comprising 1 or more heteroatoms selected from N, S and O; —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —CR3R4, —OC(O)R3, —(O)(CH2)sNR3R4 or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H, C1-6 alkyl; C5-6 aryl optionally comprising 1 or more heteroatoms selected from N, O and S, and optionally substituted with halo or C1-4 alkyl; C3-6 cycloalkyl; or R3 and R4 together with the N atom, when present, form a cyclic ring structure comprising 5-6 atoms selected from C, N, S and O; and s is an integer from 0 to 8;
  • A is C5-12 aryl or C5-7 cycloalkyl, each optionally comprising 1 or more heteroatoms selected from N, S and O;
  • R2 is H, amino, hydroxyl, halo, or straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkylthio, C1-6 alkoxycarbonyl, —CN, —CF3, —OR3, —COR3, NO2, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, —C(O)R3R4, —O(CH2)qNR3, —CNR3R4 or —(CH2)qNR3R4; where q is an integer from 1 to 6;
  • n is 0, 1, 2, 3 or 4, the groups R2, when n>1, being the same or different;
  • p is 0 or an integer from 1 to 5;
  • Y is O, S, CHOH, —NHC(O)—, —C(O)NH—, —C(O)—, —OC(O)—, NR7 or —CH═N—, and R7 is H or C1-4 alkyl; or absent; and
  • Z is CR8R9 wherein R8 and R9 are independently selected from H, and straight or branched chain C1-8 alkyl; or
  • a pharmaceutically acceptable salt, ester or prodrug thereof.
  • The present invention further provides pharmaceutical compositions comprising an effective amount of a compound of formula (I).
  • Further provided are methods of treating the symptoms of a disease or condition associated with reduced levels of acetylcholine, said method comprising administering a therapeutically effective amount of a composition comprising a compound of formula (I).
  • In yet a further embodiment, the present invention provides a method of treating the symptoms of a disease or condition associated with increased intraocular pressure, such as, for example, glaucoma, where the method comprises administering a therapeutically effective amount of a composition comprising a muscarinic receptor agonist, preferably an m1 selective agonist, preferably selected from the compounds of formula (I).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing raw data from one 96-well microtiter plate of screening of 35,000 small organic molecules in the assay described in Example XVI.
  • FIG. 2 is a graph showing data comparing the profile of the reference antagonist atropine with m1 muscarinic receptor transfected cells stimulated with either carbachol (open triangles) or compound A (Example I) (closed triangles).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides compounds preferably showing a relatively high selectivity towards the m1 receptor subtype relative to other muscarinic subtypes which may have a beneficial effect in the treatment of cognitive impairment such as Alzheimer's disease or other conditions associated with age-related cognitive decline while avoiding the adverse affects of the drugs hitherto suggested for this purpose. Compounds exhibiting this property have surprisingly been isolated by screening against m1-m5 receptor subtypes.
  • According to one embodiment, the present invention provides compounds of formula (I), wherein
  • X1, X2, X3, X4 and X5 are C; or one of X1, X2, X3, X4 or X5 is O or N and the others are C;
  • k is 0 or 1;
  • t is 1;
  • R1 is straight or branched-chain C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H and C1-6 alkyl; and skis an integer from 1 to 8;
  • n is 1, 2 or 3; and
  • A is phenyl or naphthyl;
  • where R2 is straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, —CN, —CF3, —OH, —COR3, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, or —(CH2)qNR3R4; where q is an integer from 1 to 6; or
  • A is aryl comprising 1 or more heteroatoms selected from N, S and O;
  • R2 is H, halo, straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, —CN, —CF3, OH, —COR3, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3 or —(CH2)qNR3R4; or
  • a pharmaceutically acceptable salt, ester or prodrug thereof.
  • In one preferred embodiment, the compound is of the formula (II):
    Figure US20080108618A1-20080508-C00002
  • Preferred subgeneric embodiments of compounds of formula (II) include compounds of formulas (IIa) and (IIb):
    Figure US20080108618A1-20080508-C00003
  • According to one preferred series of embodiments of compounds of formulas I, II, IIa and IIb, t is 1 and Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—. In another, X3 is C. Preferably, R1 is alkyl, where preferably R2 is alkyl, aminoalkyl, alkoxy or hydroxyl. In one embodiment, p is 3. In another, R1 is C2-8 alkyl and R2 is methyl, hydroxyl or alkoxy.
  • In one embodiment, n is 1 or 2; Y is —C(O)— or O and t is 1. Preferably, R2 is halo. According to other embodiments, t is 0; or R1 is alkoxy, benzyl or phenyl.
  • X3 may also be N, where according to one embodiment, R1 is alkyl or alkoxy; or R1 is benzyl or phenyl; where R2 is alkyl or alkoxy.
  • According to another embodiment, X3 is O, where t may be, for example, 0. Preferably, R2 is alkyl or alkoxy; or R2 is halo. Particular embodiments of the invention include:
    • 4-Methoxy-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxy-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxy-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Butoxy-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methoxymethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxymethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxymethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-(2-Methoxyethyl)-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-(2-Ethoxyethyl)-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methoxy-4-methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methoxy-4-ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methoxy-4-propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methoxy-4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxy-4-methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxy-4-ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxy-4-propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Ethoxy-4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxy-4-methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxy-4-ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxy-4-propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Propoxy-4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butoxy-4-methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butoxy-4-ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butoxy-4-propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butoxy-4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 2-[3-(4-n-Butylpiperidine)propoxy]toluene;
    • 2-[3-(4-n-Butylpiperidine)propanesulfanyl]toluene;
    • 2-[3-(4-n-Butylpiperidine)propanesulfinyl]toluene;
    • 3-(4-n-Butylpiperidine)-o-tolyl-butane-1-thione;
    • 3-(4-n-Butylpiperidinopropyl)-o-tolyl-amine;
    • N-(4-(4-n-Butylpiperidine)-1-o-tolyl-butyl)-hydroxylamine;
    • 4-n-Butyl-1-[4-(2-chlorophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-bromophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-fluorophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-mercaptophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-sulfanylmethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-sulfanylethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-aminophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methylaminophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-ethylaminophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-dimethylaminophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-diethylaminophenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(1-H-imidazol-2-yl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(1-imidazol-1-yl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(1-thiazol-2-yl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-([1,2,3]triazol-1-yl)-4-oxo-1-butyl]piperidine;
    • 2-[4-n-butyl-piperidine-1-ethyl]-8-methyl-3,4-dihydro-2H-naphthalen-1-one;
    • 2-[4-n-butyl-piperidine-1-ethyl]-7-methyl-indan-1-one;
    • 3-[4-n-butyl-piperidine-1-ethyl]-chroman-4-one;
    • 2-[4-n-butyl-piperidine-1-ethyl]-1H-benzoimidazole;
    • 4-n-Butyl-1-[4-(4-fluoro-2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-hydroxyphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methoxyphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(1-thiophen-2-yl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-ethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-ethoxyphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2,4-dimethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2,3-dimethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(3-methoxyphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-benzyloxyphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(4-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-N-phenyl-butyramide;
    • 4-Methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(naphthalen-1-yl)-4-oxo-1-butyl]piperidine;
    • 4-Benzyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 1-[4-(2-methylphenyl)-4-oxo-1-butyl]pyrrolidine;
    • 4-Benzyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 2-Propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 2-Ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Propyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 3,5-Dimethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 4-n-Hexyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 4-Hydroxyethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 4-Ethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 4-Benzyl-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine;
    • 4-Benzyl-1-[4-(4-bromophenyl)-4-oxo-1-butyl]piperidine;
    • 4-Phenyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine;
    • 3-Hydroxymethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Methyl-1-[4-(4-bromophenyl)-4-oxo-1-butyl]piperidine;
    • 1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 2-Hydroxymethyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-Benzyl-1-[4-(2-methylphenyl)-4-oxo-1-pentyl]piperazine;
    • 4-n-Hexyl-1-[4-(2-methylphenyl)-4-oxo-1-pentyl]piperazine;
    • 4-(Piperidine-1-yl)-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 1-[4-(2-methylphenyl)-4-oxo-1-butyl]-2,3-dihydro-1H-indole;
    • 4-Benzyl-1-[5-(2-methylphenyl)-5-oxo-1-pentyl]piperidine;
    • 4-n-Butyl-1-[5-(2-methylphenyl)-5-oxo-1-pentyl]piperidine;
    • 4-n-Butyl-1-[4-(2,6-dimethylphenyl)-4-oxo-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methoxymethylphenyl)-4-oxo-1-butyl]piperidine;
    • 1-(2-Methylphenyl)-2-(4-benzylpiperazin-1-yl)-ethanone;
    • 3,5-Dimethyl-1-[5-(2-methylphenyl)-5-oxo-1-pentyl]piperidine;
    • 3,5-Dimethyl-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine;
    • 1-[4-(4-Fluorophenyl) oxo-1-butyl]pyrrolidine;
    • 4-Benzyl-1-[6-(2-methylphenyl)-6-oxo-1-hexyl]piperazine;
    • 3,5-Dimethyl-1-[6-(2-methylphenyl)-6-oxo-1-butyl]piperidine;
    • 4-Benzyl-1-[5-(2-methoxyphenyl)-5-oxo-1-pentyl]piperazine;
    • 4-Benzyl-1-[3-phenyl-3-oxo-1-propyl]piperazine;
    • 4-n-Butyl-1-[5-(2-methoxyphenyl)-5-oxo-1-pentyl]piperidine;
    • 3,5-Dimethyl-1-[4-(4-fluoro-2-methylphenyl)-4-oxo-1-butyl]piperidine;
    • 3-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]azetidine;
    • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-2-methyl-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-2,2-dimethyl-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-2-ethyl-1-butyl]piperidine;
    • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-2-propyl-1-butyl]piperidine; and
    • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-2,2-diethyl-1-butyl]piperidine.
  • Compounds per se specifically excluded from the scope of formula I are 4-n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine; 2-[3-(3-n-Butylpiperidine)propanesulfanyl]toluene; and 4-Propyloxy-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine (i.e., compounds where —(CH2)p—Y— is —(CH2)3—C(O)— or —(CH2)3—S—; and X1 through X5 are C; such that -A-(R2)n and R1 are not together: o-methyl-phenyl and n-butyl, respectively; phenyl and n-butyl, respectively; or p-fluoro-phenyl and —O—(CH2)2CH3, respectively).
  • The present invention further provides a method of agonizing a muscarinic receptor comprising contacting the receptor with an effective amount of a compound of formula (I), inclusive of all compounds within the scope of formula (I) (i.e., including 4-n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine; 2-[3-(3-n-Butylpiperidine)propanesulfanyl]toluene; and 4-Propyloxy-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine).
  • The present further provides pharmaceutical compositions comprising an effective amount of a compound of formula (I), inclusive of all compounds within the scope of formula (I)(i.e., including -n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine; 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine; 2-[3-(3-n-Butylpiperidine)propane-sulfanyl]toluene; and 4-Propyloxy-1-[4-(4-fluorophenyl)-4-oxo-1-butyl]piperidine).
  • The present invention further also provides methods of treating the symptoms of a disease or condition associated with reduced levels of acetylcholine, the method comprising administering a therapeutically effective amount of a composition described herein. Exemplary diseases or conditions include neurogenerative disease, cognitive impairment, age-related cognitive decline or dementia.
  • The present invention further provides a method of treating the symptoms of a disease or condition associated with increased intraocular pressure, the method comprising administering a therapeutically effective amount of a muscarinic receptor agonist. Glaucoma is a disease in which an abnormality is observed in the circulation-control mechanism of the aqueous humor filling up the anterior chamber, i.e., the space formed between the cornea and the lens. This leads to an increase in the volume of the aqueous humor and an increase in intraocular pressure, consequently leading to the visual field defect and even to loss of eyesight due to the compulsion and contraction of the papillae of the optic nerve. It has been found that muscarinic receptor agonists, and in particular m1 receptor selective agonists, are effective in reducing intraocular pressure. Thus, in a preferred embodiment, the method comprises administering a therapeutically effective amount of a muscarinic receptor agonist comprising m1 receptor agonist activity, and more preferably m1 selective agonist activity. Although any known m1 receptor selective agonist may be used in practicing the invention, preferably the compounds of the present invention as described herein are used. Exemplary agonists available in the art include xanomeline (3-[4-(hexyloxy)-1,2,5-thiadiazol-3-yl]-1,2,5,6-tetrahydro-1-methylpyridine) (Novo Nordisk); talsaclidine (3-(2-propynyloxy)-1-azabicyclo[2.2.2]octane)(Boehringer Ingelheim); Z-1-azabicyclo[2.2.1]heptane-3-one, O-[3-(3-methoxyphenyl)-2-propynyl]oxime (Parke-Davis PD-151832); sabcomeline (R-(Z)-α-(methoxyimino)-1-azabicyclo-[2,2,2]octane-3-acetonitrile HCl)(SmithKline Beecham); 2,8-dimethyl-3-methylene-1-oxa-8-azaspiro[4.5]decane (Yamanouchi Pharmaceutical Co. YM-796); WAY-132983 (Wyeth-Ayerst Research); cevimeline (2′-methyl-spiro[1-azabicyclo[2.2.2]octane-3,5′-[1,3]oxathiolane] (Snow Brand Milk Products Co); alvameline (5-(2-ethyl-2H-tetrazol-5-yl)-1,2,3,6-tetrahydro-1-methyl-pyridine)(H Lundbeck A/S); milameline (1,2,5,6-tetrahydro-1-methyl-3-pyridinecarboxaldehyde, O-methyloxime) (Parke-Davis & Co); and those described in, for example, U.S. Pat. Nos. 5,175,166, 5,403,845, 5,534,522, 4,992,457, 5,571,819, 5,093,333, 5,468,875, 5,510,478, 5,756,501, 5,834,458, EP 384288, WO 9917771, JP 61280497, WO 9700894, WO 9847900, WO 9314089, EP 805153, WO 9422861, WO 9603377, EP 429344, EP 647642, WO 9626196, WO 9800412, WO 9531457, JP 61280497, JP 6298732, JP 6305967, WO 9640687, EP 311313, EP 370415, EP 709381, EP 723781, EP 727208, EP 727209, WO 9740044 and EP 384285, all of which are incorporated herein by reference.
  • The compounds of the present invention preferably show selective agonist activity towards the m1 receptor. Such an agonist is defined as a compound that increases the activity of the m1 muscarinic receptor when it contacts the receptor. With respect to the m1 receptor, selectivity means that an agonist causes a relatively greater increase in the activity of the m1 receptor than of the m3 receptor subtype. Preferably, the agonist causes at least about a 10 fold greater increase in activity of the m1 receptor than of the m3 receptor. Preferably, the m1 selective agonist also causes a relatively greater increase, preferably at least about a 10 fold greater increase, in the activity of an m1 receptor than of an m2; preferably than of an m4; and preferably than of an m5. In one embodiment, selectivity is defined as a property of a muscarinic m1 agonist whereby an amount of agonist effective to increase the activity of the m1 receptor causes little or no increase in the activity of the m3 and m5 subtypes, and preferably the m2 and m4 subtypes.
  • As used herein, the term “alkyl” means a straight or branched-chain alkane group with 1-6 carbon atoms in the chain, for instance methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, etc. The term “heteroalkyl” is intended to indicate an alkane group containing 1 or 2 heteroatoms selected from O, S or N.
  • As used herein, the term “alkenyl” means a straight or branched-chain alkene group with 2-6 carbon atoms in the chain; the term “alkynyl” is intended to indicate a straight or branched-chain alkyne group with 2-6 carbon atoms in the chain.
  • As used herein, the terms “aryl” and “cycloalkyl” preferably refer to mono- and bicyclic ring structures comprising 5 to 12 carbon atoms, more preferably monocyclic rings comprising 5 to 6 carbon atoms. Where such rings comprise one or more heteroatoms, selected from N, S and O, (i.e., heterocyclic rings) such rings comprise a total of 5 to 12 atoms, more preferably 5 to 6 atoms. Heterocyclic rings include, but are not limited to, furyl, pyrrolyl, pyrazolyl, thienyl, imidazolyl, isoxazolyl, oxazolyl, thiazolyl, isothiazolyl, pyridyl, piperidinyl, piperazinyl, pyridazinyl, pyrimidinyl, pyrazinyl, morpholinyl, oxadiazolyl, thiadiazolyl, imidazolinyl, imidazolidinyl and the like. The ring may be substituted by one or more of the groups included in the definition of R2 above. It is understood that the substituents C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl or C1-6 alkoxycarbonyl may, if present, be substituted by one or more of hydroxyl, C1-4 alkoxy, halogen, cyano, amino or nitro.
  • As used herein, the term “halogen” or “halo” includes chlorine, fluorine, iodine and bromine.
  • It is understood that the ring represented by the structure
    Figure US20080108618A1-20080508-C00004
  • may be both saturated and unsaturated.
  • Compounds of the present invention may be prepared by methods analogous to the methods disclosed in GB 1,142,143 and U.S. Pat. No. 3,816,433. Ways of modifying those methods to include other reagents etc. will be apparent to those skilled in the art. Thus, for instance, compounds of formula I may be prepared as shown in the following reaction scheme.
    Figure US20080108618A1-20080508-C00005
  • The starting compound having formula (X) may be prepared by general methods of organic synthesis. For general methods of preparing compounds of formula (X), reference is made to Fuller, R. W. et al., J. Med. Chem. 14:322-325 (1971); Foye, W. O., et al., J. Pharm. Sci. 68:591-595 (1979); Bossier, J. R. et al., Chem. Abstr. 66:46195h and 67:21527a (1967); Aldous, F. A. B., J. Med. Chem. 17:1100-1111 (1974); Fuller, R. W. et al., J. Pharm. Pharmacol. 25:828-829 (1973); Fuller, R. W. et al., Neuropharmacology 14:739-746 (1975); Conde, S. et al., J. Med. Chem. 21:978-981 (1978); Lukovits, I. et al., Int. J. Quantum Chem. 20:429-438 (1981); and Law, B., J. Cromatog. 407:1-18 (1987), the disclosures of which are incorporated by reference herein in their entirety. The radiolabelled derivatives having formula (XX) may be prepared by, for example, using a tritiated reducing agent to form the reductive amination or by utilizing a 14C-labelled starting material.
  • Alternatively, where the starting compound comprises a carbonyl group, the compound having the formula (XXII) may be reduced with, for example, AlH3, diborane:methyl sulfide or other standard carbonyl reducing reagents to produce the ligand having the formula (XXX).
    Figure US20080108618A1-20080508-C00006
  • The receptor ligands having formula (XXXII) may be prepared by nucleophilic displacement of an electrophile (E) by the amino derivative (XXXI). Examples of electrophiles which may be used for this purpose include halides such as I, Cl, Br, tosylate or mesylate.
    Figure US20080108618A1-20080508-C00007
  • When Y in formula (XXXII) is —C(O)—, this compound may be prepared from oxidation of an sec. alcohol with for example pyridinium chlorocromate or N-chlorosuccinimide or CrO3—H2SO4 or nickel peroxide or metal (Al, K) or DCC-DMSO.
  • When Y in formula (XXXII) is —O—, this compound may be prepared by alkylation of an alcohol with arylhalides under for example Cu catalysis.
  • When Y in formula (XXXII) is —S—, this compound may be prepared by alkylation of a thiol with arylhalides under for example Cu catalysis.
  • When Y in formula (XXXII) is —CHOH—, this compound may be prepared by reduction of the corresponding ketone by catalytic hydrogenation or by the use of NaBH4 or by the use of LiAlH4.
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g. sodium or potassium salts; alkaline earth metal salts, e.g. calcium or magnesium salts; and salts formed with suitable organic ligands, e.g. quarternary ammonium salts. Examples of pharmaceutically acceptable salts include the acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium, carbonate, chloride, clavulanate, citrate, dihydrochloride, fumarate, gluconate, glutamate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, nitrate, N-methylglucamine ammonium salt, oleate, oxalate, phosphate/diphosphate, salicylate, stearate, sulfate, succinate, tannate, tartrate, tosylate, triethiodide and valerate salt.
  • The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs are inactive derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
  • Where the compounds according to the invention have at least one chiral center, they may exist as a racemate or as enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention. Furthermore, some of the crystalline forms for compounds of the present invention may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are also included in the scope of this invention.
  • Where the processes for the preparation of the compounds according to the invention give rise to mixtures of stereoisomers, such isomers may be separated by conventional techniques such as preparative chiral chromatography. The compounds may be prepared in racemic form, or individual enantiomers may be prepared either by stereoselective synthesis or by resolution. The compounds may, for example, be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (−)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary.
  • During any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J. F. W. McOmie, Plenum Press, 1973; and T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • Compounds of the present invention may be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever specific pharmacological modification of the activity of muscarinic receptors is required.
  • The present invention also provides pharmaceutical compositions comprising one or more compounds of the invention together with a pharmaceutically acceptable diluent or excipient. Preferably such compositions are in unit dosage forms such as tablets, pills, capsules (including sustained-release or delayed-release formulations), powders, granules, elixirs, tinctures, syrups and emulsions, sterile parenteral solutions or suspensions, aerosol or liquid sprays, drops, ampoules, auto-injector devices or suppositories; for oral, parenteral (e.g. intravenous, intramuscular or subcutaneous), intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation, and may be formulated in an appropriate manner and in accordance with accepted practices such as those disclosed in Remington's Pharmaceutical Sciences, Gennaro, Ed., Mack Publishing Co., Easton Pa., 1990. Alternatively, the compositions may be in sustained-release form suitable for once-weekly or once-monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection. The present invention also contemplates providing suitable topical formulations for administration to, e.g. eye or skin or mucosa.
  • For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, flavoring agents and coloring agents can also be incorporated into the mixture. Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or beta-lactose, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include, without limitation, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • For preparing solid compositions such as tablets, the active ingredient is mixed with a suitable pharmaceutical excipient, e.g. such as the ones described above, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof. By the term “homogeneous” is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. The solid preformulation composition may then be subdivided into unit dosage forms of the type described above containing from 0.1 to about 50 mg of the active ingredient of the present invention. The tablets or pills of the present composition may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner core containing the active compound and an outer layer as a coating surrounding the core. The outer coating may be an enteric layer which serves to resist disintegration in the stomach and permits the inner core to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with conventional materials such as shellac, cetyl alcohol and cellulose acetate.
  • The liquid forms in which the present compositions may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical carriers. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, gelatin, methylcellulose or polyvinyl-pyrrolidone. Other dispersing agents which may be employed include glycerin and the like. For parenteral administration, sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired. The compositions can also be formulated as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration
  • Consequently, the present invention also relates to a method of alleviating or treating a disease or condition in which modification of muscarinic receptor activity, in particular m1 receptor activity, has a beneficial effect by administering a therapeutically effective amount of a compound of the present invention to a subject in need of such treatment. Such diseases or conditions may, for instance arise from inadequate stimulation or activation of muscarinic receptors. It is anticipated that by using compounds which are selective for a particular muscarinic receptor subtype, in particular m1, the problems with adverse side effects observed with the known muscarinic drugs, such as tachycardia or bradycardia or gastrointestinal effects, may substantially be avoided.
  • The term “subject,” as used herein refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • The term “therapeutically effective amount” as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease being treated.
  • Preferably, compounds of general formula I exhibit subtype selectivity for the muscarinic m1 receptor subtype. Likewise, the compounds exhibit selectivity for the muscarinic m1 receptor subtype compared to other human G-protein coupled receptors tested including serotonin, histamine, dopamine or adrenergic receptors. One important implication of this selectivity is that these compounds may be effective in the treatment or amelioration of a number of diseases and disorders of the central nervous system without the undesirable side effects previously observed with non-selective compounds.
  • The ability of the compounds of the present invention to demonstrate muscarinic m1 receptor subtype selectivity makes them potentially very useful in treating a number of diseases and disorders characterized by cognitive impairment such as attention deficit disorder, or neurodegenerative diseases, e.g. Alzheimer's disease, other forms of age-related cognitive decline, e.g. senile dementia, or dementia-related symptoms such as decreased motor activity, mood changes, anergia, apathy, restlessness and aggressive behavior. It is currently believed that the muscarinic m1 receptor may also be involved in control of intraocular pressure, and that muscarinic m1 agonists may therefore be used to treat or alleviate ocular diseases such as glaucoma.
  • Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses two, three or four times daily. Furthermore, compounds for the present invention may be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to persons skilled in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed. A physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the disease or disorder which is being treated.
  • The daily dosage of the products may be varied over a wide range from 0.01 to 100 mg per adult human per day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A unit dose typically contains from about 0.001 mg to about 50 mg of the active ingredient, preferably from about 1 mg to about 10 mg of active ingredient. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0001 mg/kg to about 25 mg/kg of body weight per day. Preferably, the range is from about 0.001 to 10 mg/kg of body weight per day, and especially from about 0.001 mg/kg to 1 mg/kg of body weight per day. The compounds may be administered on a regimen of 1 to 4 times per day.
  • Compounds according to the present invention may be used alone at appropriate dosages defined by routine testing in order to obtain optimal pharmacological effect on a muscarinic receptor, in particular the muscarinic m1 receptor subtype, while minimizing any potential toxic or otherwise unwanted effects. In addition, co-administration or sequential administration of other agents which improve the effect of the compound may, in some cases, be desirable.
  • The pharmacological properties and the selectivity of the compounds of this invention for specific muscarinic receptor subtypes may be demonstrated by a number of different assay methods using recombinant receptor subtypes, preferably of the human receptors if these are available, e.g. conventional second messenger or binding assays. A particularly convenient functional assay system is the receptor selection and amplification assay disclosed in U.S. Pat. No. 5,707,798 describing a method of screening for bioactive compounds by utilizing the ability of cells transfected with receptor DNA, e.g. coding for the different muscarinic subtypes, to amplify in the presence of a ligand of the receptor. Cell amplification is detected as increased levels of a marker also expressed by the cells.
  • The invention is disclosed in further detail in the following examples which are not in any way intended to limit the scope of the invention as claimed.
  • EXAMPLES Example I 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (5)
  • 1-Benzyl-4-n-butylidenepiperidine (2). A 500 mL 3-necked flask fitted with a stirrer, was charged with sodium hydride (1.61 g, 67 mmol) and DMSO (40 mL). The resulting suspension was heated to 90° C. for 30 min, until the evolution of hydrogen ceased. The suspension was cooled on an ice-bath for 20 min followed by addition of a slurry of butyltriphenylphosphonium bromide (26.6 g, 67 mmol) in DMSO (70 mL). The red mixture was stirred for 15 min at rt. 1-Benzyl-4-piperidone 1 (14.0 g, 74 mmol) was slowly added over 30 min, and the mixture was stirred at room temperature over night. H2O (200 mL) was added to the reaction mixture followed by extraction with heptane (4×100 mL) and ethyl acetate (2×100 mL). The combined organic phases were dried and evaporated to dryness, producing 38.1 g of a yellow oil. The oil was distilled to give 14.9 g (88%) of 2, bp 101-105° C. (0.1 mm Hg). 1H NMR (CDCl3) δ 0.90-0.95 (t, 3H), 1.25-1.41 (m, 2H), 1.90-2.20 (m, 2H), 2.18-2.30 (m, 4H), 2.40-2.45 (m, 4H), 2.50 (s, 2H), 5.17 (t, 1H), 7.20-7.42 (m, 5H).
  • 4-n-Butylpiperidine (3). In a 500 mL flask fitted with a stirrer was added a slurry of 2 (13.2 g, 58 mmol) and 10% palladium on charcoal (1.2 g) in ethanol (70 mL), followed by addition of concentrated hydrochloric acid (1.5 mL). The reaction flask was evacuated and hydrogen was added via a reaction flask. A total of 2.5 dm3 of hydrogen was consumed. The reaction mixture was filtered and evaporated and the residue was dissolved in H2O (40 mL) and NaOH (20 mL, 2 M) followed by extraction with ethyl acetate (3×100 mL). The combined organic phases were washed with brine (30 mL) and evaporated to dryness to produce 7.1 g of crude 3. The crude product was subjected to CC [eluent: heptane:EtOAc (4:1)] to give pure 3 (2.7 g, 33%). 1H NMR (CDCl3) δ 0.85 (t, 3H), 1.0-1.38 (m, 9H), 1.65 (dd, 2H), 2.38 (s, 1H), 2.55 (dt, 2H), 3.04 (dt, 2H).
  • 4-(4-n-Butylpiperidin-1-yl)butanenitrile (4). In a 100 mL flask with a magnetic stirrer was placed 3 (2.3 g, 16.4 mmol), 4-bromobutyronitrile (2.4 g, 16.4 mmol), potassium carbonate powder (2.5 g, 18 mmol) in acetonitrile (20 mL). The reaction mixture was stirred at rt for 5 h followed by addition of H2O (15 mL). The mixture was extracted with ethyl acetate (3×30 mL) and the combined organic phases were evaporated to dryness to produce 3.9 of crude 4. The crude product was subjected to CC [eluent: heptane:EtOAc (1:1)] to give pure 4 (2.3 g, 87%). 1H NMR (CDCl3) δ 0.82 (t, 3H), 1.19-1.37 (m, 9H), 1.64-1.75 (d, 2H), 1.84-2.01 (m, 4H), 2.39-2.54 (m, 4H), 2.89-2.97 (d, 2H).
  • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (5). In a 25 mL oven-dried flask was charged Mg turnings (125 mg, 5.2 mmol) which were activated by the use of a heat-gun. Under inert atmosphere was added a suspension of 2-iodoanisole (1.13 g, 5.2 mmol) in Et2O (4 mL) and the reaction mixture was allowed to stand at rt for 1 hour. Compound 4 (720 mg, 3.4 mmol) dissolved in Et2O (4 mL) was added and the mixture was refluxed over-night. THF (15 mL) and sulfuric acid (4 mL, 2 M) was added and the reaction mixture was stirred for 4 h, followed by addition of NaOH (6 mL, 2 M). The reaction mixture was extracted with ethyl acetate (3×50 mL), and the combined organic phases were evaporated to dryness to produce 1.2 g of crude 5. The crude product was subjected to CC [eluent: CH2Cl2:CH3OH (99:1)] to give pure 5 (0.42 g, 26%). 1H NMR (CDCl3) δ 0.83 (t, 3H), 1.20-1.42 (m, 9H), 1.65-1.73 (d, 2H), 1.96-2.20 (m, 4H), 2.53 (t, 2H), 3.02-3.17 (m, 4H), 3.89 (s, 3H), 6.95-7.01 (m, 2H), 7.44 (t, 1H), 7.65 (d, 1H).
  • Example II 3-Hydroxymethyl-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (7)
  • 4-(3-Hydroxymethyl-piperidin-1-yl)-butyronitrile (6). In an oven-dried 25 mL flask was placed piperidine-3-yl-methanol (1.12 g, 10 mmol) in acetonitrile (10 mL), followed by addition of potassium carbonate (1.38 g, 10 mmol) and 4-bromobutyronitrile (0.90 mL, 9 mmol). The reaction mixture was stirred at rt. for 12 h. The mixture was filtered and evaporated to dryness. Addition of H2O (20 mL) was followed by extraction with ethyl acetate (3×20 mL) and the combined organic phases were dried (MgSO4) and evaporated to dryness to produce 1.50 g of crude 6 which was used without further purification in the synthesis of compound 7.
  • 3-Hydroxymethyl-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (7). In a 50 mL oven-dried flask was added Mg turnings (780 mg, 32 mmol), which were activated by the use of a heat-gun under vacuum, followed by addition of anhydrous THF (7 mL). Under inert atmosphere was added a suspension of 2-iodotoluene (5.3 g, 24 mmol) in THF (10 mL) and the reaction mixture was allowed to reflux for 4 hours. A suspension of compound 6 (1.50 g, 8 mmol) in THF (5 mL) was added via a syringe followed by addition of CuBr (23 mg, 0.16 mmol, 2 mol %) and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (20 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (8 mL, 2 M). Addition of THF (15 mL) was followed by extraction with CH2Cl2 (3×20 mL), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.41 g of crude 7. The crude product was subjected to preparative HPLC CC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytical pure sample of compound 7. LC-MS [M+H]+ 275 (cald. 275.2).
  • Example III 2-Propyl-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (9)
  • 4-(2-propyl-piperidin-1-yl)-butyronitrile (8). A mixture of 2-propylpiperidine (550 mg, 4.3 mmol), 4-bromobutyronitrile (430 mg, 3.0 mmol) and potassium carbonate (550 mg, 4.0 mmol) in acetonitrile (5 mL) was stirred at rt for 12 h., followed by addition of a saturated brine (25 mL). The reaction mixture was extracted with ethyl acetate (3×25 mL) and the combined organic phases were dried (MgSO4) and evaporated to dryness to produce crude 8. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (99:1)] to give pure 8 (0.48 g, 83%); LC-MS [M+H]+ 194 (cald. 194.2).
  • 2-Propyl-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (9). In a 10 mL oven-dried flask was added Mg turnings (97 mg, 4.1 mmol) which were activated by the use of a heat-gun under vacuum. Under inert atmosphere was added a suspension of 2-iodotoluene (380 μl, 2.8 mmol) in Et2O (3 mL) and the reaction mixture was allowed to reflux for 1 hour. A mixture of compound 8 (0.43 g, 2.2 mmol) in CH2Cl2 (3 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and stirred at rt for 12 hours followed by addition of NaOH (10 mL, 2 M). Addition of THF (15 mL) was followed by extraction with ethyl acetate (3×50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), dried (MgSO4) and evaporated to dryness to produce 0.43 g of crude 9. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytically pure sample of compound 9; LC-MS [M+H]+ 287 (cald. 287.2).
  • Example IV 1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperidine (11)
  • In a 10 mL oven-dried flask was added Mg turnings (97 mg, 4.1 mmol) which was activated by the use of a heat-gun under vacuum. Under inert atmosphere was added a suspension of 2-iodo-toluene (380 μL, 3.0 mmol) in Et2O (3 mL) and the reaction mixture was allowed to reflux for 1 hours. A suspension of 4-piperidin-1-yl-butanenitrile 10 (Dahlbom et. al. Acta. Chem. Scand. 1951, 5, 690-697) (0.305 mg, 2.0 mmol) in CH2Cl2 (3 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and stirred at rt for 12 hours followed by addition of NaOH (12 mL, 2 M). Addition of THF (15 mL) was followed by extraction with ethyl acetate (3×50 mL), and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and dried (MgSO4) and evaporated to dryness to produce 0.21 g of crude 11. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytically pure sample of compound II; LC-MS [M+H]+ 245 (cald. 245.2).
  • Example V 4-methyl-1-[4-(4-bromophenyl)-4-oxo-1-butyl]piperidine (12)
  • In a 10 mL dried flask was added 4-methylpiperidine (719 μL, 6 mmol), dioxane (5 mL) followed by addition of potassium carbonate (0.30 g, 2.18 mmol), potassium iodide (10 mg) and 4-bromo-4-chlorobutyrophenone (785 mg, 2.76 mmol). The reaction mixture was left at 110° C. for 12 h., followed by dilution with H2O (10 mL). The reaction mixture was extracted with Et2O (3×15 mL) and the combined organic phases are dried (MgSO4) and evaporated to dryness to produce 0.50 g of crude 12. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytical pure sample of compound 12; LC-MS [M+H]+ 322 (cald. 323.1).
  • Example VI 1-[4-(2-methylphenyl)-4-oxo-1-butyl]pyrrolidine (13)
  • In a 10 mL oven-dried flask was charged Mg turnings (30 mg, 1.2 mmol) which were activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a solution of 2-iodotoluene (0.22 g, 1.0 mmol) in Et2O (2 mL) and the reaction mixture was allowed to reflux for 1 hour. A mixture of 4-pyrrolidin-1-yl-butyronitrile (Burckhalter et. al. J. Org. Chem. 1961, 26, 4070-4076) (0.14 g, 1.0 mmol) in CH2Cl2 (2 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (10 mL, 2 M). Addition of THF (15 mL) was followed by extraction with ethyl acetate (3×20 mL), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.12 g of crude 13. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytical pure sample of compound 13. LC-MS [M+H]+ 231 (cald. 231.3).
  • Example VII 4-Methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine (15)
  • 4-(4-Methyl-piperazin-1-yl)-butyronitrile (14). In a 25 mL flask was placed 1-methylpiperazine (0.52 g, 5.1 mmol), 4-bromobutyronitrile (0.78 g, 5.3 mmol) and potassium carbonate (0.71 g, 5.3 mmol) suspended in acetonitrile (5 mL). The reaction mixture was stirred at rt for 4 h., followed by addition of H2O (20 mL) and extraction with ethyl acetate (3×25 mL). The combined organic phases were washed with brine (25 mL), dried (MgSO4) and evaporated to dryness to produce 0.72 g of crude 14 which was used without further purification in the synthesis of compound 15.
  • 4-Methyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine (15). In a 10 mL oven-dried flask was added Mg turnings (116 mg, 4.0 mmol) which were activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a mixture of 2-iodotoluene (0.65 g, 3.0 mmol) in Et2O (3 mL) and the reaction mixture was allowed to reflux for 1 hour. A solution of compound 14 (0.33 g, 2.0 mmol) in CH2Cl2 (3 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (6 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (8 mL, 2 M). Addition of THF (15 mL) was followed by extraction with CH2Cl2 (3×20 mL). The organic phases were dried (MgSO4) and evaporated to dryness to produce 0.26 g of crude 15. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytically pure sample of compound 15. LC-MS [M+H]+ 260 (cald. 260.4).
  • Example VIII 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine (17)
  • 4-(4-Butyl-piperazin-1-yl)-butyronitrile (16). In a 25 mL flask was placed 1-butylpiperazine (712 mg, 5.0 mmol), 4-bromobutyronitrile (779 mg, 5.3 mmol) and potassium carbonate (687 mg, 5.0 mmol) suspended in acetonitrile (5 mL). The reaction mixture was stirred at rt for 12 h., followed by addition of H2O (20 mL) and extraction with ethyl acetate (3×25 mL). The combined organic phases were washed with brine (25 mL), dried (MgSO4) and evaporated to dryness to produce 0.89 g of crude 16 which was used without further purification in the synthesis of compound 17.
  • 4-n-Butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]piperazine (17). In a 10 mL oven-dried flask charged with Mg tunings (100 mg, 4.0 mmol) which was activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a suspension of 2-iodotoluene (0.66 g, 3.0 mmol) in Et2O (3 mL) and the reaction mixture was allowed to reflux for 1 hours. A suspension of compound 16 (0.43 g, 2.0 mmol) in CH2Cl2 (3 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (6 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (8 mL, 2 M). Addition of THF (15 mL) was followed by extraction with CH2Cl2 (3×20 mL), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.50 g of crude 17. The crude product was subjected to preparative HPLC [eluent: Buffer A: 0.1% TFA; Buffer B: 80% CH3CN+0.1% TFA] to produce an analytical pure sample of compound 17. LC-MS [M+H]+ 302 (cald. 302.5).
  • Example IX 4-n-Butyl-1-[4-(2-ethoxyphenyl)-4-oxo-1-butyl]piperidine (18)
  • In a 10 mL oven-dried flask was added Mg turnings (94 mg, 3.8 mmol) which was activated by the use of a heat-gun under vacuum. Under inert atmosphere was added a suspension of 1-ethoxy-2-iodobenzene (0.71 g, 2.9 mmol) in Et2O (3 mL) and the reaction mixture was allowed to reflux for 3 hours. Compound 4 (0.40 g, 1.9 mmol) dissolved in CH2Cl2 (3 mL) was added and the mixture was stirred at 40° C. for additional 3 hours. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and left stirring overnight at room temperature, followed by addition of NaOH (20 mL, 2 M) until basic conditions. The reaction mixture was extracted with ethyl acetate (3×50 mL)) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the combined organic phases were dried (MgSO4) and evaporated to dryness to produce 0.60 g of crude 18. The crude product was subjected to CC [eluent: Tol:EtOAc (1:1)] to give pure 18 (0.32 g, 34%); LC-MS [M+H]+ 331 (cald. 331.5).
  • Example X 4-n-Butyl-1-[4-(2,3-dimethylphenyl)-4-oxo-1-butyl]piperidine (19)
  • In a 10 mL oven-dried flask was added Mg turnings (94 mg, 3.8 mmol) which was activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a suspension of 1-iodo-2,3-dimethylbenzene (0.69 g, 3.0 mmol) in Et2O (5 mL) under spontaneously reflux, and the reaction mixture was allowed to reflux for 4 hours. A suspension of compound 4 (0.41 g, 2.0 mmol) in CH2Cl2 (2 ml) was added to the reaction mixture and left at rt overnight. The reaction mixture was quenched by addition of H2SO4 (7 mL, 2 M) and stirred at rt for 3 hours, followed by addition of NaOH (20 mL, 2 M) until basic conditions. The reaction mixture was extracted with ethyl acetate (3×50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.69 g of crude 19. The crude product was subjected to CC [eluent: CH2Cl2 MeOH (99:1)] to give pure 19 (0.40 g, 64%); LC-MS [M+H]+ 315 (cald. 315.5).
  • Example XI 4-n-Butyl-1-[4-(2,4-dimethylphenyl)-4-oxo-1-butyl]piperidine (20)
  • In a 10 mL oven-dried flask charged with Mg turnings (95 mg, 3.9 mmol) which was activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a suspension of 1-iodo-2,4-dimethylbenzene (0.69 g, 2.9 mmol) in Et2O (4.5 mL) under spontaneously reflux, and the reaction mixture was allowed to reflux for 3 hours. Compound 4 (0.41 g, 2.0 mmol) dissolved in CH2Cl2 (2 mL) was added under inert atmosphere to the reaction mixture and left stirring at rt overnight. The reaction mixture was quenched by addition of H2SO4 (8 mL, 2 M) and stirred at rt for 4 hours, then the reaction mixture was basified by addition of NaOH (20 mL, 2 M). Addition of THF (20 mL) was followed by extraction with ethyl acetate (3×50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.61 g of crude 20. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (99:1)] to give pure 20 (0.21 g, 35%); LC-MS [M+H]+ 315 (cald. 315.5).
  • Example XII 4-n-Butyl-1-[4-(2-methoxyphenyl)-4-oxo-1-butyl]piperidine (21)
  • In a 10 mL oven-dried flask charged with Mg turnings (0.12 g, 4.9 mmol) which was activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a suspension of 1-bromo-2-ethylbenzene (0.66 g, 3.6 mmol) in Et2O (2 mL) and the reaction mixture was allowed to reflux for 2 hours. A suspension of compound 4 (0.50 g, 2.4 mmol) in CH2Cl2 (2 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (14 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (20 mL, 2 M). Addition of THF (20 mL) was followed by extraction with ethyl acetate (3×50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.75 g of crude 21. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (99:1)] to give pure 21 (0.68 g, 90%); LC-MS [M+H]+ 315 (cald. 315.5).
  • Example XIII 4-n-Butyl-1-[4-(2,4-dimethylphenyl)-4-oxo-1-butyl]piperidine (22)
  • A 10 mL oven-dried flask was charged with Mg turnings (88 mg, 3.6 mmol) and was activated under vacuum by the use of a heat-gun. Under inert atmosphere was added a suspension of 1-Iodo-2-methoxymethylbenzene (0.67 g, 2.7 mmol) in Et2O (4 mL) and the reaction mixture was allowed to reflux for 1 hours. A suspension of compound 8 (0.38 g, 1.8 mmol) in CH2Cl2 (4 mL) was added via a syringe and the reaction mixture was stirred at rt overnight. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and stirred at rt for 2 hours followed by addition of NaOH (10 mL, 2 M). Addition of THF (15 mL) was followed by extraction with ethyl acetate (3×50 mL) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the organic phases were dried (MgSO4) and evaporated to dryness to produce 0.51 g of crude 22. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (99:1)] to give pure 22 (0.14 g, 23%); LC-MS [M+H]+ 331 (cald. 331.5).
  • Example XIV 4-n-Butyl-1-[4-(2-pyridinyl)-4-oxo-1-butyl]piperidine (24)
  • 4-(4-Butyl-piperidin-1-yl)butyric acid methyl ester (23). To a 25 mL reaction flask was added 4-bromo-butyric acid methyl ester (2.04 g, 11.2 mmol), compound 3 (1.51 g, 10.8 mmol) and potassium carbonate (1.63 g, 11.8 mmol) suspended in CH3CN (10 mL). The reaction mixture was stirred over-night at rt followed by filtration and evaporation to dryness. Addition of H2O (50 mL) was followed by extraction with ethyl acetate (3×100 mL). The combined organic phases were dried (MgSO4) and evaporated to dryness, to produce 2.84; g of crude 23. The crude product was subjected to CC [eluent: CH2Cl2 MeOH (99:1)] to give pure 23 (1.93 g, 750%). LC-MS [M+H]+ 241 (cald. 241.2).
  • 4-n-Butyl-1-[4-(2-pyridinyl)-4-oxo-1-butyl]piperidine (24). To a dry 25 mL reaction flask was added 2-bromopyridine (200 mg, 1.3 mmol) dissolved in CH2Cl2 (3 mL) and the temperature was adjusted to −78° C. After being stirred for 20 min, addition of n-BuLi (0.84 mL, 1.4 mmol) was conducted under inert atmosphere. After additional 30 min, a solution of 23 dissolved in CH2Cl2 (2 mL) was added. The reaction mixture was left to warm to rt over-night before being quenched with H2SO4 (5 mL, 1 M). The reaction mixture was extracted with ethyl acetate (6×25 mL) and the combined organic phases were dried (MgSO4) and evaporated to dryness, to produce 0.31 g of crude 24. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (10:1)] to give pure 24 (75 mg, 12%). LC-MS [M+H)+ 288 (cald. 288.2).
  • Example XV 4-n-Butyl]-[4-(2-hydroxyphenyl)-4-oxo-butyl]piperidine (27)
  • 1-Benzyloxy-2-iodo-benzene (25). In a 25 mL ovendried flask 2-iodophenol (1.03 g, 4.7 mmol) and potassium carbonate (0.71 g, 5.2 mmol) were dissolved in dry acetone (10 mL). The mixture was stirred for 15 min followed by addition of benzylbromide (0.61 mL, 5.2 mmol) and left over-night at rt. Addition of H2O (50 mL) followed by extraction with ethyl acetate (3×50 mL) and the combined organic phases were dried (MgSO4) and evaporated to dryness, to produce 1.7 g of crude 25. The crude product was subjected to CC [eluent: Heptane:EtOAc (9:1)] to give pure 25 (1.2 g, 81%). LC-MS [M+H]+ 310 (cald. 310.0).
  • 4-n-Butyl-1-[4-(2-benzyloxyphenyl)-4-oxo-1-butyl]piperidine (26). In a 25 mL oven-dried flask was added Mg turnings (123 mg, 5.1 mmol) which was activated by the use of a heat-gun under vacuum. Under inert atmosphere was added a solution of 1-benzyloxy-2-iodo-benzene (25) (1.18 g, 3.8 mmol) in Et2O (10 mL) and the reaction mixture was allowed to reflux for 3.5 hours. A solution of 4-(4-n-butylpiperidin-1-yl)butanenitrile 4 (0.53 g, 2.5 mmol) dissolved in CH2Cl2 (3 mL) was added and the reaction mixture and was stirred at 40° C. over-night. The reaction mixture was quenched by addition of H2SO4 (10 mL, 2 M) and left stirring for 1 hour, followed by addition of NaOH (20 mL, 2 M) until basic conditions. The reaction mixture was extracted with ethyl acetate (3×50 mL)) and the combined organic phases are washed with brine (10 mL) and NaOH (10 mL, 2 M), and the combined organic phases were dried (MgSO4) and evaporated to dryness to produce 1.28 g of crude 26. The crude product was subjected to CC [eluent: Tol:EtOAc (1:1)] to give pure 26 (0.51 g, 51%); LC-MS [M+H]+ 393 (cald. 393.7).
  • 4-n-Butyl-1-[4-(2-hydroxyphenyl)-4-oxo-1-butyl]piperidine (27). To a 25 mL reaction flask was added a solution of 4-n-Butyl-1-[4-(2-benzyloxyphenyl)-4-oxo-1-butyl]piperidine (26) (49 mg, 1.2 mmol) dissolved in dry EtOH (10 mL) and conc. HCl (0.1 mL) followed by addition of palladium on charcoal (40 mg). The reaction flask was then charged with H2 by the use of balloon technique and left stirring at rt over-night under H2 atmosphere. The reaction mixture was basified by addition of NaOH (2 mL, 2.0 M) and filtered through celite. The aqueous phase was extracted with ethyl acetate (3×50 mL) and the combined organic phases were washed with brine (10 mL) and NaOH (10 mL, 2 M), dried (MgSO4) and evaporated to dryness to produce 0.42 g of crude 27. The crude product was subjected to CC [eluent: CH2Cl2:MeOH (99:1)] to give pure 27 (0.21 g, 58%); LC-MS [M+H]+ 303 (cald. 303.2).
  • Example XVI Screening of Test Compounds in an Assay Using Muscarinic Receptor Subtypes m1, m2, m3, m4 and m5
  • Transfection of cells with muscarinic receptor DNAs (general procedure) NIH 3T3 cells (available from the American Type Culture Collection as ATCC CRL 1658) were grown at 37° C. in a humidified atmosphere (5% CO2) incubator in Dulbecco's modified Eagle's medium (DMEM) supplemented with 4.5 g/l glucose, 4 mM glutamine, 50 units/ml penicillin, 50 units/ml streptomycin (available from Advanced Biotechnologies, Inc., Gaithersburg, Md.) and 10% calf serum (available from Sigma, St. Louis, Mich.). The cells were treated with trypsin-EDTA, spun down and plated at 2×106 per 15 cm dish in 20 ml of DMEM containing 10% calf serum.
  • The m1-m5 muscarinic receptor subtypes were cloned substantially as described by Bonner et al., Science 237, 1987, p. 527, and Bonner et al., Neuron 1, 1988, p. 403. For the m2 and m4 receptors, the cells were co-transfected with DNA encoding a chimera between the Gq protein and the five carboxy-terminal amino acids of the Gi protein (the Gq-i5 construct is described by Conklin et al., Nature 363, 1993, p. 274).
  • On day one, the cells were transfected using the Superfect transfection reagent (available from Qiagen, Valencia, Calif.) in accordance with the manufacturer's instructions. Receptor DNA, β-gal DNA (pSI-β-galactosidase available from Promega, Madison, Wis.), chimeric Gq-i5 DNA for the m2 and m4 receptor subtype assays, and salmon sperm DNA (available from Sigma, St. Louis, Mich.) as filler to a total of 20 μg DNA was added per plate. Prior to addition to the plates, 60 μl Superfect was added to the DNA and mixed thoroughly by pipetting up and down several times. The mixture was incubated at room temperature for 10-15 minutes. The media were aspirated, and 12 ml fresh DMEM containing 10% calf serum and 50 units/ml penicillin streptomycin was added to the plates. The DNA-Superfect solution was mixed once more with a pipette and added to the plate which was swirled to distribute the DNA mixture evenly over the surface. The cells were incubated overnight at 37° C. and 5% CO2.
  • After incubation, the media were aspirated and the plates were rinsed once with a 15 ml volume of Hank's Buffered Saline. The plates were swirled to ensure thorough rinsing. 20 ml fresh DMEM supplemented with 10% calf serum and 50 units/ml penicillin/streptomycin was added to the plates. The cells were incubated for 24-48 hours until the plates were 100% confluent.
  • Assay of NIH 3T3 cells transfected with muscarinic receptor subtypes (general procedure) DMEM containing 2% Cyto-SF3 was heated at 37° C. in a water bath under sterile conditions. Sterile working stock solutions of test compounds to be assayed were prepared by diluting the compounds in DMEM to 8× the final concentration for testing. The compound (carbachol) to be included in the assay as a positive control was also diluted in DMEM to 8× the final concentration. 50 μl of the DMEM containing 2% Cyto-SF3 was added to each well of 96-well microtiter plates under sterile conditions. Then, 16 μl of compound solutions were added to the top wells of the plates, and dilution of the solutions was performed by taking 16 μl of the compound solutions from the top wells and pipetting them into the next row of wells. This procedure was repeated with each subsequent row of wells, except that 50 μl medium alone was added to the baseline control wells (the wells that contain medium and cells, but not test compounds) and plate control wells (wells containing medium, but not test compounds and cells). The plates were then placed in an incubator at 37° C. to equilibrate temperature and pH.
  • When the cell cultures had reached 100% confluence, the medium was aspirated and each plate was rinsed with 15 ml Hank's Buffered Saline (HBS). The cells were left in the incubator for about 10-15 minutes until the HBS had turned slightly yellow. The HBS was then aspirated and 1 ml trypsin was added to each plate and swirled so as to completely cover the plates. The edges of the plates were gently rapped several times to loosen the cells. After the cells had been dislodged from the surface, 8 ml DMEM containing 10% calf serum and 50 units/ml penicillin and 50 units/ml streptomycin was added to inhibit the trypsin. The plates were rinsed with this medium, and the cells were pipetted into a tube. The cells were centrifuged at 1000 rpm for 5-10 min. in an IEC Centra CL2 centrifuge (produced by Sorvall). Afterwards, the medium was carefully aspirated so as not to dislodge the cells. The cell pellet was suspended in 1600 μl DMEM containing 10% calf serum and 50 units/ml penicillin and 50 units/ml streptomycin, after which 20 ml DMEM supplemented with 2% Cyto-SF3 was added. 50 μl aliquots of this cell suspension was added to the wells of the 96-well microtiter plates prepared above (except for the plate control wells). The plates were then incubated for 4 days at 37° C. and 5% CO2.
  • After incubation, the medium was removed by inverting the microtiter plates and shaking them gently, after which they were blotted on absorbent paper. 100 μl chromogenic substrate (3.5 mM o-nitrophenyl-β-D-galactopyranoside, 0.5% Nonidet NP-40, in phosphate buffered saline) was added to each well, and the plates were incubated at 30° C. until the optimum absorbance at 405 nm was obtained. The absorbance of the baseline and plate control wells were subtracted from all values.
  • Results Using the general procedure described above, NIH 3T3 cells were co-transfected with DNAs encoding the m1, m3 and m5 receptor subtypes. A compound library containing approximately 35,000 small organic compounds (1 per well) was screened against the receptors by the procedures described above. FIG. 1 illustrates data from one 96-well plate from the screen. On this plate, two compounds were active at one or more of the transfected receptors. In the total screen, four related compounds were identified that showed activity. To determine which of the receptors were activated in the screen, the compounds were tested as described above against each of the receptors transfected into separate cell cultures. Compound A only activated the m1 receptor subtype, at which it was a potent partial agonist, inducing a lower maximal response than the reference compound carbachol.
  • In further experiments, the four compounds were found to selectively activate the m1 receptor with no significant activity at the m2, m3, m4 or m5 muscarinic receptors. The most active compound, compound A, was not an antagonist of carbachol-induced responses of the five muscarinic receptor subtypes.
  • Compound A was further tested for agonist activity against several other receptors at the α-adrenergic receptor subtypes 1D, 1B, 1A, 2A, 2B and 2C, the histamine H1 and the serotonin 5-HT1A and 5-HT2A subtypes. The compound showed no significant activity in these assays. In antagonist experiments, compound A did not inhibit responses of the α-adrenergic receptor subtypes 2A, 2B or 2C, or the serotonin receptor subtypes 5-HT1A or 5-HT2A. As illustrated in FIG. 2, the responses induced by compound A were blocked by the muscarinic antagonist atropine with the same potency as were responses induced by the muscarinic agonist carbachol.
  • Example XVII R-SAT Assay
  • R-SAT assays (see U.S. Pat. No. 5,707,798, incorporated herein by reference) were carried out where cells transfected with m1, m3 or m5 receptors were exposed to seven compounds at 1.5 μM concentration. Cellular response is expressed as a percentage of the maximum response of the cells (defined as response to 10 μM carbachol). The results are presented in the following table.
    Receptor and Concentration
    m1 m3 m5
    Compound 1.5 μM 1.5 μM 1.5 μM
    A (Example I) 107 +/− 9  7 +/− 8 3 +/− 8 
    B (Example IX) 76 +/− 11 7 +/− 9 −6 +/− 10 
    C (Example XV) 91 +/− 9  4 +/− 9 0 +/− 12
    D (Example X) 72 +/− 9  13 +/− 7  2 +/− 15
    E (Example XI) 42 +/− 13 9 +/− 3 −3 +/− 2 
    F (Example XII) 65 +/− 9  9 +/− 7 5 +/− 11
    G* 66 +/− 19 16 +/− 12 7 +/− 11

    *4-n-Butyl-1-[4-phenyl-4-oxo-1-butyl]piperidine
  • As indicated above, the compounds are selective agonists of the m1 receptor.
  • The invention described and claimed herein is not to be limited in scope by the specific embodiments herein disclosed, since these embodiments are intended as illustrations of several aspects of the invention. Any equivalent embodiments are intended to be within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims.
  • Various references are cited herein, the disclosures of which are incorporated by reference in their entireties.

Claims (111)

1. A compound of formula (I):
Figure US20080108618A1-20080508-C00008
wherein
X1, X2, X3, X4 and X5 are selected from C, N and O;
k is 0 or 1;
t is 0, 1 or 2;
R1 is straight or branched-chain C1-8 alkyl, C2-8 alkenyl, C1-8 alkynyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 heteroalkyl, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, C1-8 hydroxyalkoxy, C1-8 hydroxyalkyl, —SH, C1-8 alkylthio, —O—CH2—C5-6 aryl, —C(O)—C5-6 aryl substituted with C1-3 alkyl or halo; C5-6 aryl or C5-6 cycloalkyl optionally comprising 1 or more heteroatoms selected from N, S and O; —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —CR3R4, —OC(O)R3, —(O)(CH2)sNR3R4 or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H, C1-6 alkyl; C5-6 aryl optionally comprising 1 or more heteroatoms selected from N, O and S, and optionally substituted with halo or C1-6 alkyl; C3-6 cycloalkyl; or R3 and R4 together with the N atom, when present, form a cyclic ring structure comprising 5-6 atoms selected from C, N, S and O; and s is an integer from 0 to 8;
A is C5-12 aryl or C5-7 cycloalkyl, each optionally comprising 1 or more heteroatoms selected from N, S and O;
R2 is H, amino, hydroxyl, halo, or straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, C1-6 alkylthio, —CN, —CF3, —OR3, —COR3, NO2, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, —C(O)R3R4, —O(CH2)qNR3, —CNR3R4 or —(CH2)qNR3R4; where q is an integer from 1 to 6;
n is 0, 1, 2, 3 or 4, the groups R2, when n>1, being the same or different;
p is 0 or an integer from 1 to 5;
Y is O, S, CHOH, —NHC(O)—, —C(O)NH—, —C(O)—, —OC(O)—, NR7 or —CH═N—, and R7 is H or C1-4 alkyl; or absent;
Z is CR8R9 wherein R8 and R9 are independently selected from H, and straight or branched-chain C1-8 alkyl;
provided where —(CH2)p—Y— is —(CH2)3—C(O)— or —(CH2)3—S—; and X1 through X5 are C; that -A-(R2)n and R1 are not together:
o-methyl-phenyl and n-butyl, respectively;
phenyl and n-butyl, respectively; or
p-fluoro-phenyl and —O—(CH2)2CH3, respectively; or
a pharmaceutically acceptable salt, ester or prodrug thereof.
2. The compound of claim 1, wherein:
X1, X2, X3, X4 and X5 are C; or one of X1, X2, X3, X4 or X5 is O or N and the others are C;
k is 0 or 1;
t is 1;
R1 is straight or branched-chain C1-8 alkyl, C2-9 alkenyl, C2-8 alkynyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H and C1 alkyl; and s is an integer from 1 to 8;
n is 1, 2 or 3; and
A is phenyl or naphthyl;
where R2 is straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, —CN, —CF3, —OH, —COR3, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, or —CH2)qNR3R4; where q is an integer from 1 to 6; or
A is aryl comprising 1 or more heteroatoms selected from N, S and O;
R2 is H, halo, straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, —CN, —CF3, OH, —COR3, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3 or —(CH2)qNR3R4; or
a pharmaceutically acceptable salt, ester or prodrug thereof.
3. A compound of claim 1 or 2, wherein p is 3.
4. A compound of claim 1 or 2, wherein k is 0.
5. The compound of claim 1:
Figure US20080108618A1-20080508-C00009
6. The compound of claim 5:
Figure US20080108618A1-20080508-C00010
7. A compound of claim 6, wherein t is 1 and Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—.
8. The compound of claim 7, wherein X3 is C.
9. The compound of claim 8, wherein R1 is alkyl.
10. The compound of claim 9, wherein R2 is alkyl, aminoalkyl, alkoxy or hydroxyl.
11. A compound of claim 10, wherein p is 3.
12. A compound of claim 11, wherein R1 is C2-8 alkyl and R2 is methyl, hydroxyl or alkoxy.
13. A compound of claim 12, wherein Y is —C(O)— or O.
14. The compound of claim 9, wherein R2 is halo.
15. The compound of claim 6, wherein t is 0.
16. The compound of claim 8, wherein R1 is alkoxy.
17. The compound of claim 8, wherein R1 is benzyl or phenyl.
18. The compound of claim 7, wherein X3 is N.
19. The compound of claim 18, wherein R1 is alkyl or alkoxy.
20. The compound of claim 18, wherein R1 is benzyl or phenyl.
21. The compound of claim 19, wherein R2 is alkyl or alkoxy.
22. The compound of claim 20, wherein R2 is alkyl or alkoxy.
23. The compound of claim 7, wherein X3 is O.
24. The compound of claim 23, wherein R1 is alkyl.
25. The compound of claim 24, wherein R2 is alkyl or alkoxy.
26. The compound of claim 24, wherein R2 is halo.
27. The compound of claim 5:
Figure US20080108618A1-20080508-C00011
28. A compound of claim 27, wherein Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—.
29. The compound of claim 28, wherein X3 is C.
30. The compound of claim 29, wherein R1 is alkyl.
31. The compound of claim 30, wherein R2 is alkyl, aminoalkyl, alkoxy or hydroxyl.
32. A compound of claim 31, wherein p is 3.
33. A compound of claim 32, wherein R1 is C2-8 alkyl and R2 is methyl, hydroxyl or alkoxy.
34. A compound of claim 33, wherein Y is —C(O)— or O.
35. The compound of claim 30, wherein R2 is halo.
36. The compound of claim 27, wherein t is 0.
37. The compound of claim 29, wherein R1 is alkoxy.
38. The compound of claim 29, wherein R1 is benzyl or phenyl.
39. The compound of claim 28, wherein X3 is N.
40. The compound of claim 39, wherein R1 is alkyl or alkoxy.
41. The compound of claim 39, wherein R1 is benzyl or phenyl.
42. The compound of claim 40, wherein R2 is alkyl or alkoxy.
43. The compound of claim 41, wherein R2 is alkyl or alkoxy.
44. The compound of claim 28, wherein X3 is O.
45. The compound of claim 44, wherein R1 is alkyl.
46. The compound of claim 45, wherein R2 is alkyl or alkoxy.
47. The compound of claim 45, wherein R2 is halo.
48. A method of agonizing a muscarinic receptor comprising contacting said receptor with an effective amount of a compound of formula (I):
Figure US20080108618A1-20080508-C00012
wherein
X1, X2, X3, X4 and X5 are selected from C, N and O;
k is 0 or 1;
t is 0 or 1;
R1 is straight or branched-chain C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 heteroalkyl, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, C1-8 hydroxyalkoxy, C1-9 hydroxyalkyl, —SH, C is thioalkyl, —O—CH2—C5-6 aryl, —C(O)—C5-6 aryl substituted with C1-3 alkyl or halo; C5-6 aryl or C5-6 cycloalkyl optionally comprising 1 or more heteroatoms selected from N, S and O (89620); —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —CR3R4, —OC(O)R3, —C(O)(CH2)sNR3R4 or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H, C1-6 alkyl; C5-6 aryl optionally comprising 1 or more heteroatoms selected from N, O and S, and optionally substituted with halo or C1-6 alkyl; C3-6 cycloalkyl; or R3 and R4 together with the N atom, when present, form a cyclic ring structure comprising 5-6 atoms selected from C, N, S and O; and s is an integer from 0 to 8;
A is C5-12 aryl or C5-7 cycloalkyl, each optionally comprising 1 or more heteroatoms selected from N, S and O;
R2 is H, amino, hydroxyl, halo, or straight or branched-chain C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6 alkoxycarbonyl, —CN, —CF3, —OR3, —COR3, NO2, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, —C(O)R3R4, —O(CH2)qNR3, —CNR3R4 or —(CH2)qNR3R4; where q is an integer from 1 to 6;
n is 0, 1, 2, 3 or 4, the groups R2, when n>1, being the same or different;
p is 0 or an integer from 1 to 5;
Y is O, S, CHOH, —NHC(O)—, —C(O)NH—, —C(O)—, —OC(O)—, NR7 or —CH═N—, and R7 is H or C1-4 alkyl; or absent; and
Z is CR8R9 wherein R5 and R9 are independently selected from H, and straight or branched chain C1-8 alkyl; or
a pharmaceutically acceptable salt, ester or prodrug thereof.
49. A method of claim 48:
Figure US20080108618A1-20080508-C00013
50. A method of claim 49, wherein t is 1 and Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—.
51. The method of claim 50, wherein X3 is C.
52. The method of claim 51, wherein R1 is alkyl.
53. The method of claim 52, wherein R2 is alkyl, aminoalkyl, alkoxy or hydroxyl.
54. A method of claim 53, wherein p is 3.
55. A method of claim 54, wherein R1 is C2-8 alkyl and R2 is methyl, hydroxyl or alkoxy.
56. A method of claim 55, wherein Y is —C(O)— or 0.
57. The method of claim 52, wherein R2 is halo.
58. The method of claim 49, wherein t is 0.
59. The method of claim 51, wherein R1 is alkoxy.
60. The method of claim 51, wherein R1 is benzyl or phenyl.
61. The method of claim 52, wherein X3 is N.
62. The method of claim 61, wherein R1 is alkyl or alkoxy.
63. The method of claim 61, wherein R1 is benzyl or phenyl.
64. The method of claim 62, wherein R2 is alkyl or alkoxy.
65. The method of claim 63, wherein R2 is alkyl or alkoxy.
66. The method of claim 50, wherein X3 is O.
67. The method of claim 66, wherein R1 is alkyl.
68. The method of claim 67, wherein R2 is alkyl or alkoxy.
69. The method of claim 67, wherein R2 is halo.
70. A pharmaceutical composition comprising an effective amount of a compound of formula (I):
Figure US20080108618A1-20080508-C00014
wherein
X1, X2, X3, X4 and X5 are selected from C, N and O;
k is 0 or 1;
t is 0 or 1;
R1 is straight or branched-chain C1-8 alkyl, C2-8 alkenyl, C2-8 alkyl, C1-8 alkylidene, C1-8 alkoxy, C1-8 heteroalkyl, C1-8 aminoalkyl, C1-8 haloalkyl, C1-8 alkoxycarbonyl, C1-8 hydroxyalkoxy, C1-8 hydroxyalkyl, —SH, C1-8 thioalkyl, —O—CH2—C5-6 aryl, —C(O)—C5-6 aryl substituted with C1-3 alkyl or halo; C5-6 aryl or C5-6 cycloalkyl optionally comprising 1 or more heteroatoms selected from N, S and O; —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —CR3R4, —OC(O)R3, —C(O)(CH2)sNR3R4 or —(CH2)sNR3R4; where R3, R4 and R5 are the same or different, each independently being selected from H, C1-6 alkyl; C5-6 aryl optionally comprising 1 or more heteroatoms selected from N, O and S, and optionally substituted with halo or C1-4 alkyl; C3-6 cycloalkyl; or R3 and R4 together with the N atom, when present, form a cyclic ring structure comprising 5-6 atoms selected from C, N, S and O; and s is an integer from 0 to 8;
A is C5-12 aryl or C5-7 cycloalkyl, each optionally comprising 1 or more heteroatoms selected from N, S and O;
R2 is H, amino, hydroxyl, halo, or straight or branched-chain C1 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-4 alkoxy, C1-6 heteroalkyl, C1-6 aminoalkyl, C1-6 haloalkyl, C1-6alkoxycarbonyl, —CN, —CF3, —OR3, —COR3, NO2, —NHR3, —NHC(O)R3, —C(O)NR3R4, —NR3R4, —NR3C(O)NR4R5, —OC(O)R3, —C(O)R3R4, —O(CH2)qNR3, —CNR3R4 or —(CH2)qNR3R4; where q is an integer from 1 to 6;
n is 0, 1, 2, 3 or 4, the groups R2, when n>1, being the same or different;
p is 0 or an integer from 1 to 5;
Y is O, S, CHOH, —NHC(O)—, —C(O)NH—, —C(O)—, —OC(O)—, NR7 or —CH═N—, and R7 is H or C1-4 alkyl; or absent;
Z is CR8R9 wherein R8 and R9 are independently selected from H, and straight or branched chain C1-8 alkyl; or
a pharmaceutically acceptable salt, ester or prodrug thereof; and
a pharmaceutically acceptable carrier.
71. A composition of claim 70 compromising:
Figure US20080108618A1-20080508-C00015
72. A composition of claim 71, wherein t is 1 and Y is —C(O)—, —NHC(O)—, S, O or —OC(O)—.
73. The composition of claim 72, wherein X3 is C.
74. The composition of claim 73, wherein R1 is alkyl.
75. The composition of claim 74, wherein R2 is alkyl, aminoalkyl, alkoxy or hydroxyl.
76. A composition of claim 75, wherein p is 3.
77. A composition of claim 75, wherein R1 is C2-8 alkyl and R2 is methyl, hydroxyl or alkoxy.
78. A composition of claim 77, wherein Y is —C(O)— or O.
79. The composition of claim 74, wherein R2 is halo.
80. The composition of claim 71, wherein t is 0.
81. The composition of claim 73, wherein R1 is alkoxy.
82. The composition of claim 73, wherein R1 is benzyl or phenyl.
83. The composition of claim 74, wherein X3 is N.
84. The composition of claim 82, wherein R1 is alkyl or alkoxy.
85. The composition of claim 82, wherein R1 is benzyl or phenyl.
86. The composition of claim 83, wherein R2 is alkyl or alkoxy.
87. The composition of claim 84, wherein R2 is alkyl or alkoxy.
88. The composition of claim 72, wherein X3 is O.
89. The composition of claim 88, wherein R1 is alkyl.
90. The composition of claim 89, wherein R2 is alkyl or alkoxy.
91. The composition of claim 87, wherein R2 is halo.
92. A method of treating the symptoms of a disease or condition associated with reduced levels of acetylcholine, said method comprising administering a therapeutically effective amount of one or more compounds of claim 1.
93. The method of claim 92, wherein said disease or condition is neurogenerative disease, cognitive impairment, age-related cognitive decline or dementia.
94. A method of treating the symptoms of a disease or condition associated with reduced levels of acetylcholine, said method comprising administering a therapeutically effective amount of a composition of claim 70.
95. The method of claim 94, wherein said disease or condition is neurogenerative disease, cognitive impairment, age-related cognitive decline or dementia.
96. A method of treating the symptoms of a disease or condition associated with increased intraocular pressure, said method comprising administering a therapeutically effective amount of a muscarinic receptor agonist.
97. The method of claim 96, wherein said muscarinic receptor agonist comprises m1 receptor agonist activity.
98. The method of claim 96, wherein said muscarinic receptor agonist is m1 selective.
99. The method of claim 98, wherein agonist causes at least about a 10 fold greater increase in the activity of an m1 receptor subtype than of an m3 receptor subtype.
100. The method of claim 96, wherein said method comprises administering a therapeutically effective amount of one or more compounds of claim 1.
101. The method of claim 98, wherein said method comprises administering a therapeutically effective amount of one or more compounds of claim 1.
102. The method of claim 96, wherein said disease is glaucoma.
103. A method of treating the symptoms of a disease or condition associated with increased intraocular pressure, said method comprising administering a therapeutically effective amount of a composition comprising a muscarinic receptor agonist.
104. The method of claim 103, wherein said muscarinic receptor agonist comprises m1 receptor agonist activity.
105. The method of claim 103, wherein said muscarinic receptor agonist is m1 selective.
106. The method of claim 105, wherein said agonist causes at least about a 10 fold greater increase in the activity of the m1 receptor subtype than of the m3 receptor subtype.
107. The method of claim 103 wherein said method comprises administering a therapeutically effective amount of a composition of claim 70.
108. The method of claim 105, wherein said method comprises administering a therapeutically effective amount of a composition of claim 70.
109. The method of claim 103, wherein said disease is glaucoma.
110. A method of identifying agents capable of reducing intraocular pressure, said method comprising:
contacting a putative agent with an m1 muscarinic receptor subtype;
contacting the putative agent with an m3 muscarinic receptor subtype;
comparing the increase in activity of each receptor subtype; and
identifying agents selective for the m1 muscarinic receptor subtype;
thereby identifying agents capable of reducing intraocular pressure.
111. The method of claim 110, wherein said agent causes at least about a 10 fold greater increase in the activity of the m1 receptor subtype than of the m3 receptor subtype.
US11/933,177 1998-03-31 2007-10-31 Compounds with activity on muscarinic receptors Abandoned US20080108618A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/933,177 US20080108618A1 (en) 1998-03-31 2007-10-31 Compounds with activity on muscarinic receptors

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US8013398P 1998-03-31 1998-03-31
US28277899A 1999-03-31 1999-03-31
ARP990101516 1999-03-31
PCT/US1999/007057 WO1999050247A1 (en) 1998-03-31 1999-03-31 Compounds with activity on muscarinic receptors
USPCT/US1999/007057 1999-03-31
ARP990101516A AR014974A1 (en) 1998-03-31 1999-04-05 COMPOUNDS WITH ACTIVITY ON MUSCARINIC RECEPTORS, A PHARMACEUTICAL COMPOSITION THAT INCLUDES THEM AND THE USE OF THEM FOR THE MANUFACTURE OF A MEDICINE
US09/356,202 US6528529B1 (en) 1998-03-31 1999-07-16 Compounds with activity on muscarinic receptors
US10/338,937 US7485651B2 (en) 1998-03-31 2003-01-07 Compounds with activity on muscarinic receptors
US11/933,177 US20080108618A1 (en) 1998-03-31 2007-10-31 Compounds with activity on muscarinic receptors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/338,937 Continuation US7485651B2 (en) 1998-03-31 2003-01-07 Compounds with activity on muscarinic receptors

Publications (1)

Publication Number Publication Date
US20080108618A1 true US20080108618A1 (en) 2008-05-08

Family

ID=23400557

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/356,202 Expired - Fee Related US6528529B1 (en) 1998-03-31 1999-07-16 Compounds with activity on muscarinic receptors
US10/338,937 Expired - Fee Related US7485651B2 (en) 1998-03-31 2003-01-07 Compounds with activity on muscarinic receptors
US11/933,177 Abandoned US20080108618A1 (en) 1998-03-31 2007-10-31 Compounds with activity on muscarinic receptors

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/356,202 Expired - Fee Related US6528529B1 (en) 1998-03-31 1999-07-16 Compounds with activity on muscarinic receptors
US10/338,937 Expired - Fee Related US7485651B2 (en) 1998-03-31 2003-01-07 Compounds with activity on muscarinic receptors

Country Status (3)

Country Link
US (3) US6528529B1 (en)
AU (1) AU6103000A (en)
WO (1) WO2001005763A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009152392A2 (en) * 2008-06-11 2009-12-17 University Of Toledo Muscarinic agonists for neurological disorders and methods of making the same
US9549928B2 (en) 2011-04-29 2017-01-24 The University Of Toledo Muscarinic agonists as enhancers of cognitive flexibility
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10556013B2 (en) 2017-06-20 2020-02-11 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US10835532B2 (en) 2008-05-15 2020-11-17 The University Of Toledo Muscarinic agonists as cognitive enhancers
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US11530184B2 (en) 2020-06-30 2022-12-20 Imbria Pharmaceuticals, Inc. Crystal forms of 2-[4-[(2,3,4-trimethoxyphenyl)methyl]piperazin-1-yl]ethyl pyridine-3-carboxylate
US11780811B2 (en) 2020-06-30 2023-10-10 Imbria Pharmaceuticals, Inc. Methods of synthesizing 2-[4-[(2,3,4-trimethoxyphenyl)methyl]piperazin-1-yl]ethyl pyridine-3-carboxylate
US11883396B2 (en) 2021-05-03 2024-01-30 Imbria Pharmaceuticals, Inc. Methods of treating kidney conditions using modified forms of trimetazidine

Families Citing this family (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6528529B1 (en) * 1998-03-31 2003-03-04 Acadia Pharmaceuticals Inc. Compounds with activity on muscarinic receptors
US8914114B2 (en) 2000-05-23 2014-12-16 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US6624162B2 (en) 2001-10-22 2003-09-23 Pfizer Inc. Imidazopyridine compounds as 5-HT4 receptor modulators
MXPA03000145A (en) 2002-01-07 2003-07-15 Pfizer Oxo or oxy-pyridine compounds as 5-ht4 receptor modulators.
JP2005522457A (en) * 2002-02-26 2005-07-28 ノース ショア−ロング アイランド ジューイッシュ リサーチ インスティチュート Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
TWI263497B (en) * 2002-03-29 2006-10-11 Lilly Co Eli Pyridinoylpiperidines as 5-HT1F agonists
GB0211230D0 (en) 2002-05-16 2002-06-26 Medinnova Sf Treatment of heart failure
CA2499494A1 (en) 2002-09-20 2004-04-01 Pfizer Inc. N-substituted piperidinyl-imidazopyridine compounds as 5-ht4 receptor modulators
DOP2003000703A (en) 2002-09-20 2004-03-31 Pfizer IMIDAZOPIRADINE COMPOUNDS AS 5-HT4 RECEIVER AGONISTS
TWI299664B (en) 2003-01-06 2008-08-11 Osi Pharm Inc (2-carboxamido)(3-amino)thiophene compounds
US7696225B2 (en) 2003-01-06 2010-04-13 Osi Pharmaceuticals, Inc. (2-carboxamido)(3-Amino) thiophene compounds
EP1613321A2 (en) * 2003-03-28 2006-01-11 Acadia Pharmaceuticals Inc. Muscarinic m1 receptor agonists for pain management
HU227534B1 (en) * 2003-08-04 2011-08-29 Richter Gedeon Nyrt (thio)carbamoyl-cyclohexane derivatives, process for producing them and pharmaceutical compositions containing them
WO2005016276A2 (en) * 2003-08-05 2005-02-24 Samaritan Pharmaceuticals Inc. Sigma-1 receptor ligand with acetylcholinesterase inhibition properties
US20060247248A1 (en) * 2003-08-05 2006-11-02 Vassilios Papadopoulos Sigma-1 receptor ligand with acetylcholinesterase
US10912712B2 (en) 2004-03-25 2021-02-09 The Feinstein Institutes For Medical Research Treatment of bleeding by non-invasive stimulation
JP2007530586A (en) * 2004-03-25 2007-11-01 ザ ファインスタイン インスティテュート フォー メディカル リサーチ Nervous hemostasis
GEP20084550B (en) * 2004-04-30 2008-11-25 Warner Lambert Co Substituted morpholine compounds for the treatment of central nervous system disorders
GB0409744D0 (en) * 2004-04-30 2004-06-09 Pfizer Ltd Novel compounds
MXPA06014486A (en) 2004-06-15 2007-03-01 Pfizer Benzimidazolone carboxylic acid derivatives.
US7737163B2 (en) 2004-06-15 2010-06-15 Pfizer Inc. Benzimidazolone carboxylic acid derivatives
WO2006008118A1 (en) 2004-07-16 2006-01-26 Proteosys Ag Muscarinic antagonists with parp and sir modulating activity as cytoprotective agents
KR20070061578A (en) * 2004-10-05 2007-06-13 아스텔라스세이야쿠 가부시키가이샤 Pharmaceutical composition for xerophthalmia and xerostomia treatment
NZ555320A (en) 2004-12-03 2010-11-26 Schering Corp Substituted piperazines as CB1 antagonists
US11207518B2 (en) 2004-12-27 2021-12-28 The Feinstein Institutes For Medical Research Treating inflammatory disorders by stimulation of the cholinergic anti-inflammatory pathway
ATE489132T1 (en) 2004-12-27 2010-12-15 The Feinstein Inst Medical Res TREATMENT OF INFLAMMATORY DISEASES BY ELECTRICAL STIMULATION OF THE VAGUS NERVE
ME02222B (en) 2004-12-30 2016-02-20 Janssen Pharmaceutica Nv Piperidine- and piperazine-1-carboxylic acid amide derivatives and related compounds as modulators of fatty acid amide hydrolase (faah) for the treatment of anxiety, pain and other conditions
KR20080027384A (en) * 2005-07-08 2008-03-26 마텍 바이오싸이언스스 코포레이션 Polyunsaturated fatty acids for treatment of dementia and pre-dementia-related conditions
JP2009506069A (en) 2005-08-26 2009-02-12 ブレインセルス,インコーポレイティド Neurogenesis through modulation of muscarinic receptors
EP1939189A4 (en) 2005-08-26 2013-03-13 Shionogi & Co Derivative having ppar agonistic activity
EP2258359A3 (en) 2005-08-26 2011-04-06 Braincells, Inc. Neurogenesis by muscarinic receptor modulation with sabcomelin
WO2007070760A2 (en) * 2005-12-15 2007-06-21 Boehringer Ingelheim International Gmbh Compounds which modulate the cb2 receptor
JP2009528266A (en) 2006-01-18 2009-08-06 シェーリング コーポレイション Cannabinoid receptor modifier
MX2008014450A (en) * 2006-05-18 2009-03-09 Mannkind Corp Intracellular kinase inhibitors.
EP2081905B1 (en) * 2006-07-28 2012-09-12 Boehringer Ingelheim International GmbH Sulfonyl compounds which modulate the cb2 receptor
US7998971B2 (en) 2006-09-08 2011-08-16 Braincells Inc. Combinations containing a 4-acylaminopyridine derivative
JP5030114B2 (en) * 2006-09-25 2012-09-19 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Compounds that modulate the CB2 receptor
CA2680477A1 (en) * 2007-03-13 2008-09-18 The Feinstein Institute For Medical Research Treatment of inflammation by non-invasive stimulation
US20100197564A1 (en) * 2007-04-19 2010-08-05 Schering Corporation Diaryl morpholines as cb1 modulators
HUP0700353A2 (en) * 2007-05-18 2008-12-29 Richter Gedeon Nyrt Metabolites of (thio)carbamoyl-cyclohexane derivatives
US7858666B2 (en) 2007-06-08 2010-12-28 Mannkind Corporation IRE-1α inhibitors
EP2170846A2 (en) * 2007-06-28 2010-04-07 Intervet International BV Substituted piperazines as cb1 antagonists
EP2170847A2 (en) * 2007-06-28 2010-04-07 Intervet International BV Substituted piperazines as cb1 antagonists
WO2009029614A1 (en) 2007-08-27 2009-03-05 The Feinstein Institute For Medical Research Devices and methods for inhibiting granulocyte activation by neural stimulation
EP2217565B1 (en) * 2007-11-07 2013-05-22 Boehringer Ingelheim International GmbH Compounds which modulate the cb2 receptor
US7875610B2 (en) * 2007-12-03 2011-01-25 Richter Gedeon Nyrt. Pyrimidinyl-piperazines useful as D3/D2 receptor ligands
ATE552002T1 (en) * 2008-02-21 2012-04-15 Richter Gedeon Nyrt SOLID PREPARATION FOR ORAL ADMINISTRATION
US9662490B2 (en) 2008-03-31 2017-05-30 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation and administration of an anti-inflammatory drug
US9211409B2 (en) * 2008-03-31 2015-12-15 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation of T-cell activity
US20090275997A1 (en) * 2008-05-01 2009-11-05 Michael Allen Faltys Vagus nerve stimulation electrodes and methods of use
US8178568B2 (en) * 2008-07-10 2012-05-15 Boehringer Ingelheim International Gmbh Sulfone compounds which modulate the CB2 receptor
AP2975A (en) 2008-07-16 2014-09-30 Richter Gedeon Nyrt Pharmaceutical formulations containing dopamine receptor ligands
KR20110063438A (en) * 2008-09-25 2011-06-10 베링거 인겔하임 인터내셔날 게엠베하 Sulfonyl compounds which selectively modulate the cb2 receptor
AU2009316801C1 (en) * 2008-11-18 2015-12-24 Setpoint Medical Corporation Devices and methods for optimizing electrode placement for anti-inflammatory stimulation
HU230067B1 (en) 2008-12-17 2015-06-29 Richter Gedeon Nyrt Novel piperazine salt and preparation method thereof
HUP0800766A2 (en) 2008-12-18 2010-11-29 Richter Gedeon Vegyeszet Process for the preparation of piperazine derivatives
HUP0800765A2 (en) 2008-12-18 2010-11-29 Richter Gedeon Nyrt A new process for the preparation of piperazine derivatives and their hydrochloric salts
EP2379080A1 (en) 2009-01-13 2011-10-26 ProteoSys AG Pirenzepine as otoprotective agent
MX2011010359A (en) * 2009-04-02 2012-02-23 Colucid Pharmaceuticals Inc Composition of 2,4,6-trifluoro-n-[6-(1-methyl-piperidin-4-carbony l)-pyridin-2-yl]-benzamide.
US8996116B2 (en) 2009-10-30 2015-03-31 Setpoint Medical Corporation Modulation of the cholinergic anti-inflammatory pathway to treat pain or addiction
US9211410B2 (en) 2009-05-01 2015-12-15 Setpoint Medical Corporation Extremely low duty-cycle activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US8886339B2 (en) 2009-06-09 2014-11-11 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US8788034B2 (en) 2011-05-09 2014-07-22 Setpoint Medical Corporation Single-pulse activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US8299103B2 (en) * 2009-06-15 2012-10-30 Boehringer Ingelheim International Gmbh Compounds which selectively modulate the CB2 receptor
US8383615B2 (en) 2009-06-16 2013-02-26 Boehringer Ingelheim International Gmbh Azetidine 2-carboxamide derivatives which modulate the CB2 receptor
JP2013505295A (en) * 2009-09-22 2013-02-14 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Compound that selectively modulates CB2 receptor
US11051744B2 (en) 2009-11-17 2021-07-06 Setpoint Medical Corporation Closed-loop vagus nerve stimulation
US9833621B2 (en) 2011-09-23 2017-12-05 Setpoint Medical Corporation Modulation of sirtuins by vagus nerve stimulation
EP3636314B1 (en) 2009-12-23 2021-09-08 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
JP2013517271A (en) 2010-01-15 2013-05-16 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Compounds that modulate the CB2 receptor
JP5746228B2 (en) 2010-03-05 2015-07-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Tetrazole compound that selectively modulates CB2 receptor
CA2795062A1 (en) 2010-04-02 2011-10-06 Colucid Pharmaceuticals, Inc. Compositions and methods of synthesis of pyridinoylpiperidine 5-ht1f agonists
UA108233C2 (en) 2010-05-03 2015-04-10 Fatty acid amide hydrolysis activity modulators
US8846936B2 (en) 2010-07-22 2014-09-30 Boehringer Ingelheim International Gmbh Sulfonyl compounds which modulate the CB2 receptor
GB201106817D0 (en) 2011-04-21 2011-06-01 Astex Therapeutics Ltd New compound
WO2013068371A1 (en) 2011-11-08 2013-05-16 Intervet International B.V. Soft chewable dosage form compositions of cannabinoid receptor type 1 (cb-1) antagonists
US9572983B2 (en) 2012-03-26 2017-02-21 Setpoint Medical Corporation Devices and methods for modulation of bone erosion
US8912197B2 (en) 2012-08-20 2014-12-16 Forest Laboratories Holdings Ltd. Crystalline form of carbamoyl-cyclohexane derivatives
US9980973B2 (en) 2012-10-19 2018-05-29 Astex Therapeutics Limited Bicyclic heterocycle compounds and their uses in therapy
GB201218862D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
GB201218864D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
GB201218850D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
EP2803668A1 (en) 2013-05-17 2014-11-19 Boehringer Ingelheim International Gmbh Novel (cyano-dimethyl-methyl)-isoxazoles and -[1,3,4]thiadiazoles
CN105829310B (en) 2013-12-20 2019-04-12 阿斯特克斯治疗有限公司 Bicyclic heterocycles and its therapeutical uses
US11311725B2 (en) 2014-10-24 2022-04-26 Setpoint Medical Corporation Systems and methods for stimulating and/or monitoring loci in the brain to treat inflammation and to enhance vagus nerve stimulation
US10000477B2 (en) 2014-10-31 2018-06-19 Indivior Uk Limited Dopamine D3 receptor antagonist compounds
US10633336B2 (en) 2014-12-19 2020-04-28 The Broad Institute, Inc. Dopamine D2 receptor ligands
EP3233799B1 (en) 2014-12-19 2021-05-19 The Broad Institute, Inc. Dopamine d2 receptor ligands
US11406833B2 (en) 2015-02-03 2022-08-09 Setpoint Medical Corporation Apparatus and method for reminding, prompting, or alerting a patient with an implanted stimulator
US10596367B2 (en) 2016-01-13 2020-03-24 Setpoint Medical Corporation Systems and methods for establishing a nerve block
US11471681B2 (en) 2016-01-20 2022-10-18 Setpoint Medical Corporation Batteryless implantable microstimulators
WO2017127756A1 (en) 2016-01-20 2017-07-27 Setpoint Medical Corporation Control of vagal stimulation
WO2017127758A1 (en) 2016-01-20 2017-07-27 Setpoint Medical Corporation Implantable microstimulators and inductive charging systems
US10583304B2 (en) 2016-01-25 2020-03-10 Setpoint Medical Corporation Implantable neurostimulator having power control and thermal regulation and methods of use
US11274087B2 (en) 2016-07-08 2022-03-15 Richter Gedeon Nyrt. Industrial process for the preparation of cariprazine
WO2019036470A1 (en) 2017-08-14 2019-02-21 Setpoint Medical Corporation Vagus nerve stimulation pre-screening test
US11660443B2 (en) 2018-04-20 2023-05-30 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via electrical trigeminal nerve stimulation
US11260229B2 (en) 2018-09-25 2022-03-01 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via coordinated trigeminal and vagal nerve stimulation
US11547707B2 (en) 2019-04-10 2023-01-10 Richter Gedeon Nyrt. Carbamoyl cyclohexane derivatives for treating autism spectrum disorder
AR119319A1 (en) 2019-07-09 2021-12-09 Lilly Co Eli PROCESSES AND INTERMEDIATE FOR THE LARGE-SCALE PREPARATION OF 2,4,6-TRIFLUORO-N-[6-(1-METHYL-PIPERIDINE-4-CARBONYL)-PYRIDIN-2-IL]-BENZAMIDE HEMISUCCINATE AND PREPARATION OF 2,4,6-TRIFLUORO-N-[6-(1-METHYL-PIPERIDINE-4-CARBONYL)-PYRIDIN-2-IL]-BENZAMIDE
US11938324B2 (en) 2020-05-21 2024-03-26 The Feinstein Institutes For Medical Research Systems and methods for vagus nerve stimulation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5403845A (en) * 1991-08-27 1995-04-04 University Of Toledo Muscarinic agonists
US6528529B1 (en) * 1998-03-31 2003-03-04 Acadia Pharmaceuticals Inc. Compounds with activity on muscarinic receptors

Family Cites Families (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2126329A (en) 1936-03-20 1938-08-09 Hoffmann La Roche Amide derivatives of isoxazole carboxylic acids
US2400913A (en) 1944-02-26 1946-05-28 Smith Kline French Lab New composition of matter
US2695295A (en) 1952-12-19 1954-11-23 Mcneilab Inc Unsymmetrical n, n'-substituted ethylenediamine and piperazine compounds
GB874206A (en) 1956-09-05 1961-08-02 Knoll Ag Basic derivatives of salicylamide
NL271658A (en) 1960-12-01
FR1382425A (en) 1963-01-14 1964-12-18 Ciba Geigy Process for preparing diaza-cyclo-alkanes, inter alia 3-methyl-4-phenyl-1- (2-phenylmercapto-ethyl) -piperazine
GB1053301A (en) 1963-01-14
US3816433A (en) 1965-03-24 1974-06-11 Ferrosan Ab 4-fluoro-ypsilon-(4-methylpiperidino)-butyrophenone and its pharmaceutically acceptable salts
GB1142143A (en) 1965-03-24 1969-02-05 Ferrosan Ab Piperidine-butyrophenones and a process for their production
US3488352A (en) 1966-10-11 1970-01-06 Shulton Inc Basically substituted alkoxy anthranilamides,their corresponding 2-nitro compounds and derivatives thereof
FR1543944A (en) 1967-03-10 1968-10-31 Bruneau & Cie Lab Salicylic acid amide derivatives and their preparation
BE792187A (en) 1971-12-03 1973-03-30 Sumitomo Chemical Co NEW ALKYLAMINE DERIVATIVES
DE2335432A1 (en) 1973-07-12 1975-01-30 Boehringer Mannheim Gmbh 3,4-DIHYDRO-2H-NAPHTHALINONE- (1) 5-OXY-PROPYL-PIPERAZINE DERIVATIVES AND THE METHOD FOR THEIR PRODUCTION
GB1422263A (en) 1973-01-30 1976-01-21 Ferrosan As 4-phenyl-piperidine compounds
GB1459506A (en) 1974-02-18 1976-12-22 Wyeth John & Brother Ltd Piperidine derivatives
GB1498884A (en) 1975-04-15 1978-01-25 Wyeth John & Brother Ltd Aminoacetamide-pyridyl-tetrahydropyridyl and-piperidyl derivatives
US4031101A (en) 1976-04-15 1977-06-21 Parke, Davis & Company Cis-2,6-dimethyl-α,α-diphenyl-1-piperidinebutanol compounds
US4344952A (en) 1980-05-30 1982-08-17 Ab Ferrosan Method of treating diarrhoea with gamma-piperidino-butyrophenones
FR2534912B1 (en) 1982-10-26 1985-06-28 Lafon Labor NOVEL (2,4,6-TRIMETHOXYPHENYL) - (3-PIPERIDINOPROPYL) -CETONE DERIVATIVES, USE IN THERAPEUTICS AND METHOD OF PREPARATION
US4565821A (en) * 1983-03-21 1986-01-21 The Texas A&M University System Method and ophthalmic composition for treating ocular hypertension and glaucoma with butyrophenones
KR890003966B1 (en) 1984-05-23 1989-10-14 스미또모 덴끼 고오교 가부시기가이샤 Wire electrode for wire electro-discharge machine
US4855290A (en) 1985-05-10 1989-08-08 State Of Israel, Represented By Prime Minister's Office, Israel Institute For Biological Research Derivatives of quinuclidine
DE3706585A1 (en) 1987-02-25 1988-09-08 Schering Ag ARYL- AND ARYLOXY-SUBSTITUTED TERT.-ALKYLENAMINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR PHARMACEUTICAL USE
US4992457A (en) 1987-03-18 1991-02-12 Schulman Jerome M Pharmacologically active cholinergic compositions, and methods for making same and use thereof in treating disease
DE3853758T2 (en) 1987-10-05 1995-09-07 Yamanouchi Pharma Co Ltd Heterocyclic spiro compounds and their production.
US4906634A (en) 1988-03-08 1990-03-06 Schering A.G. Novel N-[4-(aminosubstituted)phenyl]methanesulfonamides and their use as cardiovascular agents
US4838373A (en) 1988-06-30 1989-06-13 Caterpillar Inc. Suspension structure for a tracked vehicle
DE8817121U1 (en) 1988-11-22 1993-02-04 Boehringer Ingelheim Kg, 6507 Ingelheim, De
FR2663326B2 (en) 1989-11-17 1992-10-16 Sanofi Sa PYRIDAZINE DERIVATIVES, PREPARATION METHOD AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME.
US5043345A (en) 1989-02-22 1991-08-27 Novo Nordisk A/S Piperidine compounds and their preparation and use
US5276035A (en) 1990-07-26 1994-01-04 Novo Nordisk A/S 1,4-disubstituted piperazines
DK178490D0 (en) 1990-07-26 1990-07-26 Novo Nordisk As 1,4-DISUBSTITUTED PIPERAZINES
CA2050264A1 (en) 1990-08-30 1992-03-01 Raymond Baker Substituted pyrazine and its salts, compositions containing them and their use in medicine
US5093333A (en) 1991-04-29 1992-03-03 American Home Products Corporation N-substituted-2-aminoquinolines useful for treating hypofunction of the cholinergic system
FR2676444B1 (en) 1991-05-16 1995-03-10 Sanofi Elf NOVEL AMINO-3 PYRIDAZINE DERIVATIVES ACTIVE IN THE CENTRAL NERVOUS SYSTEM, PREPARATION METHOD AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME.
EP0591426A4 (en) 1991-06-27 1996-08-21 Univ Virginia Commonwealth Sigma receptor ligands and the use thereof
WO1993014089A1 (en) 1992-01-13 1993-07-22 Novo Nordisk A/S Heterocyclic compounds and their preparation and use
US5470850A (en) 1992-02-24 1995-11-28 Novo Nordisk A/S 2,3,4,5-tetrahydro-1H-3-benzazepines
US5716952A (en) 1992-03-18 1998-02-10 Allergan Method for reducing intraocular pressure in the mammalian eye by administration of muscarinic antagonists
DE4234198A1 (en) 1992-10-10 1994-04-14 Boehringer Ingelheim Kg New heterocyclyl-alkynyl N-(hetero)aryl (thio)carbamate cpds. - useful in treating Alzheimer's disease, senile dementia, cognitive disorders and glaucoma and to improve memory
JPH06298732A (en) 1993-02-16 1994-10-25 Taisho Pharmaceut Co Ltd Indole derivative
DK26793D0 (en) 1993-03-10 1993-03-10 Novo Nordisk As BENZOFURANYL- OR DIHYDROBENZOFURANYL-SUBSTITUTED TRICYCLIC BENZAZEPINES USING AND PREPARING
EP1120416A1 (en) 1993-04-05 2001-08-01 Pharmaceutical Discovery Corporation Pyrido[2.3-b][1,4]benzodiazepinones as M2 receptor ligand for the treatment of neurological disorders
JPH06305967A (en) 1993-04-27 1994-11-01 Taisho Pharmaceut Co Ltd Improving and therapeutic agent for cholinergic nervous insufficiency
AU7177694A (en) 1993-06-23 1995-01-17 Cambridge Neuroscience, Inc. Sigma receptor ligands and the use thereof
JP2983141B2 (en) 1993-10-06 1999-11-29 株式会社三和化学研究所 Novel compound and brain function improving agent containing the compound as active ingredient
FR2717479B1 (en) 1994-03-18 1996-06-07 Pf Medicament News 3- (omega-benzoylpiperidino) alcoyl) -4H-Benzopyran-4-ones, their preparation and their therapeutic application.
GB9409718D0 (en) 1994-05-14 1994-07-06 Smithkline Beecham Plc Novel compounds
AU3138595A (en) * 1994-07-20 1996-02-16 Acea Pharmaceuticals, Inc. Haloperidol analogs and the use thereof
CA2196046A1 (en) 1994-07-27 1996-02-08 Nigel Birdsall Heterocyclic compounds, useful as allosteric effectors at muscarinic receptors
US5672709A (en) 1994-10-24 1997-09-30 Eli Lilly And Company Heterocyclic compounds and their preparation and use
US5571819A (en) 1994-11-22 1996-11-05 Sabb; Annmarie L. Imidazopyridines as muscarinic agents
US5510478A (en) 1994-11-30 1996-04-23 American Home Products Corporation 2-arylamidothiazole derivatives with CNS activity
US5561127A (en) 1994-12-19 1996-10-01 Allelix Biopharmaceuticals, Inc. Muscarinic receptor ligands
US5468875A (en) 1994-12-22 1995-11-21 American Home Products Corporation 1-azabicycloheptane derivatives
TW304167B (en) 1995-01-30 1997-05-01 Lilly Co Eli
US5605701A (en) 1995-02-17 1997-02-25 Eli Lilly And Company Transdermal formulation
PE17897A1 (en) 1995-02-17 1997-06-12 Lilly Co Eli TRANSDERMIC FORMULATION IN PATCH OF 3- (4-BUTILTIO) -1,2,5-TIADIAZOL-3-IL) -1-AZABICICLO (2,2,2) OCTANO
US5889006A (en) 1995-02-23 1999-03-30 Schering Corporation Muscarinic antagonists
IL117149A0 (en) 1995-02-23 1996-06-18 Schering Corp Muscarinic antagonists
WO1996038422A1 (en) 1995-05-31 1996-12-05 Yamanouchi Pharmaceutical Co., Ltd. Fused benzodiazepinone derivatives and medicinal composition of the same
US5534522A (en) 1995-06-07 1996-07-09 Warner-Lambert Company (R)-(Z)-1-azabicyclo [2.2.1] heptan-3-one,O-[3-(3-methoxyphenyl)-2-propynyl] oxime maleate as a pharmaceutical agent
AU711104B2 (en) 1995-06-22 1999-10-07 Novo Nordisk A/S Compounds with growth hormone releasing properties
US5756501A (en) 1995-12-13 1998-05-26 American Home Products Corporation Saturated and unsaturated pyridazino 4,5-B! indolizines useful as antidementia agents
US5726179A (en) 1996-04-01 1998-03-10 The University Of Toledo Muscarinic agonists
AU3055397A (en) 1996-04-23 1997-11-12 Eli Lilly And Company Heterocyclic compounds
EP0910566B1 (en) 1996-04-24 2003-09-17 Novo Nordisk A/S Heterocyclic compounds and their preparation and use
ATE208767T1 (en) 1996-04-30 2001-11-15 Pfizer MUSCARINE RECEPTOR AGONISTS
WO1998000412A1 (en) 1996-07-01 1998-01-08 Schering Corporation Muscarinic antagonists
JP3390179B2 (en) 1996-08-15 2003-03-24 シェーリング コーポレイション Ether muscarinic antagonist
US5834458A (en) 1996-10-09 1998-11-10 Eli Lilly And Company Heterocyclic compounds and their use
ATE299504T1 (en) 1997-04-22 2005-07-15 Novo Nordisk As HETEROCYCLIC COMPOUNDS, THEIR PRODUCTION AND THEIR APPLICATION
EP1017386A1 (en) 1997-10-02 2000-07-12 H. Lundbeck A/S GRANULAR PREPARATIONS OF 5-(2-ETHYL- 2$i(H)-TETRAZOL- 5-YL)-1-METHYL-1, 2,3,6-TETRAHYDROPYRIDINE
RU2230740C2 (en) 1998-03-31 2004-06-20 Акадиа Фармасьютикалз, Инк. Derivatives of nitrogen-containing heterocyclic compounds and pharmaceutical composition based on thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5403845A (en) * 1991-08-27 1995-04-04 University Of Toledo Muscarinic agonists
US6528529B1 (en) * 1998-03-31 2003-03-04 Acadia Pharmaceuticals Inc. Compounds with activity on muscarinic receptors

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10835532B2 (en) 2008-05-15 2020-11-17 The University Of Toledo Muscarinic agonists as cognitive enhancers
WO2009152392A2 (en) * 2008-06-11 2009-12-17 University Of Toledo Muscarinic agonists for neurological disorders and methods of making the same
WO2009152392A3 (en) * 2008-06-11 2010-02-04 University Of Toledo Muscarinic agonists for neurological disorders and methods of making the same
US9549928B2 (en) 2011-04-29 2017-01-24 The University Of Toledo Muscarinic agonists as enhancers of cognitive flexibility
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10918728B2 (en) 2017-06-20 2021-02-16 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US10953102B2 (en) 2017-06-20 2021-03-23 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US10556013B2 (en) 2017-06-20 2020-02-11 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US11376330B2 (en) 2017-06-20 2022-07-05 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US11844840B2 (en) 2017-06-20 2023-12-19 Imbria Pharmaceuticals, Inc. Compositions and methods for increasing efficiency of cardiac metabolism
US11530184B2 (en) 2020-06-30 2022-12-20 Imbria Pharmaceuticals, Inc. Crystal forms of 2-[4-[(2,3,4-trimethoxyphenyl)methyl]piperazin-1-yl]ethyl pyridine-3-carboxylate
US11746090B2 (en) 2020-06-30 2023-09-05 Imbria Pharmaceuticals, Inc. Crystal forms of 2-[4-[(2,3,4- trimethoxyphenyl)methyl]piperazin-1-yl]ethyl pyridine-3-carboxylate
US11780811B2 (en) 2020-06-30 2023-10-10 Imbria Pharmaceuticals, Inc. Methods of synthesizing 2-[4-[(2,3,4-trimethoxyphenyl)methyl]piperazin-1-yl]ethyl pyridine-3-carboxylate
US11883396B2 (en) 2021-05-03 2024-01-30 Imbria Pharmaceuticals, Inc. Methods of treating kidney conditions using modified forms of trimetazidine

Also Published As

Publication number Publication date
US20030144285A1 (en) 2003-07-31
AU6103000A (en) 2001-02-05
WO2001005763A3 (en) 2002-01-17
WO2001005763A2 (en) 2001-01-25
US7485651B2 (en) 2009-02-03
US6528529B1 (en) 2003-03-04

Similar Documents

Publication Publication Date Title
US6528529B1 (en) Compounds with activity on muscarinic receptors
AU762726B2 (en) Compounds with activity on muscarinic receptors
RU2351588C2 (en) N-phenyl(piperidine-2-yl)methyl-benzamide derivatives, and their application in therapy
US5292761A (en) Piperidine, tetrahydropyridine and pyrrolidine compounds
JPH03181461A (en) Antiarrhythmic agent
JP2009508963A (en) Amide compounds and their use as pharmaceutical compositions
CN1468227A (en) Non-imidazole aryloxyalkylamines
JP2714391B2 (en) Tetrahydropyridine oxime cholinergic agent
US6916822B2 (en) Phenoxyalkylamine derivatives useful as opioid δ receptor agonists
US20070185168A1 (en) Piperidine derivatives
DE69433005T2 (en) benzothiazine derivatives
KR100196969B1 (en) Condensed diazepinones, process for their preparation and pharmaceutical compositions containing them
CA2543133A1 (en) Compounds with activity on muscarinic receptors
US3928369A (en) Compounds useful as antidepressive agents, and a process for their preparation
EP1546100B1 (en) New benzoyl piperidine compounds
EP1785415B1 (en) Piperidine derivative or pharmaceutically acceptable salt thereof
WO1995025100A1 (en) Use of 4-amino-5-chloro-2-methoxybenzoic esters as 5-ht4 agonists
NZ270500A (en) 1-indenylethyl-4-naphthyl piperazines and pharmaceutical compositions thereof
EP3666757A1 (en) Process for preparing a piperidin-4-one
FR2717174A1 (en) Use of 4-amino-5-chloro-2-methoxy-benzoic acid piperidino-ethyl ester(s)
CH605924A5 (en) Antidepressant 4-benzofuranyl-piperidine and pyridine derivs.

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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