WO1999062505A2 - Method for treating neurodegenerative disorders - Google Patents

Method for treating neurodegenerative disorders Download PDF

Info

Publication number
WO1999062505A2
WO1999062505A2 PCT/US1999/011702 US9911702W WO9962505A2 WO 1999062505 A2 WO1999062505 A2 WO 1999062505A2 US 9911702 W US9911702 W US 9911702W WO 9962505 A2 WO9962505 A2 WO 9962505A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
disease
hydrogen
aralkyl
compound
Prior art date
Application number
PCT/US1999/011702
Other languages
French (fr)
Other versions
WO1999062505A3 (en
Inventor
Allen B. Reitz
David A. Demeter
Daniel H. S. Lee
Hoau-Yan Wang
Robert H. Chen
Tina Morgan Ross
Malcolm K. Scott
Carlos R. Plata-Salaman
Original Assignee
Ortho-Mcneil Pharmaceutical, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ortho-Mcneil Pharmaceutical, Inc. filed Critical Ortho-Mcneil Pharmaceutical, Inc.
Priority to JP2000551761A priority Critical patent/JP4637351B2/en
Priority to EP99928342A priority patent/EP1083889B1/en
Priority to AT99928342T priority patent/ATE255888T1/en
Priority to DE69913520T priority patent/DE69913520T2/en
Priority to AU45433/99A priority patent/AU765142B2/en
Priority to DK99928342T priority patent/DK1083889T3/en
Priority to CA2333951A priority patent/CA2333951C/en
Publication of WO1999062505A2 publication Critical patent/WO1999062505A2/en
Publication of WO1999062505A3 publication Critical patent/WO1999062505A3/en

Links

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
    • C07D295/08Heterocyclic 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 singly bound oxygen or sulfur atoms
    • C07D295/096Heterocyclic 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 singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/46Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C215/64Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with rings other than six-membered aromatic rings being part of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/54Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C217/74Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with rings other than six-membered aromatic rings being part of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/06Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton
    • C07C229/10Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings
    • C07C229/14Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of carbon skeletons containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/24Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/25Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • 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
    • C07D295/08Heterocyclic 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 singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic 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 singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/088Heterocyclic 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 singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/14Radicals substituted by singly bound hetero atoms other than halogen
    • C07D333/20Radicals substituted by singly bound hetero atoms other than halogen by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/30Phosphinic acids R2P(=O)(OH); Thiophosphinic acids, i.e. R2P(=X)(XH) (X = S, Se)
    • C07F9/32Esters thereof
    • C07F9/3258Esters thereof the ester moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/3282Esters with hydroxyaryl compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline

Definitions

  • the present invention provides a method for treating neurodegenerative disorders. More particularly, a method for treating neurodegenerative disorders (e.g., Alzheimer's disease) by inhibiting the interaction of amyloid beta with alpha- 7 nicotinic acetylcholine receptors.
  • a method for treating neurodegenerative disorders e.g., Alzheimer's disease
  • amyloid beta with alpha- 7 nicotinic acetylcholine receptors.
  • AD Alzheimer's disease
  • AD Parkinson's disease afflict civilization with great suffering and financial loss.
  • AD is characterized by neurofibrillary tangles, neu tic plaques, and neuronal cell death. AD appears as either the familial, early onset ( ⁇ 60 yrs) or late-onset (>60 yrs) forms, with the latter being more prevalent. AD is the major cause of age-related dementia and cognitive impairment
  • amyloid precursor protein APP
  • ⁇ -amyloid ⁇ -40 and A ⁇ - 2 amyloid precursor protein
  • the A ⁇ peptides are derived from APP by proteolytic processing. Dramatic evidence implicating the A ⁇ peptides, particularly A ⁇ 1-42 , in AD comes from various recently identified mutations accounting for certain types of inherited AD. Such mutations in the presenilin (PS1 and PS2) genes are probably the cause of the most frequent form of familial, early-onset AD (Rogaev, E. I. Molecular Biology 1998, 32, 58).
  • a ⁇ -42 has a greater ability than A ⁇ 1-40 to aggregate into the amyloid fibrils that constitute the plaques characteristic of AD (Lansbury, P. T., Jr. Accts. Chem. Res. 1996, 29, 317). Even though is generally present to a much larger degree in the cerebrospinal fluid than A ⁇ 1-42 , it is A ⁇ 2 which is the major A ⁇ peptide found in AD plaques.
  • the A ⁇ peptides can inhibit cholinergic neurotransmitter function independent of neurotoxicity (Auld, D. S.; Kar, S.; Quirion, R. Trends Neurosci. 1998, 21, 43).
  • a ⁇ peptides bind to a number of natural substances such as apoE3, apoE4, apoJ, transthyretin, and albumin.
  • a ⁇ has been reported to interact with a membrane-bound receptor for advanced glycation end products and to the class A scavenger receptor (SR) associated with the production of reactive oxygen species. Stimulation of the alpha-7 subtype of the nicotinic acetylcholine receptors (nAChRs) can protect neurons against A ⁇ cytotoxicity (Kihara, T.
  • nAChRs are members of the ligand-gated ion channel family and appear to be formed from five protein subunits associating together around a central pore (Lindstrom, J. Molecular Neurobiology 1997 ' , 15, 193). These subunits include ⁇ 1- ⁇ 9, ⁇ 1- ⁇ 4, ⁇ , ⁇ , and ⁇ .
  • the ⁇ .7 subtype forms functional homomers which bind to ⁇ - bungarotoxin, a 75-amino acid peptide, with high affinity (0.65-1.7 nM Kd) and nicotine with relatively low affinity (ca. micromolar K d ) (Holladay, M. W.; Dart, M. J.; Lynch, J. K. J. Med. Chem. 1997, 40, 4169).
  • Compounds which block the aggregation of A ⁇ peptides are potentially useful drugs for the treatment of AD.
  • rifampicin inhibits A ⁇ aggregation and neurotoxicity and may show an effect in vivo in diminishing plaque burden when compared with age-matched controls (Tomiyama, T. et al. J. Biol. Chem. 1996, 277, 6839).
  • compounds can be found to either bind to ⁇ 7 nAChRs, to A ⁇ itself, or to both.
  • a ⁇ 1-42 to alpha-7 nAChRs provides a seed for crystallization or deposition of A ⁇ into insoluble deposits, which have the potential to grow into the fibrillar amyloid deposits characteristic of AD. Therefore, blocking the interaction of A ⁇ 1- 2 with alpha-7 nAChRs should reduce the amount of insoluble aggregated A ⁇ that is formed, and thus prevent the neurotoxicity and pathology associated with such aggregated amyloid deposits.
  • Still another object of the invention is to provide a method for identifying compounds which inhibit the binding of A ⁇ peptides with ⁇ 7 nAChRs, either by binding to A ⁇ peptides or to ⁇ 7 nAChRs.
  • the present invention is directed to a method of treating a neurodegenerative disorder in a subject (preferably, a human) in need thereof which comprises administering to the subject an amount of a compound effective to inhibit the binding of an amyloid beta peptide, preferably A ⁇ 1-42 , to alpha-7 nAChRs, preferably, human alpha-7 nAChRs. Since alpha-8 and alpha-9 nAChRs are similar with respect to structure and function to alpha-7 nAChRs, it is possible that blocking the interaction of ⁇ -amyloid with alpha-8 and alpha-9 nAChRs would have therapeutic benefit as well.
  • Neurodegenerative disorders included within the methods of the present invention include, but are not limited to, Alzheimer's disease, Pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson's-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette's disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy's disease), primary lateral sclerosis, familial spastic
  • age-related dementia and other dementias and conditions with memory loss include vascular dementia, diffuse white matter disease (Binswanger's disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica and frontal lobe dementia.
  • vascular dementia diffuse white matter disease
  • dementia of endocrine or metabolic origin dementia of head trauma and diffuse brain damage
  • dementia pugilistica and frontal lobe dementia dementia pugilistica and frontal lobe dementia.
  • other neurodegenerative disorders resulting from cerebral ischemia or infaction including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type (including, but not limited to, epidural, subdural, subarachnoid and intracerebral), and intracranial and intravertebral lesions (including, but not limited to, contusion, penetration, shear, compression and laceration).
  • the neurodegenerative disorder is selected from Alzheimer's disease, Parkinson's disease, Tourette's syndrome, amyotrophic lateral sclerosis, age-related memory loss, senility and age-related dementia, most preferably, the neurodegenerative disorder is Alzheimer's disease.
  • the neurodegenerative disorder is Alzheimer's disease, also.defmed as an amyloidosis
  • other conditions within the methods of the present invention include other amyloidosis which share features including, but not limited to, hereditary cerebral angiopathy, nonneuropathic hereditary amyloid, Down's syndrome, macroglobulinemia, secondary familial Mediterranean fever, Muckle-Wells syndrome, multiple myeloma, pancreatic- and cardiac-related amyloidosis, chronic hemodialysis arthropathy, and Finnish and Iowa amyloidosis.
  • In one embodiment of the invention is a method of treating and/or preventing dementia in an Alzheimer's patient (as well as a method for treating and/or preventing other clinical manifestations of Alzheimer's disease that include, but are not limited to, cognitive and language deficits, apraxias, depression, delusions and other neuropsychiatric symptoms and signs, and movement and gait abnormalities) which comprises administering to the subject a therapeutically effective amount of a compound to inhibit the binding of an amyloid beta peptide (preferably, A ⁇ ! - 42 ) with nAChRs, preferable alpha-7 nAChRs, most preferably, human alpha-7 nAChRs.
  • an amyloid beta peptide preferably, A ⁇ ! - 42
  • a second embodiment of the invention is a method of improving memory and/or mental status and/or of halting the progression of mental deterioration in an Alzheimer's disease patient which comprises administering to the subject a therapeutically effective amount of a compound to inhibit the binding of an amyloid beta peptide (preferably, A ⁇ - 42 ) with nAChRs, preferably alpha-7 nAChRs, most preferably, human alpha-7 nAChRs.
  • an amyloid beta peptide preferably, A ⁇ - 42
  • the compound used in the methods of treating neurodegenerative disorders, treating and/or preventing Alzheimer's disease, and improving memory and/or halting the progression of mental deterioration in an Alzheimer's disease patient is not estrogen, raloxifene, droloxifene, tamoxifen, idoxifene or levomeloxifene; more preferably, the compound is not estrogen or a selective estrogen receptor modulator (SERM).
  • SERM is an estrogen receptor ligand that exhibits estrogen agonist activity in the cardiovascular system, CNS and bone, and estrogen antagonist activity in reproductive tissues, such as breast and uterus.
  • Also included in the invention is the use of a compound which inhibits the binding of an amyloid beta peptide (preferably to an alpha-7 nAChR (preferably, a human alpha-7 nAChR) in the preparation of a medicament for the treatment of a neurodegenerative disorder in a subject (preferably, a human) in need thereof.
  • an amyloid beta peptide preferably to an alpha-7 nAChR (preferably, a human alpha-7 nAChR) in the preparation of a medicament for the treatment of a neurodegenerative disorder in a subject (preferably, a human) in need thereof.
  • Another illustration of the invention is the use of a compound which inhibits the binding of an amyloid beta peptide (preferably A ⁇ - 2 ) to alpha-7 nAChRs (preferably, human alpha-7 nAChRs) in the preparation of a medicament for: a) improving memory, b) halting the progression of the mental deterioration seen in Alzheimer's disease patients, c) treating dementia, d) preventing dementia in an Alzheimer's patient, and e) treating and/or preventing other clinical manifestations of Alzheimer's disease that include, but are not limited to, cognitive and language deficits, apraxias, depression, delusions and other neuropsychiatric symptoms and signs, and movement and gait abnormalities in an Alzheimer's patient.
  • an amyloid beta peptide preferably A ⁇ - 2
  • alpha-7 nAChRs preferably, human alpha-7 nAChRs
  • Ri is hydrogen or C ⁇ -C 4 alkyl
  • R is selected from hydrogen, d-C ⁇ alkyl, aryl or C 7 -C ⁇ 0 aralkyl;
  • R3 is selected from hydrogen, C ⁇ -C 6 alkyl, C3-C 10 alkenyl,
  • R 2 and R 3 together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
  • R is C C 6 alkyl, aryl, or C 7 -C ⁇ 0 aralkyl; and Rsand R 6 are each independently selected from hydrogen, C ⁇ -C 6 alkyl, C3-C10 alkenyl, C ⁇ -C 8 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
  • Ri is hydrogen; R 2 is selected from hydrogen or C ⁇ -C 4 alkyl;
  • R 3 is selected from C ⁇ -C alkyl, C3-C10 alkenyl, C 5 -C 6 cycloalkylC ⁇ -C4 6 alkyl, d-Ce alkoxycarbonylC ⁇ -C 4 alkyl, C ⁇ -C 6 alkylthio, heteroaryiC ⁇ -C 4 alkyl, or unsubstituted or substituted C 7 -C ⁇ o aralkyl wherein the substituent on the aralkyl are one or two substituents independently selected from the group consisting of halogen, hydroxy, C ⁇ -C 4 alkyl and unsubstituted or substituted C ⁇ -C alkoxy wherein the substituents on the alkoxy are one or two substituents independently selected from amino, C ⁇ -C alkylamino, C ⁇ -C 4 dialkylamino, pyrrolidinyl, or piperidinyl; or
  • R 4 is C ⁇ -C 4 alkyl
  • R 5 and R 6 are each independently selected from hydrogen, C 1 -C alkyl, C 3 -C 6 alkenyl, C ⁇ -C 6 alkylcarbonyl, or diphenylphosphinyl.
  • Ri is hydrogen or C-i-C 4 alkyl
  • R 2 and R 3 are each independently selected from hydrogen, Ci-Ce alkyl, aryl or C 7 -C ⁇ o aralkyl; and l ⁇ is C ⁇ -C 6 alkyl, aryl, or C7-C10 aralkyl; and pharmaceutically acceptable salts thereof.
  • Compounds of formula I are novel compounds which block the interaction of beta-amyloid with alpha-7 nAChRs. More specifically, the compounds of formula I inhibit the binding of human alpha-7 nAChRs by binding to A ⁇ -42 .
  • the orientation between the nitrogen atom and R. 1 on the appropriate ring can be either cis or trans.
  • the compound is 5,8-dihydroxy-frans-2-di( ⁇ /-propylamino)-3-methyl-1 ,2,3,4- tetrahydronaphthalene, and pharmaceutically acceptable salts thereof.
  • Other compounds useful in the methods of the present invention inhibit the binding of A ⁇ - 42 with human alpha-7 nAChRs by binding to human alpha- 7 nAChRs directly.
  • An example of such a compound which binds to human alpha-7 nAChRs is ⁇ -bungarotoxin .
  • the present invention provides methods of treating neurodegenerative disorders by inhibiting the binding of amyloid beta peptides to alpha-7 nAChRs.
  • Neurodegenerative disorders included within the methods of the present invention include, but are not limited to, Alzheimer's disease, Pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson's-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette's disease, bulbar
  • age-related dementia and other dementias and conditions with memory loss include vascular dementia, diffuse white matter disease (Binswanger's disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica and frontal lobe dementia.
  • vascular dementia diffuse white matter disease
  • dementia of endocrine or metabolic origin dementia of head trauma and diffuse brain damage
  • dementia pugilistica and frontal lobe dementia dementia pugilistica and frontal lobe dementia.
  • other neurodegenerative disorders resulting from cerebral ischemia or infaction including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type (including, but not limited to, epidural, subdural, subarachnoid and intracerebral), and intracranial and intravertebral lesions (including, but not limited to, contusion, penetration, shear, compression and laceration).
  • the neurodegenerative disorder is selected from Alzheimer's disease, Parkinson's disease, Tourette's syndrome, amyotrophic lateral sclerosis, age-related memory loss, senility and age-related dementia, most preferably, the neurodegenerative disorder is Alzheimer's disease. Because, most preferably, the neurodegenerative disorder is Alzheimer's disease, also .
  • amyloidosis other conditions within the methods of the present invention include other amyloidosis which share features including, but not limited to, hereditary cerebral angiopathy, nonneuropathic hereditary amyloid, Down's syndrome, macroglobulinemia, secondary familial Mediterranean fever, Muckle-Wells syndrome, multiple myeloma, pancreatic- and cardiac-related amyloidosis, chronic hemodialysis arthropathy, and Finnish and Iowa amyloidosis.
  • amyloid beta refers to amyloid beta peptides and include the A ⁇ -4 0 , peptides and their fragments.
  • fragments of amyloid beta peptides include, but not limited to, fragment 1-28, and fragment 25-35 (e.g., Yatin SM, Aksenov M, Butterfield DA. Neurochem Res 1999 Mar;24(3):427-35; Hirakura Y, Satoh Y, Hirashima N, Suzuki T, Kagan BL, Kirino Y.
  • 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 or disorder being treated.
  • alkyl shall mean straight or branched chain alkanes of one to ten carbon atoms, or any number within this range.
  • alkyl radicals include, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, 3-(2-methyl)butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl and 2-methylpentyl.
  • Alkoxy radicals are oxygen ethers formed from the previously described straight or branched chain alkyl groups.
  • Cycloalkyl groups contain 3 to 8 ring carbons and preferably 5 to 7 carbons.
  • alkenyl and alkynyi groups include straight and branched chain alkenes and aikynes having 2 to 10 carbon atoms, or any number within this range.
  • aryl indicates aromatic groups such as phenyl and naphthyl.
  • C7-C10 aralkyl means an alkyl group substituted with an aryl group wherein the total number of carbon atoms is between 7 and 10 (e.g., benzyl, phenylethyl, phenylpropyl).
  • heteroaryl represents an unsubstituted or substituted stable five or six membered monocyclic aromatic ring system or an unsubstituted or substituted nine or ten membered benzo-fused heteroaromatic ring system or bicyclic heteroaromatic ring system which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heteroaryl group may be attached at any heteroatom or carbon atom that results in the creation of a stable structure.
  • heteroaryl groups include, but are not limited to pyridyl, pyridazinyl, thienyl, furanyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzoxazolyl, benzopyrazolyl, indolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl adeninyl or quinolinyl.
  • Prefered heteroaryl groups include pyridyl, pyrrolyl, pyrazinyl, thiadiazolyl, pyrazolyl, thienyl, triazolyl and quinolinyl.
  • N(CH 2 ) 5 means a piperidinyl group.
  • cC 6 Hn refers to a cyclohexyl group.
  • substituted e.g., aryl, aralkyl
  • that group may have one or more substituents, preferably from one to five substituents, more preferably from one to three substituents, most preferably from one to two substituents, independently selected from the list of substituents.
  • a "phenyld-C 6 alkylamidoC ⁇ -C 6 alkyl" substituent refers to a group of the formula
  • Compounds which are useful in the methods of the present invention for inhibiting the interaction of A ⁇ -40 and A ⁇ -42 to the alpha-7 subtype of nAChRs for either the purpose of direct therapeutic intervention or in order to screen for compounds which act via this mechanism include compounds of formula I, especially 5, 8-dihydroxy-fra/?s-2-di( ⁇ /-propylamino)-3-methyl-1 , 2,3,4- tetrahydronaphthalene (Compound 9), (-)-nicotine, (rac)-epibatidine, ⁇ - bungarotoxin, and pharmaceutically acceptable salts thereof.
  • Certain peptide stretches of the human alpha-7 nAChR bind to amyloid beta and can be used in place of the alpha-7 nAChR together with amyloid beta for the purpose of screening libraries to find compounds which block the interaction of amyloid beta and the human alpha-7 nAChR. Included among these peptide stretches of the human alpha-7 nAChR are alpha-7 nAChRI 93- 224 and smaller peptides derived thereof as listed in the Table. Table
  • the salts of the compounds of this invention refer to non-toxic "pharmaceutically acceptable salts.”
  • Other salts may, however, be useful in the preparation of compounds according to this invention or of their pharmaceutically acceptable salts.
  • 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 with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, 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., quaternary ammonium salts.
  • alkali metal salts e.g., sodium or potassium salts
  • alkaline earth metal salts e.g., calcium or magnesium salts
  • suitable organic ligands e.g., quaternary ammonium salts.
  • the present invention includes within its scope prodrugs of the compounds of this invention.
  • a prodrug is inactive as administered, but becomes activated in vivo.
  • the prodrug is converted to the parent drug chemically or by specific enzyme(s).
  • prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the required compound.
  • the term “administering” shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
  • the compounds according to this invention may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention.
  • (-)-Nicotine is (-)-1-methyl-2-(3-pyridinyl)pyrrolidine and is readily available from Sigma Chemical Company.
  • (+/-)-Epibatidine is exo-(+/-)-2-(6-chloro-3-pyridinyl)-7- azabicyclo[2.2.1]heptane and is readily available from Sigma Chemical Company.
  • ⁇ -Bungarotoxin is a 74 amino acid peptide which is commercially available from Research Biochemicals Inc. ⁇ -Bungarotoxin and its amino acid sequence are described in Lee, C. Y. Annu. Rev. Pharmacol. 1972, 12, 265- 281.
  • 125, -A ⁇ - 0 , and anti-alpha-7 nAChR antibodies are commercially available Amersham Pharmacia Biotech, Advanced Bioconcepts and Research Biochemicals International, respectively.
  • 125 l- ⁇ -bungarotoxin is commercially available from Amersham Pharmacia Biotech.
  • the present invention therefore provides a method of treating a neurodegenerative disorder, which comprises administering any of the compounds as defined herein in a quantity effective to treat the neurodegenerative disorder.
  • the compound is not estrogen, raloxifene, droloxifene, tamoxifen, idoxifene or levomeloxifene; more preferably, the compound is not estrogen or a selective estrogen receptor modulator (SERM).
  • SERM selective estrogen receptor modulator
  • the compound may be administered to a patient afflicted with a neurodegenerative disorder by any conventional route of administration, including, but not limited to, intravenous, oral, subcutaneous, intramuscular, intradermal, buccal, intracerebral and other parenteral routes.
  • the quantity of the compound which is effective for treating a neurodegenerative disorder is between 0.01 mg per kg and 10 mg per kg of subject body weight.
  • the method of treating neurodegenerative disorders described in the present invention may also be carried out using a pharmaceutical composition
  • a pharmaceutical composition comprising any of the compounds as defined herein and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may contain between about 0.5 mg and 200 mg of the compound, and may be constituted into any form suitable for the mode of administration selected.
  • Carriers include necessary and inert pharmaceutical excipients, including, but not limited to, binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, dyes, and coatings.
  • Compositions suitable for oral administration include solid forms, such as pills, capsules, granules, tablets, caplets, and powders, and liquid forms, such as solutions, syrups, elixers, and suspensions.
  • Forms useful for intracerebral and other parenteral routes of administration include sterile solutions, emulsions and suspensions.
  • Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular compound used, the mode of administration, the strength of the preparation, the mode of administration, and the advancement of the disease condition. In addition, factors associated with the particular patient being treated, including patient age, body weight, diet, physical activity and time of administration, and associated co-morbidities and clinical conditions will result in the need to adjust dosages.
  • the present invention also provides diagnostic tools useful for diagnosing Alzheimer's disease.
  • Alzheimer's disease exhibits neuropathological abnormalities in the olfactory system located in the nasal cavity. These include the presence of dystrophic neurites that exhibit immunoreactivity for tau, neurofilaments, apolipoprotein E and other proteins, abnormal tau protein, increase in superoxide dismutase, and beta-amyloid deposition in the primary sensory (olfactory receptor) cells and nerve fibres of the nasal mucosa tissue (Arnold et al., Ann N Y Acad Sci 1998 Nov 30;855:762-75; Hock et al., Eur Neurol 1998 Jul;40(1):31-6; Johnson et al., Neurobiol Aging 1994 Nov- Dec;15(6):675-80; Kulkami-Narla et al., Exp Neurol 1996 Aug; 140(2): 115-25; Lee et al., Exp Neurol 1993 May;121(1 ):93-105; Tabat
  • AD patients e.g., central nervous system neurons from AD patients.
  • Routine access to these sensory neurons and fibers can be done with nasal biopsy in AD patients (e.g., Feron et al., Arch Otolaryngol Head Neck Surg 1998 Aug;124(8):861-6).
  • Olfactory neuroblasts olfactory neurons obtained by biopsy and placed in primary cell culture
  • AD patients produce carboxy terminal amyloid precursor protein (APP) fragments that contain beta-amyloid (A-beta)(Crino et al., Ann Otol Rhinol Laryngol 1995 Aug;104(8):655-61).
  • Crino et al. showed labeling of A-beta in the basal third of the olfactory neuroepithelium and in axons projecting through the lamina intestinal of AD patients.
  • Thioflavin-S staining that detects amyloid deposition was also observed in the basal third of the olfactory neuroepithelium from AD patients.
  • Alpha 7 nicotinic acetylcholine receptors are present in olfactory neurons probably including olfactory receptor cells in the nasal cavity (Alkondon et al., Neurosci Lett 1994 Aug 1;176(2): 152- 6; Alkondon et al., Eur J Neurosci 1997 Dec;9(12):2734-42; Bouvet et al., Neurosci Res 1988 Feb;5(3):214-23; Edwards et al., Experientia 1987 Aug 15;43(8):868-73; Edwards et al., Experientia 1988 Mar 15;44(3):208-11; Seguela et al., J Neurosci 1993 Feb;13(2):596-604).
  • Beta-amyloid peptide increases cytosolic-free Ca 2+ in AD lymphoblasts (Ibarreta et al., Alzheimer Dis Assoc Disord 1997 Dec;11(4):220-7), and elevates mitogen-induced Ca 2+ responses in freshly prepared human lymphocytes (Eckert et al., Life Sci 1994;55(25-26):2019-29).
  • Amyloid precursor protein can be induced on the cell surface of human lymphocytes upon stimulation (Bullido et al., Biochim Biophys Acta 1996 Aug 21 ;1313(1 ):54-62) and increased APP-770 isoform occurs in lymphocytes from AD patients (Ebstein et al., Brain Res Mol Brain Res 1996 Jan;35(1-2):260-8). Lymphoblastoid cells from patients with early-onset and late-onset familial AD show increased expression of beta-APP mRNA and protein (Matsumoto et al., Eur J Biochem 1993 Oct 1 ;217(1 ):21-7). Lymphocytes from AD patients also exhibit an increased mRNA level for alpha 7 nicotinic receptor (Hellstrom- Lindahl et al., Brain Res Mol Brain Res 1999 Mar 20;66(1-2):94-103)
  • AD alpha 7 nicotinic acetylcholine receptor - beta amyloid peptides interaction in circulating blood cells and olfactory neuroepithelial neurons/neuronal processes or olfactory neuroblasts obtained from AD patients could be used as AD diagnostic tools, markers of AD progression and prognosis, and markers of therapeutic efficacy for any intervention or treatment targeting AD.
  • the present invention provides methods for diagnosing
  • Alzheimer's disease monitoring the progression and prognosis of Alzheimer's disease and/or monitoring the therapeutic efficacy of any intervention or treatment of Alzheimer's disease comprising:
  • test sample comprises circulating blood cells and/or olfactory neuroepithelial neuronal cell bodies or their neuronal processes (i.e., dendrites and axon of a nueron); and
  • amyloid beta peptide including, but not limited to, A ⁇ 1-40 , A ⁇ 1-42 and A ⁇ - 3 peptides and their fragments
  • alpha-7 nicotinic acetylcholine receptors alpha-7 nicotinic acetylcholine receptors
  • BSA bovine serum albumin
  • nAChR nicotinic acetylcholine receptor
  • Ph phenyl
  • PCC pyridinium chlorochromate TEA triethyl amine THF tetrahydrofuran TLC thin layer chromatography
  • This material can then be subjected to a reductive amination reaction such as with propionaldehyde as shown to yield (8).
  • Compound (8) is then treated with HBr in acetic acid to cleave the methyl ethers and form dihydroxy compound (9).
  • BBr 3 may be used to cleave the methyl ethers and form dihydroxy compound (9).
  • a deficiency of BBr 3 may be used to provide compounds in which only one of the methyl ethers has been removed to afford compounds with one hydroxy and one methoxy group.
  • a further means of preparing compounds of the present invention is illustrated in Scheme 2.
  • This material is then subjected to an aminomercuration reaction to afford a 2- amino-1 ,4-butadiene such as compound (12).
  • Diels-Alder reaction of compound (12) with benzophenone (1) gives (13) which can be isomerized as in Scheme 1 with base to give the 1 ,5-dihyroxy compound (14).
  • Compounds of type 9 can be treated with a base such as triethylamine in a suitable solvent such as dioxane or methylene chloride along with electrophiles such as acid halides, phosphinyl halides, or alkyl halides to give the products of substitution on one or both of the phenolic hydroxyls.
  • a base such as triethylamine
  • a suitable solvent such as dioxane or methylene chloride
  • electrophiles such as acid halides, phosphinyl halides, or alkyl halides
  • Fraction 1 (0.306g, 1.0 mmol) and fumaric acid (0.141 g, 1.2 mmol) were dissolved in ethanol (2 mL) with heating. The ethanol was evaporated and the residue triturated with diethyl ether affording the fumarate salt of 18 as a white solid. Similarly obtained from Fraction 2 (0.301 g, 1.0 mmol) and fumaric acid (0.141 g, 1.2 mmol) was the fumarate sale of 17.
  • Ketone 6 (0.299 g, 1.36 mmol) was dissolved in toluene (1 mL) followed by the addition of morphoiine (0.130 g, 0.13 mL, 1.5 mmol) and Ti(/-PrO) (0.611 g, 0.64 mL, 2.15 mmol). The reaction mixture was stirred for 15 hrs at room temperature. Methanol (2 mL) was added followed by portionwise addition of NaBH 4 (0.15 g, 3.9 mmol) over 1 hr. Dichloromethane and 1N
  • Ketone 6 (1 g, 4.5 mmol) was dissolved in acetonitrile (75 mL) followed by the addition of acetic acid (0.54 mL, 9 mmol, 2 eq.) and propargylamine (0.6 mL, 9 mmol, 2 eq.). The reaction mixture was stirred at room temperature for 1 hr. Sodium cyanoborohydride (0.6 g, 9 mmol, 2 eq.) was added portionwise (3x) over 1.5 hrs (every 30 min), and the reaction mixture was stirred overnight at room temperature. Ethyl acetate and 1 N NaOH were added with thorough mixing and the organic layer was separated, dried with MgS0 4 , filtered and evaporated to an oil.
  • acetic acid (0.54 mL, 9 mmol, 2 eq.
  • propargylamine 0.6 mL, 9 mmol, 2 eq.
  • the product was purified with flash chromatography (50:50/ethyl acetate:hexane) to yieid an oil of 5,8-dimethoxy-c/s and trans-2-N- (propargyl)amino-3-methyl-1 ,2,3,4-tetrahydronaphthalene (1.2 g, 100%).
  • This material was dissolved in acetonitrile along with propionaldehyde (0.65 g, 9 mmol, 2 eq.) and acetic acid (0.5 mL, 9 mmol) and stirring for 1 hr at room temperature.
  • Example 11 As a specific embodiment of an oral composition, 100 mg of Compound 9 from Example 1 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size O hard gel capsule.
  • 100 mg of Compound 9 from Example 1 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size O hard gel capsule.
  • Example 11
  • a ⁇ 1-42 and A ⁇ _4 0 binding to alpha-7 nAChRs can be demonstrated in competitive binding experiments with ⁇ -bungarotoxin
  • SK-N-MC cell membranes were incubated with 0.5 nM of 125 l- ⁇ - bungarotoxin in the presence of various concentrations of A ⁇ _ 2 and A ⁇ 1-40 at 25°C for 1 hr. The assay mixture was then rapidly filtered and the radioactivity that was retained on the filter was determined.
  • a ⁇ 1- 2 binding to alpha-7 nAChRs is supported by competitive binding experiments with 125 I-A ⁇ _ 40 and provides a seed for amyloid deposition and, thus, incipient plaque formation
  • Alpha-7 nAChRs were immobilized on wheat germ agglutinin coupled yittrium SPA beads and allowed to incubate with 125 l-A ⁇ 1-40 in the presence of various concentrations of cold A ⁇ or A ⁇ 1-42 .
  • the data showed that 0.1 femtoM of cold A ⁇ - 42 completely abolished the binding, suggesting that A ⁇ -42 interacted with alpha-7 nAChRs with high affinity.
  • 10 nM of cold to alpha-7 nAChRs was dramatically increased.
  • a ⁇ _42 binding to alpha-7 nAChRs is of higher affinity than A ⁇ o binding to alpha-7 nAChRs
  • Alpha-7 nAChRs were allowed to interact with or A ⁇ 1-42 and the mixture was then immunoprecipitated with anti-alpha-7 nAChR antibodies.
  • Western analyses of the immunoprecipitated proteins identified only the presence of A ⁇ 1-42 , indicating that the A ⁇ 1- 2 /alpha-7 nAChR interaction is robust and of high affinity. Since it has already been shown that A ⁇ 1-40 binds to the alpha-7 receptor, the failure in detecting in this co-precipitation experiment suggests that the A ⁇ 1- 0 /alpha-7 nAChR interaction is of lower affinity than the A ⁇ 1-42 /alpha-7 nAChR interaction.
  • a ⁇ s are known to form aggregates leading to the formation of amyloid plaques that are characteristic of Alzheimer's disease. This phenomenon can be demonstrated in vitro by using Synthaloid plates, coated with A ⁇ crystallization centers, and labelled A ⁇ s for detecting aggregation. We have validated this aggregation assay using both 125 l-A ⁇ 1-40 and in a buffer containing 50 mM HEPES, pH 7.4, 0.1% BSA, 10% FCS and protease inhibitors.
  • Compounds which inhibit A ⁇ aggregation with an IC 50 ⁇ 100 micromolar may be effective in inhibiting the binding of A ⁇ to alpha-7 in such a manner as to have a positive therapeutic effect useful for the treatment of neurodegenerative disorders.
  • Synaptosomes from guinea pig hippocampus were incubated with 0.1 ⁇ M 3 H-choline and then subjected to repeated washes to remove unincorporated 3 H-choline.
  • the synaptosomes were treated with 65 mM K + for 30 seconds to elicit 3H-acetylcholine release. While A ⁇ o and A ⁇ z at 100 pM both inhibited acetylcholine release from these preparations (33% inhibition for both A ⁇ - 0 and A ⁇ - 42 ) .
  • pretreatment of the synaptosomes with both 10 nM of Compound 9 and 100 pM of either stimulation was found to have no effect on acetylcholine release.
  • alpha-7 nAChR contained SK-N-MC cell membranes were immobilized on wheat germ agglutinin coupled yittrium SPA beads and allowed to incubate with 25 l-A ⁇ 1-40 in the presence of various concentrations of compound 9.
  • the result demonstrated that Compound 9 efficiently inhibited the binding of 125 l- SK-N-MC cells with a 300 pM IC50.
  • Compounds which inhibit the binding of A ⁇ to alpha-7 nAChRs with an IC50 ⁇ 1 micromolar may have a positive therapeutic effect useful for the treatment of neurodegenerative disorders.
  • a 2-5 ug quantity of the 11 amino acid peptide comprised of the 206- 216 stretch of the ⁇ 7 nAChR (N-Ac, C(O)NH 2 ) are added to a 96-well microtiter plate in 50 mL 10mM HEPES, pH 7.4, or any buffer 50ul.
  • 125 l- ⁇ -amyloidi-40 (2000Ci/mmol, 50pM) was added to the wells in the presence and absence of inhibitors (1 uM to 10 uM) dissolved in 1uL of 30% or 100% DMSO. Unbound ligands were removed and bound radioactivity was measured using a Microbeta liquid scintillation counter. Ligand binding can be inhibited by ⁇ - bungarotoxin, the peptide itself, and Compound 9.

Abstract

The invention is directed to a method of treating a neurodegenerative disorder in a subject in need thereof which comprises administering to the subject an amount of a compound effective to inhibit the interaction of amyloid-beta with alpha-7 nicotinic acetylcholine receptors.

Description

Title of the Invention
Method for Treating Neurodegenerative Disorders
Cross-Reference to Related Application
This application claims priority from United States provisional application Serial No. 60/087,577, filed June 1 , 1998, the contents of which are hereby incorporated by reference.
Field of the Invention
The present invention provides a method for treating neurodegenerative disorders. More particularly, a method for treating neurodegenerative disorders (e.g., Alzheimer's disease) by inhibiting the interaction of amyloid beta with alpha- 7 nicotinic acetylcholine receptors.
Background of the Invention
Neurodegenerative disorders such as Alzheimer's disease (AD) and
Parkinson's disease (PD) afflict humanity with great suffering and financial loss. AD is characterized by neurofibrillary tangles, neu tic plaques, and neuronal cell death. AD appears as either the familial, early onset (<60 yrs) or late-onset (>60 yrs) forms, with the latter being more prevalent. AD is the major cause of age-related dementia and cognitive impairment
(Wisniewski, T.; Ghiso, J.; Frangione, B. Neυrobiol. of Disease 1997, 4, 313-328). The amyloid precursor protein (APP), β-amyloidι-40
Figure imgf000003_0001
and
Figure imgf000003_0002
(Aβι- 2) are keenly involved in the pathology of AD. The Aβ peptides are derived from APP by proteolytic processing. Dramatic evidence implicating the Aβ peptides, particularly Aβ1-42, in AD comes from various recently identified mutations accounting for certain types of inherited AD. Such mutations in the presenilin (PS1 and PS2) genes are probably the cause of the most frequent form of familial, early-onset AD (Rogaev, E. I. Molecular Biology 1998, 32, 58). In these cases, as with APP mutations, more
Figure imgf000004_0001
is observed relative to Aβ1-40. Extensive studies have shown that Aβι-42 has a greater ability than Aβ1-40 to aggregate into the amyloid fibrils that constitute the plaques characteristic of AD (Lansbury, P. T., Jr. Accts. Chem. Res. 1996, 29, 317). Even though
Figure imgf000004_0002
is generally present to a much larger degree in the cerebrospinal fluid than Aβ1-42, it is Aβι^2 which is the major Aβ peptide found in AD plaques.
The Aβ peptides can inhibit cholinergic neurotransmitter function independent of neurotoxicity (Auld, D. S.; Kar, S.; Quirion, R. Trends Neurosci. 1998, 21, 43). Aβ peptides bind to a number of natural substances such as apoE3, apoE4, apoJ, transthyretin, and albumin. In addition, Aβ has been reported to interact with a membrane-bound receptor for advanced glycation end products and to the class A scavenger receptor (SR) associated with the production of reactive oxygen species. Stimulation of the alpha-7 subtype of the nicotinic acetylcholine receptors (nAChRs) can protect neurons against Aβ cytotoxicity (Kihara, T. et al. Ann. Neurol. 1997, 42, 159). Also, a set of compounds that activate nAChRs, especially of the alpha-7 subtype, have been found to have in vivo activity in models of cognition enhancement (US Patent No. 5,741 ,802, issued April 21 , 1998).
We now describe specific binding of Aβ1- 0 and Aβι-42 to the alpha-7 subtype of nAChRs. This new finding has broad ramifications for the etiology and treatment of AD. nAChRs are members of the ligand-gated ion channel family and appear to be formed from five protein subunits associating together around a central pore (Lindstrom, J. Molecular Neurobiology 1997 ', 15, 193). These subunits include α1-α9, β1-β4, γ, δ, and ε. The α.7 subtype forms functional homomers which bind to α- bungarotoxin, a 75-amino acid peptide, with high affinity (0.65-1.7 nM Kd) and nicotine with relatively low affinity (ca. micromolar Kd) (Holladay, M. W.; Dart, M. J.; Lynch, J. K. J. Med. Chem. 1997, 40, 4169).
Compounds which block the aggregation of Aβ peptides are potentially useful drugs for the treatment of AD. For example, rifampicin inhibits Aβ aggregation and neurotoxicity and may show an effect in vivo in diminishing plaque burden when compared with age-matched controls (Tomiyama, T. et al. J. Biol. Chem. 1996, 277, 6839). In order to block the interaction of the Aβ peptides with α7 nAChRs, compounds can be found to either bind to α7 nAChRs, to Aβ itself, or to both. Any of these mechanisms of action would be expected to provide significant protection against Aβ-mediated neurotoxicity and inhibition of cholinergic functioning mediated by nAChRs and be extremely useful for the treatment of AD. The binding of Aβ1-42 to alpha-7 nAChRs provides a seed for crystallization or deposition of Aβ into insoluble deposits, which have the potential to grow into the fibrillar amyloid deposits characteristic of AD. Therefore, blocking the interaction of Aβ1- 2 with alpha-7 nAChRs should reduce the amount of insoluble aggregated Aβ that is formed, and thus prevent the neurotoxicity and pathology associated with such aggregated amyloid deposits.
Accordingly, it is an object of the invention to provide a method for treating neurodegenerative disorders by inhibiting the binding of amyloid beta peptides to alpha-7 nicotinic acetylcholine receptors. It is a further object of the invention to provide a method for treating Alzheimer's disease and or for slowing the progression of Alzheimer's disease by inhibiting the binding of amyloid beta peptides to alpha-7 nicotinic acetylcholine receptors. Another object of the invention is to provide a predictive method, a method for diagnosis, a method to monitor prognosis, a method to monitor the progression, and a method to monitor the therapeutic efficacy for any therapeutic intervention used in Alzheimer's disease. Still another object of the invention is to provide a method for identifying compounds which inhibit the binding of Aβ peptides with α7 nAChRs, either by binding to Aβ peptides or to α7 nAChRs. Summary of the Invention
The present invention is directed to a method of treating a neurodegenerative disorder in a subject (preferably, a human) in need thereof which comprises administering to the subject an amount of a compound effective to inhibit the binding of an amyloid beta peptide, preferably Aβ1-42 , to alpha-7 nAChRs, preferably, human alpha-7 nAChRs. Since alpha-8 and alpha-9 nAChRs are similar with respect to structure and function to alpha-7 nAChRs, it is possible that blocking the interaction of β-amyloid with alpha-8 and alpha-9 nAChRs would have therapeutic benefit as well.
Neurodegenerative disorders included within the methods of the present invention include, but are not limited to, Alzheimer's disease, Pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson's-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette's disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy's disease), primary lateral sclerosis, familial spastic paraplegia, Werdnig- Hoffmann disease, Kugelberg-Welander disease, Tay-Sach's disease, Sandhoff disease, familial spastic disease, Wohlfart-Kugelberg-Welander disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt-Jakob, Gerstmann-Straussler-Scheinker disease, Kuru and fatal familial insomnia).
Other conditions also included within the methods of the present invention include age-related dementia and other dementias and conditions with memory loss including vascular dementia, diffuse white matter disease (Binswanger's disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica and frontal lobe dementia. Also other neurodegenerative disorders resulting from cerebral ischemia or infaction including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type (including, but not limited to, epidural, subdural, subarachnoid and intracerebral), and intracranial and intravertebral lesions (including, but not limited to, contusion, penetration, shear, compression and laceration).
Preferably, the neurodegenerative disorder is selected from Alzheimer's disease, Parkinson's disease, Tourette's syndrome, amyotrophic lateral sclerosis, age-related memory loss, senility and age-related dementia, most preferably, the neurodegenerative disorder is Alzheimer's disease. Because, most preferably, the neurodegenerative disorder is Alzheimer's disease, also.defmed as an amyloidosis, other conditions within the methods of the present invention include other amyloidosis which share features including, but not limited to, hereditary cerebral angiopathy, nonneuropathic hereditary amyloid, Down's syndrome, macroglobulinemia, secondary familial Mediterranean fever, Muckle-Wells syndrome, multiple myeloma, pancreatic- and cardiac-related amyloidosis, chronic hemodialysis arthropathy, and Finnish and Iowa amyloidosis.
In one embodiment of the invention is a method of treating and/or preventing dementia in an Alzheimer's patient (as well as a method for treating and/or preventing other clinical manifestations of Alzheimer's disease that include, but are not limited to, cognitive and language deficits, apraxias, depression, delusions and other neuropsychiatric symptoms and signs, and movement and gait abnormalities) which comprises administering to the subject a therapeutically effective amount of a compound to inhibit the binding of an amyloid beta peptide (preferably, Aβ!-42) with nAChRs, preferable alpha-7 nAChRs, most preferably, human alpha-7 nAChRs. In a second embodiment of the invention is a method of improving memory and/or mental status and/or of halting the progression of mental deterioration in an Alzheimer's disease patient which comprises administering to the subject a therapeutically effective amount of a compound to inhibit the binding of an amyloid beta peptide (preferably, Aβι-42 ) with nAChRs, preferably alpha-7 nAChRs, most preferably, human alpha-7 nAChRs.
Preferably, the compound used in the methods of treating neurodegenerative disorders, treating and/or preventing Alzheimer's disease, and improving memory and/or halting the progression of mental deterioration in an Alzheimer's disease patient is not estrogen, raloxifene, droloxifene, tamoxifen, idoxifene or levomeloxifene; more preferably, the compound is not estrogen or a selective estrogen receptor modulator (SERM). A SERM is an estrogen receptor ligand that exhibits estrogen agonist activity in the cardiovascular system, CNS and bone, and estrogen antagonist activity in reproductive tissues, such as breast and uterus.
Also included in the invention is the use of a compound which inhibits the binding of an amyloid beta peptide (preferably
Figure imgf000008_0001
to an alpha-7 nAChR (preferably, a human alpha-7 nAChR) in the preparation of a medicament for the treatment of a neurodegenerative disorder in a subject (preferably, a human) in need thereof.
Another illustration of the invention is the use of a compound which inhibits the binding of an amyloid beta peptide (preferably Aβι- 2) to alpha-7 nAChRs (preferably, human alpha-7 nAChRs) in the preparation of a medicament for: a) improving memory, b) halting the progression of the mental deterioration seen in Alzheimer's disease patients, c) treating dementia, d) preventing dementia in an Alzheimer's patient, and e) treating and/or preventing other clinical manifestations of Alzheimer's disease that include, but are not limited to, cognitive and language deficits, apraxias, depression, delusions and other neuropsychiatric symptoms and signs, and movement and gait abnormalities in an Alzheimer's patient.
In another aspect of the invention is a compound of the formula I:
Figure imgf000009_0001
wherein Ri is hydrogen or Cι-C4 alkyl;
R is selected from hydrogen, d-Cβ alkyl, aryl or C7-Cι0 aralkyl; R3 is selected from hydrogen, Cι-C6 alkyl, C3-C10 alkenyl,
C3-C8 cycloalkylCι-C6 alkyl, Cι-C6 alkoxycarbonylCι-C6 alkyl, Cι-C6 alkylthio, heteroarylCι-C4 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted C7-C10 aralkyl wherein the substituent on the aryl or aralkyl are one or more substituents independently selected from the group consisting of halogen, hydroxy, Ci-Ce alkyl and unsubstituted or substituted Cι-C6 alkoxy wherein the substituents on the alkoxy are one or more substituents independently selected from amino, Cι-C6 alkylamino, Cι-C6 dialkylamino, pyrrolidinyl, piperidinyl, azepinyl or morpholinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
R is C C6 alkyl, aryl, or C7-Cι0 aralkyl; and Rsand R6 are each independently selected from hydrogen, Cι-C6 alkyl, C3-C10 alkenyl, Cι-C8 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
In preferred compounds of formula I, Ri is hydrogen; R2 is selected from hydrogen or Cι-C4 alkyl;
R3 is selected from Cι-C alkyl, C3-C10 alkenyl, C5-C6 cycloalkylCι-C46 alkyl, d-Ce alkoxycarbonylCι-C4 alkyl, Cι-C6 alkylthio, heteroaryiCι-C4 alkyl, or unsubstituted or substituted C7-Cιo aralkyl wherein the substituent on the aralkyl are one or two substituents independently selected from the group consisting of halogen, hydroxy, Cι-C4 alkyl and unsubstituted or substituted Cι-C alkoxy wherein the substituents on the alkoxy are one or two substituents independently selected from amino, Cι-C alkylamino, Cι-C4 dialkylamino, pyrrolidinyl, or piperidinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a morpholinyl ring;
R4 is Cι-C4 alkyl; and
R5and R6 are each independently selected from hydrogen, C1-C alkyl, C3-C6 alkenyl, Cι-C6 alkylcarbonyl, or diphenylphosphinyl.
In a subclass of compounds of formula I are compounds having the formula
Figure imgf000010_0001
wherein Ri is hydrogen or C-i-C4 alkyl;
R2 and R3 are each independently selected from hydrogen, Ci-Ce alkyl, aryl or C7-Cιo aralkyl; and l^ is Cι-C6 alkyl, aryl, or C7-C10 aralkyl; and pharmaceutically acceptable salts thereof.
Compounds of formula I are novel compounds which block the interaction of beta-amyloid with alpha-7 nAChRs. More specifically, the compounds of formula I inhibit the binding of
Figure imgf000010_0002
human alpha-7 nAChRs by binding to Aβι-42. The orientation between the nitrogen atom and R.1 on the appropriate ring can be either cis or trans. Preferably, the compound is 5,8-dihydroxy-frans-2-di(Λ/-propylamino)-3-methyl-1 ,2,3,4- tetrahydronaphthalene, and pharmaceutically acceptable salts thereof.
Other compounds useful in the methods of the present invention inhibit the binding of Aβι-42 with human alpha-7 nAChRs by binding to human alpha- 7 nAChRs directly. An example of such a compound which binds to human alpha-7 nAChRs is α-bungarotoxin .
Detailed Description of the Invention
The present invention provides methods of treating neurodegenerative disorders by inhibiting the binding of amyloid beta peptides to alpha-7 nAChRs. Neurodegenerative disorders included within the methods of the present invention include, but are not limited to, Alzheimer's disease, Pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson's-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette's disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy's disease), primary lateral sclerosis, familial spastic paraplegia, Werdnig- Hoffmann disease, Kugelberg-Welander disease, Tay-Sach's disease, Sandhoff disease, familial spastic disease, Wohlfart-Kugelberg-Welander disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt-Jakob, Gerstmann-Straussler-Scheinker disease, Kuru and fatal familial insomnia). Other conditions also included within the methods of the present invention include age-related dementia and other dementias and conditions with memory loss including vascular dementia, diffuse white matter disease (Binswanger's disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica and frontal lobe dementia. Also other neurodegenerative disorders resulting from cerebral ischemia or infaction including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type (including, but not limited to, epidural, subdural, subarachnoid and intracerebral), and intracranial and intravertebral lesions (including, but not limited to, contusion, penetration, shear, compression and laceration).
Preferably, the neurodegenerative disorder is selected from Alzheimer's disease, Parkinson's disease, Tourette's syndrome, amyotrophic lateral sclerosis, age-related memory loss, senility and age-related dementia, most preferably, the neurodegenerative disorder is Alzheimer's disease. Because, most preferably, the neurodegenerative disorder is Alzheimer's disease, also.defined as an amyloidosis, other conditions within the methods of the present invention include other amyloidosis which share features including, but not limited to, hereditary cerebral angiopathy, nonneuropathic hereditary amyloid, Down's syndrome, macroglobulinemia, secondary familial Mediterranean fever, Muckle-Wells syndrome, multiple myeloma, pancreatic- and cardiac-related amyloidosis, chronic hemodialysis arthropathy, and Finnish and Iowa amyloidosis.
The terms "amyloid beta", "amyloid beta peptide" or "beta-amyloid" as used herein, refer to amyloid beta peptides and include the Aβι-40,
Figure imgf000012_0001
peptides and their fragments. Examples of fragments of amyloid beta peptides that have been shown to have biological activity and are useful in the methods of the present invention include, but not limited to, fragment 1-28, and fragment 25-35 (e.g., Yatin SM, Aksenov M, Butterfield DA. Neurochem Res 1999 Mar;24(3):427-35; Hirakura Y, Satoh Y, Hirashima N, Suzuki T, Kagan BL, Kirino Y. Biochem Mol Biol Int 1998 Nov;46(4):787-94; Mazziotti M, Perlmutter DH. Biochem J 1998 Jun 1 ;332 ( Pt 2):517-24; Perovic S, Bohm M, Meesters E, Meinhardt A, Pergande G, Muller WE. Mech Ageing Dev 1998 Mar 16;101(1-2):1-19; Muller WE, Eckert GP, Scheuer K, Cairns NJ, Maras A, Gattaz WF. Amyloid 1998 Mar;5(1):10-5; Butterfield DA, Martin L, Carney JM, Hensley K. Life Sci 1996; 58(3):217-28; Forioni G, Lucca E, Angeretti N, Delia Torre P, Salmona M. J Neurochem 1997 Nov;69(5):2048-54; Heese K, Hock C, Otten U. J Neurochem 1998 Feb;70(2):699-707; Blanchard BJ, Konopka G, Russell M, Ingram VM. Brain Res 1997 Nov 21;776(1-2):40-50; Wu A, Derrico CA, Hatem L, Colvin RA. Neuroscience 1997 Oct;80(3):675-84; Muller WE, Romero FJ, Perovic S, Pergande G, Pialoglou P. J Neurochem 1997
Jun;68(6):2371-7; Suh YH. J Neurochem 1997 May;68(5):1781-91 ; Parpura- Gill A, Beitz D, Uemura E. Brain Res 1997 Apr 18;754(1-2):65-71; Fletcher TG, Keire DA. Protein Sci 1997 Mar;6(3):666-75; Scorzieilo A, Meucci O, Calvani M, Schettini G. Neurochem Res 1997 Mar;22(3):257-65); Miguel-Hidalgo JJ, Vecino B, Femandez-Novoa L, Alvarez A, Cacabelos R. Eur
Neuropsychopharmacol 1998 Aug;8(3):203-8; Maneiro E, Lombardi VR, Lagares R, Cacabelos R. Methods Find Exp Clin Pharmacol 1997 Jan- Feb;19(1):5-12).
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 or disorder being treated.
The term "alkyl" shall mean straight or branched chain alkanes of one to ten carbon atoms, or any number within this range. For example, alkyl radicals include, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, 3-(2-methyl)butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl and 2-methylpentyl. Alkoxy radicals are oxygen ethers formed from the previously described straight or branched chain alkyl groups. Cycloalkyl groups contain 3 to 8 ring carbons and preferably 5 to 7 carbons. Similarly, alkenyl and alkynyi groups include straight and branched chain alkenes and aikynes having 2 to 10 carbon atoms, or any number within this range.
The term "aryl" indicates aromatic groups such as phenyl and naphthyl.
The term "C7-C10 aralkyl" means an alkyl group substituted with an aryl group wherein the total number of carbon atoms is between 7 and 10 (e.g., benzyl, phenylethyl, phenylpropyl).
The term "heteroaryl" as used herein represents an unsubstituted or substituted stable five or six membered monocyclic aromatic ring system or an unsubstituted or substituted nine or ten membered benzo-fused heteroaromatic ring system or bicyclic heteroaromatic ring system which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heteroaryl group may be attached at any heteroatom or carbon atom that results in the creation of a stable structure. Examples of heteroaryl groups include, but are not limited to pyridyl, pyridazinyl, thienyl, furanyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzoxazolyl, benzopyrazolyl, indolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl adeninyl or quinolinyl.
Prefered heteroaryl groups include pyridyl, pyrrolyl, pyrazinyl, thiadiazolyl, pyrazolyl, thienyl, triazolyl and quinolinyl.
The term "N(CH2)5" means a piperidinyl group.
The term "cC6Hn" refers to a cyclohexyl group. When a particular group is "substituted" (e.g., aryl, aralkyl), that group may have one or more substituents, preferably from one to five substituents, more preferably from one to three substituents, most preferably from one to two substituents, independently selected from the list of substituents.
Under standard nomenclature used throughout this disclosure, the terminal portion of the designated side chain is described first, followed by the adjacent functionality toward the point of attachment. Thus, for example, a "phenyld-C6 alkylamidoCι-C6alkyl" substituent refers to a group of the formula
Figure imgf000015_0001
Compounds which are useful in the methods of the present invention for inhibiting the interaction of Aβι-40 and Aβι-42 to the alpha-7 subtype of nAChRs for either the purpose of direct therapeutic intervention or in order to screen for compounds which act via this mechanism include compounds of formula I, especially 5, 8-dihydroxy-fra/?s-2-di(Λ/-propylamino)-3-methyl-1 , 2,3,4- tetrahydronaphthalene (Compound 9), (-)-nicotine, (rac)-epibatidine, α- bungarotoxin, and pharmaceutically acceptable salts thereof.
Certain peptide stretches of the human alpha-7 nAChR bind to amyloid beta and can be used in place of the alpha-7 nAChR together with amyloid beta for the purpose of screening libraries to find compounds which block the interaction of amyloid beta and the human alpha-7 nAChR. Included among these peptide stretches of the human alpha-7 nAChR are alpha-7 nAChRI 93- 224 and smaller peptides derived thereof as listed in the Table. Table
Compound Amelioration of the Aβ1- 2 Meditated Inhibition of ACh
Release in Rat Cortical Synaptosomes (%) human alpha-7 81 % at 10 μM nAChRI 93-224 79% at 1 μM
AC-NGEWDLVGIPGKRSERFYECCKEPYPDVTFTV-NH2 human alpha-7 79% at 10 μM nAChR200-214 71 % at 1 μM
AC-GIPGKRSERFYECCK-NH2 human alpha-7 81 % at 10 μM nAChR206-216 77% at 1 μM
AC-SERFYECCKEP-NH2 human alpha-7 84% at 10 μM nAChR206-216 82% at 1 μM oxidized (cyclic) CC:
AC-SERFYECCKEP-NH2
human alpha-7 22% at 10 μM nAChR206-216 10% at 1 μM SERFYECCKEP human alpha-7 72% at 10 μM nAChR210-213 59% at 1 μM
AC-YECC-NH2
The standard one-letter code for the amino acids has been employed for the compounds. This code is listed in Lehninger, A. I. "Biochemistry" Second Edition, Worth Publishers, Inc., New York, 1976, p 73-75.
For use in medicine, the salts of the compounds of this invention refer to non-toxic "pharmaceutically acceptable salts." Other salts may, however, be useful in the preparation of compounds according to this invention or of their pharmaceutically acceptable salts. 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 with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. Furthermore, where the compounds of 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., quaternary ammonium salts.
The present invention includes within its scope prodrugs of the compounds of this invention. A prodrug is inactive as administered, but becomes activated in vivo. The prodrug is converted to the parent drug chemically or by specific enzyme(s). Higuchi, T.; Stella, V., Eds. "Pro-Drugs as Novel Drug Delivery Systems"; American Chemical Society: Washington, DC, 1976. In general, such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. (-)-Nicotine is (-)-1-methyl-2-(3-pyridinyl)pyrrolidine and is readily available from Sigma Chemical Company.
Figure imgf000018_0001
(-)-Nicotine
(+/-)-Epibatidine is exo-(+/-)-2-(6-chloro-3-pyridinyl)-7- azabicyclo[2.2.1]heptane and is readily available from Sigma Chemical Company.
Figure imgf000018_0002
(rac)-Epibatidine
α-Bungarotoxin is a 74 amino acid peptide which is commercially available from Research Biochemicals Inc. α-Bungarotoxin and its amino acid sequence are described in Lee, C. Y. Annu. Rev. Pharmacol. 1972, 12, 265- 281.
125, -Aβι- 0,
Figure imgf000018_0003
and anti-alpha-7 nAChR antibodies are commercially available Amersham Pharmacia Biotech, Advanced Bioconcepts and Research Biochemicals International, respectively.
125l-α-bungarotoxin is commercially available from Amersham Pharmacia Biotech. The present invention therefore provides a method of treating a neurodegenerative disorder, which comprises administering any of the compounds as defined herein in a quantity effective to treat the neurodegenerative disorder. Preferably, the compound is not estrogen, raloxifene, droloxifene, tamoxifen, idoxifene or levomeloxifene; more preferably, the compound is not estrogen or a selective estrogen receptor modulator (SERM). The compound may be administered to a patient afflicted with a neurodegenerative disorder by any conventional route of administration, including, but not limited to, intravenous, oral, subcutaneous, intramuscular, intradermal, buccal, intracerebral and other parenteral routes. The quantity of the compound which is effective for treating a neurodegenerative disorder is between 0.01 mg per kg and 10 mg per kg of subject body weight.
The method of treating neurodegenerative disorders described in the present invention may also be carried out using a pharmaceutical composition comprising any of the compounds as defined herein and a pharmaceutically acceptable carrier. The pharmaceutical composition may contain between about 0.5 mg and 200 mg of the compound, and may be constituted into any form suitable for the mode of administration selected. Carriers include necessary and inert pharmaceutical excipients, including, but not limited to, binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, dyes, and coatings. Compositions suitable for oral administration include solid forms, such as pills, capsules, granules, tablets, caplets, and powders, and liquid forms, such as solutions, syrups, elixers, and suspensions. Forms useful for intracerebral and other parenteral routes of administration include sterile solutions, emulsions and suspensions.
Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular compound used, the mode of administration, the strength of the preparation, the mode of administration, and the advancement of the disease condition. In addition, factors associated with the particular patient being treated, including patient age, body weight, diet, physical activity and time of administration, and associated co-morbidities and clinical conditions will result in the need to adjust dosages.
The present invention also provides diagnostic tools useful for diagnosing Alzheimer's disease. Alzheimer's disease (AD) exhibits neuropathological abnormalities in the olfactory system located in the nasal cavity. These include the presence of dystrophic neurites that exhibit immunoreactivity for tau, neurofilaments, apolipoprotein E and other proteins, abnormal tau protein, increase in superoxide dismutase, and beta-amyloid deposition in the primary sensory (olfactory receptor) cells and nerve fibres of the nasal mucosa tissue (Arnold et al., Ann N Y Acad Sci 1998 Nov 30;855:762-75; Hock et al., Eur Neurol 1998 Jul;40(1):31-6; Johnson et al., Neurobiol Aging 1994 Nov- Dec;15(6):675-80; Kulkami-Narla et al., Exp Neurol 1996 Aug; 140(2): 115-25; Lee et al., Exp Neurol 1993 May;121(1 ):93-105; Tabaton et al., Neurology 1991 Mar;41(3):391-4; Talamo et al., Ann N Y Acad Sci 1991;640:1-7; Yamagishi et al., Ann Otol Rhinol Laryngol 1998 May; 107(5 Pt 1 ):421-6; Yamagishi et al., Nippon Jibiinkoka Gakkai Kaiho 1994 Jan;97(1 ):51-60). These observations recapitulate the neuropathological profile and neurodegenerative abnormalities (e.g., cytoskeletal changes, protein immunoreactivity and beta-amyloid deposition) observed in central nervous system neurons from AD patients. Routine access to these sensory neurons and fibers can be done with nasal biopsy in AD patients (e.g., Feron et al., Arch Otolaryngol Head Neck Surg 1998 Aug;124(8):861-6).
Olfactory neuroblasts (olfactory neurons obtained by biopsy and placed in primary cell culture) from AD patients produce carboxy terminal amyloid precursor protein (APP) fragments that contain beta-amyloid (A-beta)(Crino et al., Ann Otol Rhinol Laryngol 1995 Aug;104(8):655-61). Crino et al. showed labeling of A-beta in the basal third of the olfactory neuroepithelium and in axons projecting through the lamina propria of AD patients. Thioflavin-S staining that detects amyloid deposition was also observed in the basal third of the olfactory neuroepithelium from AD patients. Alpha 7 nicotinic acetylcholine receptors are present in olfactory neurons probably including olfactory receptor cells in the nasal cavity (Alkondon et al., Neurosci Lett 1994 Aug 1;176(2): 152- 6; Alkondon et al., Eur J Neurosci 1997 Dec;9(12):2734-42; Bouvet et al., Neurosci Res 1988 Feb;5(3):214-23; Edwards et al., Experientia 1987 Aug 15;43(8):868-73; Edwards et al., Experientia 1988 Mar 15;44(3):208-11; Seguela et al., J Neurosci 1993 Feb;13(2):596-604).
Beta-amyloid peptide increases cytosolic-free Ca2+ in AD lymphoblasts (Ibarreta et al., Alzheimer Dis Assoc Disord 1997 Dec;11(4):220-7), and elevates mitogen-induced Ca2+ responses in freshly prepared human lymphocytes (Eckert et al., Life Sci 1994;55(25-26):2019-29). Amyloid precursor protein (APP) can be induced on the cell surface of human lymphocytes upon stimulation (Bullido et al., Biochim Biophys Acta 1996 Aug 21 ;1313(1 ):54-62) and increased APP-770 isoform occurs in lymphocytes from AD patients (Ebstein et al., Brain Res Mol Brain Res 1996 Jan;35(1-2):260-8). Lymphoblastoid cells from patients with early-onset and late-onset familial AD show increased expression of beta-APP mRNA and protein (Matsumoto et al., Eur J Biochem 1993 Oct 1 ;217(1 ):21-7). Lymphocytes from AD patients also exhibit an increased mRNA level for alpha 7 nicotinic receptor (Hellstrom- Lindahl et al., Brain Res Mol Brain Res 1999 Mar 20;66(1-2):94-103)
Based on the information described above, we propose that the analysis of the alpha 7 nicotinic acetylcholine receptor - beta amyloid peptides interaction in circulating blood cells and olfactory neuroepithelial neurons/neuronal processes or olfactory neuroblasts obtained from AD patients could be used as AD diagnostic tools, markers of AD progression and prognosis, and markers of therapeutic efficacy for any intervention or treatment targeting AD.
Thus, the present invention provides methods for diagnosing
Alzheimer's disease, monitoring the progression and prognosis of Alzheimer's disease and/or monitoring the therapeutic efficacy of any intervention or treatment of Alzheimer's disease comprising:
(a) obtaining a test sample from a subject wherein the test sample comprises circulating blood cells and/or olfactory neuroepithelial neuronal cell bodies or their neuronal processes (i.e., dendrites and axon of a nueron); and
(b) analyzing the test sample for interaction of an amyloid beta peptide (including, but not limited to, Aβ1-40, Aβ1-42and Aβι- 3 peptides and their fragments) with alpha-7 nicotinic acetylcholine receptors.
The compounds of formula I, such as 5,8-Dihydroxy-fraπs-2-di(Λ/- propylamino)-3-methyl-1 ,2,3,4-tetrahydronaphthalene (Compound 9), are made according to the procedures described in the Schemes and Examples which follow.
Abbreviations used in the instant specification, particularly the Schemes and Examples, are as follows:
Ac = acetyl
Ach = acetylcholine
AcOH = acetic acid
BSA = bovine serum albumin
DMF = N,N-dimethyl formamide
DMSO = dimethyl suifoxide
EtaN = triethyl amine
EtOAc = ethyl acetate
FCS = fetal calf serum i-Pr = isopropyl
Me = methyl
Mel = methyl iodide nAChR = nicotinic acetylcholine receptor
Ph = phenyl
PCC = pyridinium chlorochromate TEA triethyl amine THF tetrahydrofuran TLC thin layer chromatography
SCHEME 1
Figure imgf000023_0001
Compounds of the invention can be prepared as shown in Scheme 1. First a Diels-Alder reaction is performed on benzophenone (1 ) with a suitable diene such as isoprene to give a dione such as (2). Then, base-catalyzed isomerization of (2) affords a 1 ,4-dihydroxyphenyl compound (3), which is converted to dimethyl ether (4). Hydroboration followed by oxidation give alcohol (5), which is then oxidized to give ketone (6). Reductive amination with an amine such as propyl amine is then carried out using a suitable hydride reducing agent such as sodium cyanoborohydride, to give, for example, compound 7. This material can then be subjected to a reductive amination reaction such as with propionaldehyde as shown to yield (8). Compound (8) is then treated with HBr in acetic acid to cleave the methyl ethers and form dihydroxy compound (9). Alternatively, BBr3 may be used to cleave the methyl ethers and form dihydroxy compound (9). Moreover, a deficiency of BBr3 may be used to provide compounds in which only one of the methyl ethers has been removed to afford compounds with one hydroxy and one methoxy group.
Figure imgf000024_0001
Scheme 2
A further means of preparing compounds of the present invention is illustrated in Scheme 2. A suitable propargyl alcohol, such as 3-hydroxy-3-methyl- 1-butyne (10, R. = Me) is dehydrated to give an enyne such as compound 11. This material is then subjected to an aminomercuration reaction to afford a 2- amino-1 ,4-butadiene such as compound (12). Diels-Alder reaction of compound (12) with benzophenone (1) gives (13) which can be isomerized as in Scheme 1 with base to give the 1 ,5-dihyroxy compound (14). Reduction of the double bond of (14) catalytically with hydrogen and palladium on carbon, such as 10% palladium on carbon, yields compounds of the invention such as (15). Compounds of type 9 can be treated with a base such as triethylamine in a suitable solvent such as dioxane or methylene chloride along with electrophiles such as acid halides, phosphinyl halides, or alkyl halides to give the products of substitution on one or both of the phenolic hydroxyls. Such compounds can be active by themselves, or serve as prodrugs for compound 9.
The following Examples are set forth to aid in the understanding of the invention, and are not intended and should not be construed to limit in any way the invention set forth in the claims which follow thereafter.
Example 1
Preparation of 5,8-Dihydroxy-frans-2-di(/V-propylamino)-3-methyl-1 ,2,3,4- tetrahydronaphthalene (9)
a. 5,8-Dihydroxy-2-methyl-1 ,4-dihvdronaphthalene (3)
To a solution of p-benzoquinone (10.00 g, 92.5 mmol) in 1M solution of lithium perchlorate, in nitromethane (300 mL) was added isoprene (9.24 mL, 92.3 mmol) at room temperature. The resultant reaction mixture was stirred at room temperature under N2 for 4 hours. The reaction mixture was partitioned between EtOAc (500mL) and water (200 mL). The organic layer was washed with brine, dried over sodium sulfate and concentrated to give 2 as brown solid. The reaction was repeated on 15 g of benzoquinone to afford additional 2. These two runs were combined and carried on without further purification. To a solution of 2 (35.00 g, 199 mmol) in methylene chloride (400 mL) was added triethylamine (40 mL). The reaction mixture was stirred at room temperature for 1 hr. The product was precipitated out of solution by the addition of hexane (300 mL). The solid was collected by filtration and dried in a dessicator under high vaccuum overnight to provide 3 as a light brown solid. 1H NMR (300 MHz, DMSO-d6): δ 1.75 (s, 3H), 3.00-3.01 (m, 2H), 3.09 (s, 2H), 5.53 (s, 1 H), 6.43 (s, 2H), 8.49-8.52 (m, 2H). b. 5,8-Dimethoxy-2-methyl-1 ,4-dihvdronaphthalene (4)
To a cold (-78°C) solution of hydroquinone 3 (17.00 g, 96.6 mmol) in DMF under N2 was added unwashed 60% sodium hydride in mineral oil (8.90 g, 222.5 mmol). The resultant reaction mixture was warmed to room temperature over a 30 min period and subsequently treated with methyl iodide (14.3 mL, 230 mmol). After stirring the reaction mixture for 1 hr at room temperature the reaction mixture was diluted with ethyl acetate (1.5 L) and washed with brine (3X500mi). The organic solution was dried over sodium sulfate, filtered, concentrated, and the residue purified on silica gel (elution with 20% ethyl acetate\hexane) to afford 4 as a light yellow oil which solidified upon standing.
1H NMR (300 MHz, CD3OD): δ 1.77 (s, CH3), 3.07-3.09 (m, 2H), 3.16 (br s, 2H), 3.73(s, 3H), 3.75 (s, 3H), 5.52-5.53 (m, 1 H), 6.65 (s, 2H).
c. 5,8-Dimethoxy-frans-2-hvdroxy-3-methyl-1,2,3,4-tetrahydronaphthalene
(51
A solution of 4 (5.00 g, 24.5 mmol) in tetrahydrofuran (100ml) was cooled to 0 °C and treated with 1 M borane-tetrahydrofuran complex (24.5mL, 24.5mmol) and the resulting solution was stirred at room temperature for 4 h. Then, 12mL of 3 N NaOH solution was added slowly to the stirring reaction solution followed by the addition of 6 mL of 30% aqueous hydrogen peroxide. After stirring for 30 min, the resultant mixture was diluted with 500 mL of ethyl acetate and washed by brine (2X1 OOmL). The organic layer was dried over sodium sulfate, filtered and concentrated, and the residue was purified on silica gel (30% ethyl acetate\hexane) to give 5 as a white solid.
1H NMR (300 MHz, CDCI3): δ 1.13 (d, 3H, J=6.51 Hz), 1.60 (d, 1H), 1.75-1.90 (m, 1H), 2.26 (dd, 1H, J=10.0 Hz, 17.57 Hz), 2.48 (dd, 1H, J=8.93 Hz, 17.10 Hz), 2.96 (dd, 1 H, J=5.31,17.7Hz), 3.16 (dd, 1 H, J=5.33, 5.35 Hz), 3.63-3.69 (m, 1H), 3.78 (s, 6H), 6.62 (s, 2H).
d. 5,8-Dimethoxy-3-methyl-2-tetralone (6) A solution of alcohol 5 (10 g, 45 mmol) and dichloromethane (200 mL) at -30°C was treated dropwise with oxalyl chloride (4.92 g, 24 mL, 0.275 mol). The reaction was stirred at -30°C for 30 min, cooled to -60°C, and DMSO (6.4 mL) was added slowly over 15 min. The reaction was stirred for one hour, cooled to -78°C for 30 min, and then treated dropwise with triethylamine (40 mL). The reaction was warmed to room temperature and stirred for 1 hr, followed by the addition of water and dichloromethane with thorough mixing. The organic layer was separated, dried with MgSO4, filtered and the solvent was evaporated to an oil. Purification of this material using flash chromatography (flash silica, 70/30: hexane/EtOAc) afforded 6 as a white crystalline solid.
1H NMR (300 MHz, CDCI3) δ 6.7 (q, 2 H), 3.78 (s, 3 H), 3.75 (s, 3 H), 3.5 (dd, 2 H), 3.3 (m, 1 H), 2.55 (m, 2 H), 1.2 (d, 3 H).
e. 5,8-Dimethoxy-2-Λ/-propylamino-3-methyl-1 , 2,3,4- tetrahydronaphthalene (7)
To a solution of 6 (0.15 g, 0.68 mmol) in acetonitrile (10 mL) was added sodium cyanoborohydride (0.085 g, 1.30 mmol), 0.05ml of acetic acid and N- propylamine (0.08 mL, 1.3 mmol). The resulting solution was stirred at room temperature overnight and poured into 10 mL of 1N NaOH solution. The mixture was extracted with EtOAc, dried over sodium sulfate, concentrated, and the residue purified by preparative TLC (elution with 30% ethyl acetate in hexane) to give amine 7 as a colorless oil.
1H NMR (300 MHz, CD3OD): δ 0.85 (d, 3H, J= 7.07 Hz), 0.93-0.97 (m, 3H), 1.09 (t, 2H), 1.49-1.61 (m, 2H), 2.12-2.42 (m, 2H), 2.51-2.76 (m, 4H),
2.82-3.08 (m, 2H), 3.73 (s, 3H), 3.74 (s, 3H), 6.65 (s, 2H).
f. 5.8-Dimethoxy-fra/7s-2-di(Λ/-propylamino)-3-methyl-1.2.3.4- tetrahvdronaphthalene (8) To a solution of 7 (0.11 g, 0.42 mmol) in acetonitrile (10ml) was added sodium cyanoborohydride (0.05 gm, 0.6 mmol), 0.03 mL of acetic acid and propionaldehyde (0.045 mL, 0.60 mmol). The resulting solution was stirred at room temperature overnight and poured into 10 mL of 1 N NaOH solution. The mixture was extracted with EtOAc, dried over sodium sulfate, concentrated, and the residue was purified by preparative TLC (elution with 30% ethyl acetate in hexane) to give amine 8 as a light yellow solid. 1H NMR (300 MHz, CD3OD): δ 0.84 (d, 3H, J= 6.94 Hz), 0.92 (t, 2H, J= 7.30, 7.32Hz), 1.27-1.59 (m, 4H), 2.33-2.42 (m, 2H), 2.54-3.02 (m, 8H), 3.73 (s, 3H), 3.76 (s, 3H), 6.66 (s, 2H).
In a similar manner was prepared compounds 19, 21, 25, 27, 29, 35, 37, 39, 41, 52, and 54. In order to separate the enantiomers of 8, 2.859 g of 8 was passed through a CHIRALPAK® AD™ chiral cellulose-based high pressure liquid chromatography column (8 cm X 30 cm) at 25° C using hexane/isopropyl alcohol (99/1 ) as the eluant to give two fractions as oils. Fraction 1 (18) had a 98.6% enantiomeric excess (e.e.) and fraction 2 (17) had a 97.9% e.e. Fraction 1 (0.306g, 1.0 mmol) and fumaric acid (0.141 g, 1.2 mmol) were dissolved in ethanol (2 mL) with heating. The ethanol was evaporated and the residue triturated with diethyl ether affording the fumarate salt of 18 as a white solid. Similarly obtained from Fraction 2 (0.301 g, 1.0 mmol) and fumaric acid (0.141 g, 1.2 mmol) was the fumarate sale of 17.
g. 5,8-Dihvdroxy- raπs-2-di(Λ/-propylamino)-3-methyl-1 , 2,3,4- tetrahvdronaphthalene (9)
A mixture of 8 (1.4 g, 4.6 mmol) and 3.5 mL of 48 % aqueous HBr in acetic acid (3.5 mL) was stirred at 100°C under N2 overnight. The reaction mixture was then cooled to 0°C and extracted with diethyl ether (20 mL). The organic solution was neutralized with aqueous NaHCO3 and separated. The aqueous layer was extracted with ether (20 mL) and the combined ether solutions were dried over Na2SO4, filtered, and concentrated to give 9.
1H NMR (600 MHz, CDCI3): δ 0.86-0.92 (dt, 6H), 1.13 (d, 3H, J=6.42 Hz), 1.42-1.50 (m, 4H), 1.88-1.93 (m, 1 H), 2.23-2.28 (m, 1 H), 2.39-2.52 (m, 6H), 2.61-2.63 (m, 1 H), 2.90 (dd, 1 H, J=4.75, 16.28 Hz), 2.95 (dd, 1 H, J=5.05, 16.75Hz), 4.84 (br s, 2H), 6.50 (dd, 2H, J=8.52Hz). CI-MS: m/e MH+ 278 (40%).
In a similar manner 17 and 18 were converted to 15 and 16 respectively. Additionally, demethylation of the appropriate dimethyl ethers by the procedure described here led to the preparation of compounds 20, 22, 26, 28, 30, 32, 34, 36, 40, 42, 53, and 55. In the course of the demethylation of 37, the butyl ether was also removed to obtain 38.
Example 2
Preparation of 5,8-Dihydroxy-cιs-2-(1 -morpholinyl)-3-methyl-1 ,2,3,4- tetrahydronaphthalene (24)
a. 5,8-Dimethoxy-c/s-2-(1 -morpholiny0-3-methyl-1 ,2,3,4- tetrahvdronaphthalene (23).
Ketone 6 (0.299 g, 1.36 mmol) was dissolved in toluene (1 mL) followed by the addition of morphoiine (0.130 g, 0.13 mL, 1.5 mmol) and Ti(/-PrO) (0.611 g, 0.64 mL, 2.15 mmol). The reaction mixture was stirred for 15 hrs at room temperature. Methanol (2 mL) was added followed by portionwise addition of NaBH4 (0.15 g, 3.9 mmol) over 1 hr. Dichloromethane and 1N
NaOH were added with thorough mixing and the organic layer was separated, dried with MgS04, filtered and evaporated to an oil. Purification of this material using flash chromatography (flash silica, 75/25: hexane/EtOAc) afforded product 23. 1H NMR (300 MHz, DMSO-d6) δ 6.6 (s, 2 H), 3.6 (d, 6 H), 3.5 (s, 4 H),
3.2 (s, 4 H), 3.2 (s, 6 H), 2.8 (d, 1 H), 2.7 (d, 1 H), 2.3 (m, 2 H), 2.05 (m, 2 H), 0.6 (d, 3 H).
b. 5,8-Dihvdroxy-c/s-2-(1-morpholinyl)-3-methyl-1 ,2,3,4-tetrahydronaphthalene (241
A solution of 23 (20 mg, 0.066 mmol) and dichloromethane (2 mL) at - 78°C was treated with a 1 M solution of BBr3 in dichloromethane (1 mL, 1 mmol.) and stirred for 1 hr at -78°C. After warming to room temperature, methanol (5 mL) was added and the solvents were evaporated. Methanol addition to the residue followed by evaporation was done twice. The residue was dissolved in acetonitrile (25 mL) and treated with triethylamine (2 mL). After stirring for 2 hrs, water and ethyl acetate were added with thorough mixing. The organic layer was separated, dried with MgSO , filtered, and solvent evaporated to yield 24 as an oil.
1H NMR (300 MHz, CD3OD) δ 6.7 (s, 2 H), 3.4 (m, 4 H), 3.3 (m, 1 H), 2.6 (m, 5 H), 1.9 (m, 1 H), 1.5 (m, 2 H), 1.05 (d, 3 H), 0.9 (m, 3 H).
Example 3
Preparation of 5,8-Dimethoxy-c/s and fraπs-2-{/V-propyl- V- propargyl)amino-3-methyl-1,2,3,4-tetrahydronaphthalene (31 and 33)
Ketone 6 (1 g, 4.5 mmol) was dissolved in acetonitrile (75 mL) followed by the addition of acetic acid (0.54 mL, 9 mmol, 2 eq.) and propargylamine (0.6 mL, 9 mmol, 2 eq.). The reaction mixture was stirred at room temperature for 1 hr. Sodium cyanoborohydride (0.6 g, 9 mmol, 2 eq.) was added portionwise (3x) over 1.5 hrs (every 30 min), and the reaction mixture was stirred overnight at room temperature. Ethyl acetate and 1 N NaOH were added with thorough mixing and the organic layer was separated, dried with MgS04, filtered and evaporated to an oil. The product was purified with flash chromatography (50:50/ethyl acetate:hexane) to yieid an oil of 5,8-dimethoxy-c/s and trans-2-N- (propargyl)amino-3-methyl-1 ,2,3,4-tetrahydronaphthalene (1.2 g, 100%). This material was dissolved in acetonitrile along with propionaldehyde (0.65 g, 9 mmol, 2 eq.) and acetic acid (0.5 mL, 9 mmol) and stirring for 1 hr at room temperature. One hour later, sodium cyanoborohydride (0.6 g, 9 mmol, 2 eq.) was added portionwise (3x) over 1.5 hours (every 30 mins). The reaction was stirred overnight at room temperature. Ethyl acetate and 1 N NaOH were added to the reaction with thorough mixing and the organic layer was separated, dried with MgSO , filtered and evaporated to a mixture of products 31 and 33. Purification of this material using flash chromatography (flash silica,
80:20/ hexane:ether) afforded product 31 and 33. Compound H NMR (300 MHz, CDCI3) δ 6.6 (s, 2 H), 3.8 (s, 6 H), 3.4 (q, 2 H), 3.0 (dd, 2 H), 2.6 (m, 4 H), 2.2 (m, 2 H), 1.85 (m, 1 H), 1.5 (m, 2 H), 1.1 (d, 3 H), 0.9 (t, 3 H). Compound 33: 1 H NMR (300 MHz, CDCI3) δ 6.6 (s, 2 H), 3.78 (s, 3 H), 3.75 (s, 3 H), 3.6 (q, 2 H), 3.05 (dd, 1 H), 2.75 (m, 2 H), 2.61 (m, 3 H), 2.3 (m, 2 H), 2.1 (s, 1 H), 1.55 (m, 2 H), 0.9 (t, 3 H), 0.8 (d, 3 H).
Example 4
Preparation of 5-[2-(3-Methyl)butenyl]-8-hydroxy-frans-2-di(Λ - propylamino)-3-methyl-1,2,3,4-tetrahydronaphthalene (43); and 5- Hydroxy-8-[2-(3-methyl)butenyl]-frans-2-di(yV-propylamino)-3-methyl- 1 ,2,3,4-tetrahydronaphthaiene (44) Diol 9 (0.3 g, 1.2 mmol) was dissolved in acetone (40 mL) followed by the addition of potassium carbonate (0.19 g) and 1 -bromo-3-methyl-2-butene (0.1 mL, 0.87 mmol). The reaction mixture was stirred for 48 hrs at room temperature. Dichloromethane and water were added with thorough mixing and the organic layer was separated, dried with MgSO4, filtered and the solvent was evaporated to yield an oil. Purification of this material using flash chromatography (flash silica, 70:30/ hexane:ether) afforded products 43 and 44. The regiochemical structural assignment between the two of them is uncertain. Compound 43: 1H NMR (300 MHz, CDCI3) δ 6.5 (s, 2 H), 5.5 (t, 1 H), 4.4 (m, 2 H), 3.0 (m, 2 H), 2.4 (m, 8 H), 1.8 (m, 1 H), 1.72 (s, 3 H), 1.65 (s, 3 H), 1.4 (m, 4 H), 1.1 (dd, 3 H), 0.85 (t, 6 H). Compound 44: 1H NMR (300 MHz, CDCI3) δ 6.55 (s, 2 H), 5.45 (t, 1 H), 4.4 (m, 2 H), 3.05 (dd, 1 H), 2.85 (dd, 1 H), 2.4 (m, 7 H), 1.65 (s, 3 H), 1.6 (s, 3 H), 1.55 (m, 2 H), 1.4 (m, 4 H), 1.1 (m, 4 H), 1.1 (d, 3 H), 0.9 (t, 6 H). Example 5
Preparation of 5-Hydroxy-8-methoxy-fraπs-2-di(W-propylamino)-3- methyl-1,2,3,4-tetrahydronaphthalene (45); and 5-Methoxy-8-hydroxy- trans-2-di(/V-propylamino)-3-methyl-1 ,2,3,4-tetrahydronaphthalene (46)
Compound 8 (1.85 g, 6 mmol) was dissolved in dichloromethane (20 mL) and cooled to -78°C, followed by the addition of boron tribromide (6 mL of a 1 M solution, 6 mmol, 1 eq.). The reaction mixture was stirred at -78°C for 1 hr and then warmed to room temperature for 2 hrs. The reaction was quenched with methanol (5 mL), and the solvent was evaporated (methanol quench repeated twice more). The resulting oil was dried overnight under high vacuum (5 mm Hg). The residue was dissolved in dichloromethane and NaHC03 was added, mixed thoroughly, and the organic layer was separated, dried with MgSO4, filtered and the solvent evaporated to a mixture of two regioisomers 45 and 46 (1:1 ). The purification of a mixture (200 mg) was conducted on a reverse phase HPLC (C18 column, 69:30:1/ water: acetonitrile:triflouroacetic acid) to afford 45 and 46 as triflouroacetate salts. Compound 45 (TFA salt): 1H NMR (300 MHz, CDCI3) δ 6.65 (q, 2H), 4.7 (bs, 1 H), 3.7 (s, 3 H), 3.5 (m, 1 H), 3.3 (m, 1 H), 3.0 (m, 4H), 2.7 (m, 2 H), 2.3 (m, 1 H), 2.0 (m, 5 H), 1.15 (d, 3 H), 0.9 (m, 6 H). Compound 46 (free base): 1H NMR (300 MHz, CDCI3) δ 6.55 (q, 2 H), 4.3 (bs, 1 H), 3.75 (s, 3 H), 3.05 (dd, 1 H), 3.0 (dd, 1 H), 2.6 (m, 1 H), 2.4 (m, 6 H), 2.2 (m, 1 H), 1.8 (m, 1 H), 1.4 (m, 4 H), 1.1 (d, 3 H), 0.85 (t, 6 H). The regiochemistry of the structure of 46 was confirmed through careful analysis of HMBC (Heteronuclear Multiple Bond Correlation) connectivities and nOe effects. The structure of 46 was established by connectivity between the methoxy substituent on the aromatic ring and the benzylic methylene adjacent to the methyl-substituted carbon.
Example 6
Preparation of 5,8-Dimethoxy-2-[_V-(L-a_any. methyl ester)] -3- methyl-1 ,2,3,4-tetrahydronaphthalene (48) Ketone 6 (1 g, 4.5 mmol) was dissolved in acetonitrile (75 mL) and acetic acid (0.54 mL, 9 mmol, 2 eq.), and alanine methyl ester hydrochloride (1.3 g, 9 mmol, 2 eq.) was added. The reaction was stirred at room temperature for 1 hr, and then sodium cyanoborohydride (0.6 g, 9 mmol, 2 eq.) was added portionwise (3x) over 1.5 hrs (every 30 min). The reaction was stirred overnight at room temperature. Ethyl acetate and 1 N NaOH were added with thorough mixing and the organic layer was separated, dried with MgSO4, filtered and evaporated to an oil. The product was purified with flash chromatography (50:50/ ethyl acetate: hexane) to yield 48 as an oil. Compound 48, (mixture of cis and trans stereoisomers) 1 H NMR (300 MHz, CDCI3) δ 6.64 (d, 2 h), 4.2 (m, 1 H), 3.85 (m, 3 H, 3.75 (s, 6 H), 3.5 (m, 1 H), 3.1 (m, 2 H), 2.8 (m, 2H), 2.55 (m, 1 H), 2.3 (m, 1 H), 1.7 (m, 3 H), 1.2 (m, 3 H).
Example 7
Preparation of 5,8-Dimethoxy-2-[Λ/-(L-alanyl methyl ester)-Λ/- propyl]-3-methyl-1 ,2,3,4-tetrahydronaphthalene (47)
Compound 48 (0.28 g, 0.8 mmol) was dissolved in acetonitrile along with propionaldehyde (0.1 mL, 1.6 mmol, 2 eq.) and acetic acid (0.2 mL, 1.6 mmol), and stirred for 1 hr at room temperature. One hour later, sodium cyanoborohydride (0.2 g, 1.6 mmol, 2 eq.) was added portionwise (3x) over a period of 1.5 hrs (every 30 minutes). The reaction was stirred overnight at room temperature. Ethyl acetate and 1 N NaOH were added to the reaction with thorough mixing and the organic layer was separated, dried with MgSO , filtered and evaporated to an oil. This material was purified using flash chromatography (flash silica, 80:20/ hexane:ether) to afford (1 :1 mixture of cis and trans stereoisomers) 47. 1H NMR (300 MHz, CDCI3) δ 6.6 (d, 2 H), 3.75 (s, 6 H), 3.7 (d, 4 H), 3.0 (m, 2 H), 2.65 (m, 3 H), 2.2 (m, 1 H), 1.7 (m, 2 H), 1.45 (m, 2 H), 1.25 (d, 3 H), 1.0 (m, 3 H), 0.8 (t, 3 H). Example 8
Preparation of 5-Diphenylphosphinoyl-8-hydroxy-fraπs-2-di(Λ/- propylamino)-3-methyl-1 ,2,3,4-tetrahydronaphthalene (58); and 5,8- bis(Diphenylphosphinoyl)-3-methyl-1 ,2,3,4-tetrahydronaphthalene (59)
Compound 9 (0.5 g, 2.0 mmol) was dissolved in dioxane (100 mL) at 0°C. Triethylamine (6 mL) was added and the reaction mixture was stirred at 0°C for 30 mins, followed by the addition of diphenylphosphinyl chloride (0.84 mL, 4.4 mmol). The reaction mixture was stirred overnight at room temperature. NaHCO3 and dichloromethane were then added with thorough mixing. The organic layer was separated, dried with MgSO4, filtered and the solvent was evaporated to a crude oil which was purified by flash chromatography (flash silica, 70:30/ hexane:ether) to yield 59 and 58. Compound 59: 1H NMR (300 MHz, CDCI3) δ 7.85 (d, 4 H), 7.8 (d, 4 H), 7.5 (m, 12 H), 6.8 (q, 2 H), 3.0 (m, 2 H), 2.4 (m, 7 H), 1.8 (m, 1 H), 1.4 (m, 4 H), 1.05 (d, 3 H), 0.85 (t, 6 H). Compound 58 (1 :1 mix of regioisomers): 1H NMR (300 MHz, CDCI3) δ 7.85 (m, 4 H), 7.5 (m, 6 H), 6.6 (q, 1 H), 6.48 (s, 1 H), 6.21 (d, 1 H), 2.9 (m, 3 H), 2.3 (m, 9 H), 1.65 (m, 1 H), 1.4 (m, 1 H), 1.05 (d, 3 H), 0.85 (m, 6 H). In a similar manner, starting from 32, was prepared both compounds 57 and 56. In addition, in a similar manner, starting from 9, was prepared compounds 49-51 using the appropriate reagents.
Example 9
Preparation of 5,8-Dimethoxy-c/s-2-[Λ/-(4-(2-(1 - piperidinyl)ethoxy)phenyl)]amino-3-methyl-1,2,3,4-tetrahydronaphthalene
(60)
A solution of 1-(2-chloroethoxy)-4-nitrobenzene (2.01 g, 0.01 mol), piperidine (2.55 g, 0.03 mol), and toluene (10 mL) was refluxed overnight. The reaction mixture was filtered and the filtrate evaporated to give 2.97 g of 1 -[2- (1-piperidinyl)ethoxy]-4-nitrobenzene as an orange oil, MS M+ (m/e) 251.18. This material (1.25 g, 0.004 mol) was hydrogenated in ethanol (25 mL) in the presence of 10% Pd-C catalyst at 40 psig and 25° C overnight. Filtration and evaporation of the reaction mixture gave 0.95 g of 4-[2-(1- piperidinyl)ethoxy]aniline as a light brown oil, MS M+ (m/e) 221.24. A solution of this oil (0.220 g, 1.0 mmol), 3-methyl-5,8-dimethoxy-2-tetralone (6, 0.206, 0.94 mmol), and 1 ,2-dichloroethane (3.5 mL) was treated with sodium triacetoxyborohydride (0.300 g, 1.4 mmol) and acetic acid (0.060 g, 1.0 mmol) and the resulting mixture was stirred overnight at 25° C. The reaction was treated with 3N sodium hydroxide solution with vigorous stirring. The organic layer was separated, washed with saturated sodium chloride solution, dried over potassium carbonate, filtered and evaporated to a red oil. This material was purified by C-18 reverse phase high pressure liquid chromatography affording compound 60 as a pink solid. 1H NMR (300 MHz, CDCI3): δ 7.10- 7.22 (m, 2H), 6.52-6.82 (m, 4H), 4.25-4.33 (m, 2H), 3.60-3.80 (m, 3H), 3.75 (s, 3H), 3.71 (s, 3H), 3.48-3.38 (m, 2H), 2.80-3.05 (m, 4H), 1.80-2.10 (m, 4H), 1.10-1.50 (m, 3H), 1.00 (d, 3H). MS M+ (m/e) 425.38.
The compounds shown in Table 1 , below, were prepared according to the procedures described herein.
Table 1
Figure imgf000035_0001
Cp. R5 R6 R2 R3 Config. MS M* (m/e)
9 H H Pr Pr trans 278.24
15 H H Pr Pr trans, ent. 278.25
16 H H Pr Pr trans, ent. 278.21
7 Me Me H Pr trans
8 Me Me Pr Pr trans 306.28
17 Me Me Pr Pr trans, ent. 306.24
18 Me Me Pr Pr trans, ent. 306.27 Cp. R5 Re R2 R3 Config. MS M+ (m/e)
19 Me Me Pr Ph(CH2)3 trans 382.28
20 H H Pr Ph(CH2)3 trans 354.27
21 Me Me Pr cCeHnCH trans 360.32
22 H H Pr cCeHιιCH trans 332.32
23 Me Me -(CH2)20(CH2)2- cis 292.66
24 H H -(CH2)20(CH2)2- cis 264.28
25 Me Me Pr CH2Ph trans 354.74
26 H H Pr CH2Ph trans 326.21
27 Me Me Pr MeS(CH2)3 trans 352.29
28 H H Pr MeS(CH2)3 trans 324.24
29 Me Me Pr (2-thienyl)CH2 trans 360.24
30 H H Pr (2-thienyl)CH2 trans 332.14
31 Me Me Pr CH2CCH trans 302.21
32 H H Pr CH2CCH trans 274.16
33 Me Me Pr CH2CCH cis 302.22
34 H H Pr CH2CCH cis 274.16
35 Me Me Pr CH2(4-IPh) trans 480.11
36 H H Pr CH2(4-IPh) trans 452.14
37 Me Me Pr CH2(4-BuOPh) trans 426.25
38 H H Pr CH2(4-OHPh) trans 342.16
39 Me Me Pr CH2(2-CIP ) trans 388.18
40 H H Pr CH2(2-CIPh) trans 360.12
41 Me Me Pr CH2(4-MeP ) trans 368.29
42 H H Pr CH2(4-MePh) trans 340.18
43 H X Pr Pr trans 346.29
44 X H Pr Pr trans 346.30
45 Me H Pr Pr trans 292.22
46 H Me Pr Pr trans 292.22
47 Me Me Pr CH(Me)C02Me cis & trans 350.26
48 Me Me H CH(Me)C02Me cis & trans 308.19
49 Y Y Pr Pr trans 446.39
50 H Y Pr Pr trans 362.26
51 Y H Pr Pr trans 362.30
52 Me Me Pr Me trans 278.19
53 H H Pr Me trans 250.18
54 Me Me Pr Z trans 402.28
55 H H Pr z trans 374.25
56 H X' Pr CH2CCH trans 474.13
57 X' X' Pr CH2CCH trans 674.11
58 H X' Pr Pr trans 478.21
59 X' X' Pr Pr trans 678.11
60 Me Me H Y' cis 425.38
X = -CH2CH= CMe2
Y = -C(0)-t-Bu
Z = -CH2CH2CH(Me)CI- l2CH2CH=CMe2
X' = P(0)Ph2
Y' = 4-[(2-(1 -piperidinyl) ethyl)oxy]Ph
Example 10
As a specific embodiment of an oral composition, 100 mg of Compound 9 from Example 1 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size O hard gel capsule. Example 11
1-42 and Aβι_40 binding to alpha-7 nAChRs can be demonstrated in competitive binding experiments with α-bungarotoxin
SK-N-MC cell membranes were incubated with 0.5 nM of 125l-α- bungarotoxin in the presence of various concentrations of Aβι_ 2 and Aβ1-40 at 25°C for 1 hr. The assay mixture was then rapidly filtered and the radioactivity that was retained on the filter was determined. These studies showed that both Aβι- 2 and
Figure imgf000037_0001
inhibited 125l-α-bungarotoxin binding in a concentration- dependent manner with ICso values of 1 pM and 100 pM, respectively.
Example 12
1- 2 binding to alpha-7 nAChRs is supported by competitive binding experiments with 125I-Aβι_40 and provides a seed for amyloid deposition and, thus, incipient plaque formation Alpha-7 nAChRs were immobilized on wheat germ agglutinin coupled yittrium SPA beads and allowed to incubate with 125l-Aβ1-40 in the presence of various concentrations of cold Aβ^ or Aβ1-42. The data showed that 0.1 femtoM of cold Aβι-42 completely abolished the binding, suggesting that Aβι-42 interacted with alpha-7 nAChRs with high affinity. However, in the presence of 10 nM of cold
Figure imgf000037_0002
to alpha-7 nAChRs was dramatically increased. We attribute this increase of binding at higher concentrations to
Figure imgf000037_0003
precipitation of the Aβ peptides on the membranes. Prolonged incubation at lower concentrations also revealed that total binding was increased in reactions even with 100 pM of cold Aβ1-42. In control experiments, 80 pM 25l-Aβ1-40 did not bind appreciably, along with 1 pM to 1 nM of Aβ1-40, to 30 μg of Bowes melanoma membranes. Thus, Aβ deposition can occur on biological cell membranes containing alpha-7 nAChRs, but does not occur non-specifically on control membranes, suggesting that alpha-7 nAChRs can specifically seed the aggregation of β- amyloid in biological systems. Example 13
Aβι_42 binding to alpha-7 nAChRs is of higher affinity than Aβ^o binding to alpha-7 nAChRs
Alpha-7 nAChRs were allowed to interact with
Figure imgf000038_0001
or Aβ1-42 and the mixture was then immunoprecipitated with anti-alpha-7 nAChR antibodies. Western analyses of the immunoprecipitated proteins identified only the presence of Aβ1-42, indicating that the Aβ1- 2/alpha-7 nAChR interaction is robust and of high affinity. Since it has already been shown that Aβ1-40 binds to the alpha-7 receptor, the failure in detecting
Figure imgf000038_0002
in this co-precipitation experiment suggests that the Aβ1- 0/alpha-7 nAChR interaction is of lower affinity than the Aβ1-42/alpha-7 nAChR interaction.
Example 14
Compound 9 inhibited Aβ aggregation
Aβs are known to form aggregates leading to the formation of amyloid plaques that are characteristic of Alzheimer's disease. This phenomenon can be demonstrated in vitro by using Synthaloid plates, coated with Aβ crystallization centers, and labelled Aβs for detecting aggregation. We have validated this aggregation assay using both 125l-Aβ1-40 and
Figure imgf000038_0003
in a buffer containing 50 mM HEPES, pH 7.4, 0.1% BSA, 10% FCS and protease inhibitors. About 500 pM of 125l-Aβ1- 0 or 100 nM of fluo-Aβ^o in 100 μl of each well of the 96-well Synthaloid plate was allowed to incubate for 2.5 hr at room temperature in the presence or absence of inhibitors. At the end of the incubation, unbound protein in the wells was removed by three washes using the above buffer. The amount of bound protein which represented Aβ aggregation was measured either by scintillation counting or fluorescence measurements. Compound 9 was found to potently inhibit Aβ aggregation with 10nM ICδo- Time course studies also showed that prolonged incubation of amyloid aggregates with Compound 9 resulted in disaggregation.
Compounds which inhibit Aβ aggregation with an IC50 <100 micromolar may be effective in inhibiting the binding of Aβ to alpha-7 in such a manner as to have a positive therapeutic effect useful for the treatment of neurodegenerative disorders.
Example 15
Compound 9 blocked the effect of Aβs on acetylcholine release from synaptosomes
Synaptosomes from guinea pig hippocampus were incubated with 0.1 μM 3H-choline and then subjected to repeated washes to remove unincorporated 3H-choline. The synaptosomes were treated with 65 mM K+ for 30 seconds to elicit 3H-acetylcholine release. While Aβ^o and Aβ^z at 100 pM both inhibited acetylcholine release from these preparations (33% inhibition for both Aβι- 0 and Aβι-42). pretreatment of the synaptosomes with both 10 nM of Compound 9 and 100 pM of either
Figure imgf000039_0001
stimulation was found to have no effect on acetylcholine release.
Example 16
Compound 9 inhibited 125l-Aβ1-40 binding to alpha-7 nAChRs
To determine the effect of compound 9 on 125l-Aβι-40to alpha-7 nAChRs, alpha-7 nAChR contained SK-N-MC cell membranes were immobilized on wheat germ agglutinin coupled yittrium SPA beads and allowed to incubate with 25l-Aβ1-40 in the presence of various concentrations of compound 9. The result demonstrated that Compound 9 efficiently inhibited the binding of 125l- SK-N-MC cells with a 300 pM IC50. Compounds which inhibit the binding of Aβ to alpha-7 nAChRs with an IC50 <1 micromolar may have a positive therapeutic effect useful for the treatment of neurodegenerative disorders.
Example 17
Assay for peptide binding to the 206-216 stretch of the human α7 nAChR
A 2-5 ug quantity of the 11 amino acid peptide comprised of the 206- 216 stretch of the α7 nAChR (N-Ac, C(O)NH2) are added to a 96-well microtiter plate in 50 mL 10mM HEPES, pH 7.4, or any buffer 50ul. 125l-β-amyloidi-40 (2000Ci/mmol, 50pM) was added to the wells in the presence and absence of inhibitors (1 uM to 10 uM) dissolved in 1uL of 30% or 100% DMSO. Unbound ligands were removed and bound radioactivity was measured using a Microbeta liquid scintillation counter. Ligand binding can be inhibited by α- bungarotoxin, the peptide itself, and Compound 9.
Biological data for representative compounds of the present invention is provided in Table 2 which follows.
Table 2
Cp. % Inh Agg. % α7 %ACh
# (cone μM) Release at nAChR 10 μM
(ICs>. μM)
206-216
Binding
9 >90 (10) 94 94 (0.1)
15 50-90 (10) 85 (0.25)
16 50-90 (10) 44 (10)
7 50(100) NT NT
8 50-90 (10) NT (0.7)
17 >90 (10) NT 63 (1)
18 50-90 (10) NT 70 (10)
19 30 (5) NT NT
20 39 (5) NT NT
21 20 (5) NT NT
22 38 (50) NT NT
23 NT 54 NT
24 NT 43 NT
25 NT 48 NT
26 <30 (10) 64 NT
27 NT NT NT
28 50-90 (10) 34 NT Cp. % Inh Agg. % α7 %ACh
# (cone μM) Release at nAChR 10 μM
206-216 (ICso, μM) Binding
29 12(10) NT NT
30 38(5) NT NT
31 34(10) NT NT
32 69(10) NT NT
33 NT NT NT
34 69 (10) NT NT
35 38(10) NT NT
36 <30(10) NT NT
37 <30(10) NT NT
38 <30 (10) NT NT
39 85(10) NT NT
40 75(10) NT NT
41 <30(10) 19 14
42 60 (10) NT NT
43 50-90(10) 10 0
44 50-90(10) 23 0
45 30-50 (78) NT 78
46 50-90(10) 18 0
47 NT NT NT
48 <30(10) 2 0
49 50-90(10) 0 17
50 <30(10) 0 67
51 30-50(10) 0 54
52 60(10) 0 0
53 50-90(10) 0 18
54 50-90 (10) 0 0
55 50-90(10) 0 15
56 <30(10) 78 8
57 <30(10) 63 11
58 <30(10) 42 18
59 <30 (10) 72 25
60 very active 0 24
While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, it will be understood that the practice of the invention encompasses all of the usual variations, adaptations and/or modifications as come within the scope of the following claims and their equivalents.

Claims

WHAT IS CLAIMED IS:
1. A method of treating a neurodegenerative disorder in a subject in need thereof which comprises administering to the subject an amount of a compound effective to inhibit the binding of an amyloid beta peptide with alpha-7 nicotinic acetylcholine receptors.
2. The method of Claim 1 , wherein the alpha-7 nicotinic acetylcholine receptors are human alpha-7 nicotinic acetylcholine receptors.
3. The method of Claim 2, wherein the neurodegenerative disorder is selected from Alzheimer's disease, Pick's disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy- Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson's-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington's disease, Parkinson's disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette's disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy's disease), primary lateral sclerosis, familial spastic paraplegia, Werdnig-Hoffmann disease, Kugelberg-Welander disease, Tay-Sach's disease, Sandhoff disease, familial spastic disease, Wohlfart-Kugelberg-Welander disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt-Jakob, Gerstmann-Straussler-Scheinker disease, Kuru and fatal familial insomnia, age-related dementia, vascular dementia, diffuse white matter disease (Binswanger's disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica or frontal lobe dementia, neurodegenerative disorders resulting from cerebral ischemia or infection including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type, intracranial and intravertebral lesions, hereditary cerebral angiopathy, nonneuropathic hereditary amyloid, Down's syndrome, ma╬▒oglobulinemia, secondary familial Mediterranean fever, Muckle- Wells syndrome, multiple myeloma, pancreatic- and cardiac-related amyloidosis, chronic hemodialysis arthropathy, or Finnish and Iowa amyloidosis.
4. The method of Claim 3, wherein the amyloid beta peptide is A╬▓1- 2.
5. The method of Claim 3, wherein the neurodegenerative disorder is Alzheimer's disease.
6. The method of Claim 5, wherein the compound inhibits the binding of Aβχ-42 with the human alpha-7 nicotinic acetylcholine receptor by binding to
7. The method of Claim 3, wherein the compound inhibits the binding of A╬▓╬╣-42 with the human alpha-7 nicotinic acetylcholine receptor by binding to human alpha-7 nicotinic acetylcholine receptors.
8. The method of Claim 3, wherein the compound inhibits the binding of A╬▓╬╣-42 with the human alpha-7 nicotinic acetylcholine receptor by inhibiting aggregation of amyloid beta peptides.
9. The method of Claim 5, wherein the compound is of the formula I
Figure imgf000043_0001
wherein Ri is hydrogen or C╬╣-C alkyl;
R2 is selected from hydrogen, C╬╣-C6 alkyl, aryl or C7-C╬╣o aralkyl; R3 is selected from hydrogen, Ci-Ce alkyl, C3-C10 alkenyl, C3-C8 cycloalkylC╬╣-C6 alkyl, d-C6 alkoxycarbonylC╬╣-C6 alkyl, C╬╣-C6 alkylthio, heteroarylC╬╣-C4 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted C7-C10 aralkyl wherein the substituent on the aryl or aralkyl are one or more substituents independently selected from the group consisting of halogen, hydroxy, C╬╣-C6 alkyl and unsubstituted or substituted C╬╣-C6 alkoxy wherein the substituents on the alkoxy are one or more substituents independently selected from amino, C╬╣-C6 alkylamino, C╬╣-C6 dialkylamino, pyrrolidinyl, piperidinyl, azepinyl or morpholinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
R_┬╗ is C╬╣-C6 alkyl, aryl, or C7-C10 aralkyl; and R5and R6 are each independently selected from hydrogen, Ci-C6 alkyl, C3-C╬╣0 alkenyl, C╬╣-C8 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
10. A method of treating and/or preventing dementia in an Alzheimer's disease patient which comprises administering to the subject a therapeutically effective amount of a compound which inhibits the binding of an amyloid beta peptide with alpha-7 nicotinic acetylcholine receptors.
11. The method of Claim 10, wherein the amyloid beta peptide is A╬▓╬╣-42 and the alpha-7 nicotinic acetylcholine receptors are human alpha-7 nicotinic acetylcholine receptors.
12. The method of Claim 11 , wherein the compound is of the formula
Figure imgf000044_0001
wherein Ri is hydrogen or C╬╣-C alkyl; R2 is selected from hydrogen, C╬╣-C6 alkyl, aryl or C7-C10 aralkyl;
R3 is selected from hydrogen, C╬╣-C6 alkyl, C3-C10 alkenyl, C3-C8 cycloalkylCi-Ce alkyl, C╬╣-C6 alkoxycarbonylC╬╣-C6 alkyl, C╬╣-C6 alkylthio, heteroarylC1-C4 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted C7-C10 aralkyl wherein the substituent on the aryl or aralkyl are one or more substituents independently selected from the group consisting of halogen, hydroxy, C╬╣-C6 alkyl and unsubstituted or substituted C╬╣-C6 alkoxy wherein the substituents on the alkoxy are one or more substituents independently selected from amino, C╬╣-C6 alkylamino, Ci-Ce dialkylamino, pyrrolidinyl, piperidinyl, azepinyl or morpholinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
R4 is C╬╣-C6 alkyl, aryl, or C7-C10 aralkyl; and R5and R6 are each independently selected from hydrogen,
Ci-Ce alkyl, C3-C10 alkenyl, Ci-Ce alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
13. A method of improving memory and/or of halting the progression of mental deterioration in an Alzheimer's disease patient which comprises administering to the subject a therapeutically effective amount of a compound to inhibit the binding of an amyloid beta peptide with alpha-7 nicotinic acetylcholine receptors.
14. The method of Claim 13, wherein the amyloid beta peptide is A╬▓^Γêæ and the alpha-7 nicotinic acetylcholine receptors are human alpha-7 nicotinic acetylcholine receptors.
15. The method of Claim 14, wherein the compound is of the formula I
Figure imgf000046_0001
wherein Ri is hydrogen or C╬╣-C4 alkyl; R2 is selected from hydrogen, C╬╣-C6 alkyl, aryl or C7-C10 aralkyl; R3 is selected from hydrogen, C╬╣-C6 alkyl, C3-C10 alkenyl,
C3-C8 cycloalkylCi-Ce alkyl, C╬╣-C6 alkoxycarbonylC╬╣-C6 alkyl, C╬╣-C6 alkylthio, heteroarylC╬╣-C alkyl, unsubstituted or substituted aryl or unsubstituted or substituted C7-C10 aralkyl wherein the substituent on the aryl or aralkyl are one or more substituents independently selected from the group consisting of halogen, hydroxy, C╬╣-C6 alkyl and unsubstituted or substituted C╬╣-C6 alkoxy wherein the substituents on the alkoxy are one or more substituents independently selected from amino, C╬╣-C6 alkylamino, C╬╣-C6 dialkylamino, pyrrolidinyl, piperidinyl, azepinyl or morpholinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
R4 is C╬╣-C6 alkyl, aryl, or C7-C10 aralkyl; and R5and R6 are each independently selected from hydrogen, C╬╣-C6 alkyl, C3-C10 alkenyl, C╬╣-C8 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
16. A method for identifying compounds which are useful for the treatment of neurodegenerative disorders involving screening test compounds for their ability to block the interaction of a peptide selected from the group consisting of
Figure imgf000046_0002
with nicotine acetylcholine receptors.
17. The method of Claim 16, wherein the nicotine acetycholine receptors are human alpha-7, human alpha-8, and/or human alpha-9 nicotinic acetylcholine receptors.
18. The method of Claim 17, wherein the nicotine acetylcholine receptors are human alpha-7 nicotine acetylcholine receptors.
19. The method of Claim 18, wherein the peptide is 125i l-A╬▓╬╣- 0.
20. A compound of the formula I:
Figure imgf000047_0001
wherein Ri is hydrogen or C╬╣-C4 alkyl; R2 is selected from hydrogen, C╬╣-C6 alkyl, aryl or C7-C10 aralkyl;
R3 is selected from hydrogen, C╬╣-C6 alkyl, C3-C10 alkenyl, C3-C8 cycloalkylCi-Ce alkyl, C╬╣-C6 alkoxycarbonylC╬╣-C6 alkyl, C╬╣-C6 alkylthio, heteroarylC╬╣-C4 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted C7-C╬╣o aralkyl wherein the substituent on the aryl or aralkyl are one or more substituents independently selected from the group consisting of halogen, hydroxy, C╬╣-C6 alkyl and unsubstituted or substituted C╬╣-C6 alkoxy wherein the substituents on the alkoxy are one or more substituents independently selected from amino, C╬╣-C6 alkylamino, C╬╣-C6 dialkylamino, pyrrolidinyl, piperidinyl, azepinyl or morpholinyl; or R2 and R3, together with the nitrogen to which they are attached, form a five or six-membered heterocyclic ring selected from pyrrolidinyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl;
R_t is C -C6 alkyl, aryl, or C7-C10 aralkyl; and R5 and R6 are each independently selected from hydrogen, C^Ce alkyl, C3-C10 alkenyl, C╬╣-C8 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
21. The compound of Claim 20, wherein
Ri is hydrogen;
R2 is selected from hydrogen or C╬╣-C4 alkyl;
R3 is selected from C1-C4 alkyl, C3-C10 alkenyl, C5-Ce cycloalkylC╬╣-C46 alkyl, d-C6 alkoxycarbonylC╬╣-C alkyl,
C╬╣-C6 alkylthio, heteroarylC╬╣-C alkyl, or unsubstituted or substituted C7-C10 aralkyl wherein the substituent on the aralkyl are one or two substituents independently selected from the group consisting of halogen, hydroxy, C╬╣-C4 alkyl and unsubstituted or substituted C╬╣-C4 alkoxy wherein the substituents on the alkoxy are one or two substituents independently selected from amino, C╬╣-C4 alkylamino, C╬╣-C4 dialkylamino, pyrrolidinyl, or piperidinyl; or
R2 and R3, together with the nitrogen to which they are attached, form a morpholinyl ring;
R4 is C╬╣-C4 alkyl; and R5and R6 are each independently selected from hydrogen,
C╬╣-C4 alkyl, C3-C6 alkenyl, C╬╣-C6 alkylcarbonyl, or diphenylphosphinyl; and pharmaceutically acceptable salts and prodrugs thereof.
22. The compound of Claim 20 of the formula
Figure imgf000048_0001
wherein Ri is hydrogen or C╬╣-C4 alkyl; R2 and R3 are each independently selected from hydrogen, C╬╣-C6 alkyl, aryl or C7-C10 aralkyl; and
R is C╬╣-C6 alkyl, aryl, or C7-C10 aralkyl; and pharmaceutically acceptable salts and prodrugs thereof.
23. A compound of Claim 22 which is 5,8-dihydroxy-fraπs-2-di(Λ/- propylamino)-3-methyl-1 ,2,3,4-tetrahydronaphthalene and pharmaceutically acceptable salts and prodrugs thereof.
24. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Claim 20.
25. A pharmaceutical composition made by mixing a compound of Claim 20 and a pharmaceutically acceptable carrier.
26. A process for making a pharmaceutical composition comprising mixing a compound of Claim 20 and a pharmaceutically acceptable carrier.
27. A method for diagnosing Alzheimer's disease, monitoring the progression and prognosis of Alzheimer's disease and/or monitoring the therapeutic efficacy of any intervention or treatment of Alzheimer's disease comprising:
(a) obtaining a test sample from a subject wherein the test sample comprises circulating blood cells and/or olfactory neuroepithelial neuronal cell bodies or their neuronal processes; and
(b) analyzing the test sample for interaction of an amyloid beta peptide with alpha-7 nicotinic acetylcholine receptors.
PCT/US1999/011702 1998-06-01 1999-05-27 Method for treating neurodegenerative disorders WO1999062505A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2000551761A JP4637351B2 (en) 1998-06-01 1999-05-27 Treatment of neurodegenerative diseases
EP99928342A EP1083889B1 (en) 1998-06-01 1999-05-27 Tetrahydronaphtalene compounds and their use for the treatment of neurodegenerative diseases
AT99928342T ATE255888T1 (en) 1998-06-01 1999-05-27 TETRAHYDRONAPHTALENE COMPOUNDS AND THEIR USE FOR THE TREATMENT OF NEURODEGENERATIVE DISEASES
DE69913520T DE69913520T2 (en) 1998-06-01 1999-05-27 TETRAHYDRONAPHTALENE COMPOUNDS AND THE USE THEREOF FOR THE TREATMENT OF NEURODEGENERATIVE DISEASES
AU45433/99A AU765142B2 (en) 1998-06-01 1999-05-27 Method for treating neurodegenerative disorders
DK99928342T DK1083889T3 (en) 1998-06-01 1999-05-27 Tetrahydronaphthalene compounds and their use in the treatment of neurodegenerative diseases
CA2333951A CA2333951C (en) 1998-06-01 1999-05-27 Method for treating neurodegenerative disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US8757798P 1998-06-01 1998-06-01
US60/087,577 1998-06-01

Publications (2)

Publication Number Publication Date
WO1999062505A2 true WO1999062505A2 (en) 1999-12-09
WO1999062505A3 WO1999062505A3 (en) 2000-04-06

Family

ID=22206004

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/011702 WO1999062505A2 (en) 1998-06-01 1999-05-27 Method for treating neurodegenerative disorders

Country Status (12)

Country Link
US (2) US6441049B2 (en)
EP (1) EP1083889B1 (en)
JP (1) JP4637351B2 (en)
AR (1) AR018431A1 (en)
AT (1) ATE255888T1 (en)
AU (1) AU765142B2 (en)
CA (1) CA2333951C (en)
DE (1) DE69913520T2 (en)
DK (1) DK1083889T3 (en)
ES (1) ES2212570T3 (en)
PT (1) PT1083889E (en)
WO (1) WO1999062505A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001070733A2 (en) * 2000-03-21 2001-09-27 Suntory Limited Di-substituted iminoheterocyclic compounds
WO2001073446A1 (en) * 2000-03-29 2001-10-04 Synaptica Limited Alpha 7 nicotinic receptor screening assays
WO2001085093A2 (en) * 1999-12-23 2001-11-15 Neurochem, Inc. Compounds and methods for modulating cerebral amyloid angiopathy
WO2002014351A2 (en) * 2000-08-14 2002-02-21 Ortho-Mcneil Pharmaceutical, Inc. α7 NICOTINIC RECEPTOR PEPTIDES AS LIGANDS FOR β AMYLOID P EPTIDES
EP1541547A1 (en) * 2003-12-11 2005-06-15 Newron Pharmaceuticals S.p.A. Hydroxylamine derivatives
WO2005092890A2 (en) 2004-03-25 2005-10-06 Memory Pharmaceuticals Corporation Indazoles, benzothiazoles, benzoisothiazoles, benzisoxazoles, and preparation and uses thereof
US7049337B2 (en) * 2000-05-19 2006-05-23 Wayne State University Derivatives of 2-aminotetralins and pharmaceutical analogs thereof exhibiting differential CNS receptor activity and behavior
US7067261B2 (en) 2001-12-14 2006-06-27 Targacept, Inc. Methods and compositions for treatment of central nervous system disorders
US7214686B2 (en) 1997-06-30 2007-05-08 Targacept, Inc. Pharmaceutical compositions and methods for effecting dopamine release
WO2008020131A1 (en) * 2006-08-18 2008-02-21 Les Laboratoires Servier Method of screening for compounds with anti-amyloid properties
US7402592B2 (en) 2003-10-15 2008-07-22 Targacept, Inc. Pharmaceutical compositions and methods for relieving pain and treating central nervous system disorders
WO2008087529A1 (en) 2007-01-16 2008-07-24 Siena Biotech S.P.A. Nicotinic acetylcholine receptor modulators
US7732607B2 (en) 2005-08-22 2010-06-08 Anatoly Mazurov Heteroaryl-substituted diazatricycloalkanes and methods of use thereof
EP2298758A1 (en) 2002-08-30 2011-03-23 Memory Pharmaceuticals Corporation Anabaseine derivatives useful in the treatment of neurodegenerative diseases
EP2305675A1 (en) 2002-09-25 2011-04-06 Memory Pharmaceuticals Corporation Indazoles, Benzothiazoles, and Benzoisothiazoles, and preparation and uses thereof
US7981906B2 (en) 2007-08-02 2011-07-19 Targacept, Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl-benzofuran-2-carboxamide, novel salt forms, and methods of use thereof
DE102010020553A1 (en) 2010-05-14 2011-11-17 Bayer Schering Pharma Aktiengesellschaft Substituted 8-alkoxy-2-aminotetralin derivatives and their use
US8114891B2 (en) 2007-10-01 2012-02-14 Comentis, Inc. 4-substituted quinuclidine derivatives, methods of production, pharmaceutical uses thereof
EP2633868A1 (en) 2008-02-13 2013-09-04 Targacept, Inc. Combination of alpha 7 nicotinic agonists and antipsychotics
WO2014012054A1 (en) 2012-07-13 2014-01-16 Pain Therapeutics, Inc. Alzheimer's disease assay in a living patent
US8884017B2 (en) 2001-12-27 2014-11-11 Bayer Intellectual Property Gmbh 2-heteroarylcarboxylic acid amides
US8901151B2 (en) 2009-01-26 2014-12-02 Targacept, Inc. Preparation and therapeutic applications of (2S, 3R)-N-2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]OCT-3-yl)-3,5-difluorobenzamide
US8981104B2 (en) 2012-07-20 2015-03-17 Bayer Pharma Aktiengesellschaft 5-aminotetrahydroquinoline-2-carboxylic acids and their use
US8987453B2 (en) 2006-11-06 2015-03-24 Abbvie Inc. Azaadamantane derivatives and methods of use
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9387203B2 (en) 2012-07-20 2016-07-12 Bayer Pharma Aktiengesellschaft Substituted aminoindane- and aminotetralinecarboxylic acids and the use thereof
US9434724B2 (en) 2014-07-11 2016-09-06 Alpharmagen, Llc Quinuclidines for modulating alpha 7 activity
US9464078B2 (en) 2010-09-23 2016-10-11 Abbvie Inc. Monohydrate of azaadamantane derivatives
US9724340B2 (en) 2015-07-31 2017-08-08 Attenua, Inc. Antitussive compositions and methods
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9923078D0 (en) * 1999-09-29 1999-12-01 Phytopharm Plc Sapogenin derivatives and their use
US8188043B2 (en) 1999-07-28 2012-05-29 The Board Of Trustees Of The Leland Stanford Jr. University Nicotine in therapeutic angiogenesis and vasculogenesis
MXPA02000927A (en) * 1999-07-28 2003-07-14 Johnson & Johnson Amine and amide derivatives as ligands for the neuropeptide y y5 receptor useful in the treatment of obesity and other disorders.
NZ531786A (en) 2001-10-02 2006-10-27 Upjohn Co Azabicyclic-substituted fused-heteroaryl compounds for the treatment of disease
WO2003042210A1 (en) 2001-11-09 2003-05-22 Pharmacia & Upjohn Company Azabicyclic-phenyl-fused-heterocyclic compounds and their use as alpha7 nachr ligands
US7045534B2 (en) * 2002-02-12 2006-05-16 The Board Of Trustees Of The Leland Stanford Junior University Methods of reducing angiogenesis
EP1476448A2 (en) * 2002-02-19 2004-11-17 PHARMACIA &amp; UPJOHN COMPANY Azabicyclic compounds for the treatment of disease
CA2476681A1 (en) * 2002-02-19 2003-08-28 Bruce N. Rogers Fused bicyclic-n-bridged-heteroaromatic carboxamides for the treatment of disease
WO2004039815A2 (en) * 2002-11-01 2004-05-13 Pharmacia & Upjohn Company Llc Compounds having both alpha7 nachr agonist and 5ht antagonist activity for treatment of cns diseases
US7238715B2 (en) * 2002-12-06 2007-07-03 The Feinstein Institute For Medical Research Treatment of pancreatitis using alpha 7 receptor-binding cholinergic agonists
EP2062595A1 (en) * 2002-12-06 2009-05-27 The Feinstein Institute for Medical Research Inhibition of inflammation using alpha 7 receptor-binding cholinergic agonists
WO2004074973A2 (en) * 2003-02-19 2004-09-02 Balin Advertising Ltd. A system for providing private offers to customers of web sites
AU2004251599A1 (en) * 2003-05-20 2005-01-06 Transtech Pharma, Inc. Rage antagonists as agents to reverse amyloidosis and diseases associated therewith
WO2005013910A2 (en) * 2003-08-07 2005-02-17 University Of South Florida Cholinergic modulation of microglial activation via alpha-7 nicotinic receptors
US20050170360A1 (en) * 2004-01-30 2005-08-04 Papke Roger L. Variant neuronal nicotinic alpha-7 receptor and methods of use
DE102004014841B4 (en) * 2004-03-24 2006-07-06 Schwarz Pharma Ag Use of rotigotine for the treatment and prevention of Parkinson-Plus syndrome
US9216966B2 (en) 2004-10-04 2015-12-22 John Manfredi Compounds for Alzheimer's disease
US7678823B2 (en) 2004-10-04 2010-03-16 Myriad Pharmaceticals, Inc. Compounds for alzheimer's disease
EP1928827B1 (en) * 2005-09-27 2012-08-22 Myriad Genetics, Inc. Pyrrole derivatives as therapeutic compounds
US8316104B2 (en) 2005-11-15 2012-11-20 California Institute Of Technology Method and apparatus for collaborative system
JP2009519962A (en) * 2005-12-19 2009-05-21 コメンティス,インコーポレーテッド Topical mecamylamine formulation for ophthalmic administration and use thereof
WO2007149392A1 (en) * 2006-06-16 2007-12-27 University Of Kentucky Mono quaternary ammonium salts and methods for modulating neuronal nicotinic acetylcholine receptors
EP2059238A4 (en) * 2006-09-07 2011-04-06 Myriad Genetics Inc Therapeutic compounds for diseases and disorders
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
US8580833B2 (en) 2009-09-30 2013-11-12 Transtech Pharma, Inc. Substituted imidazole derivatives and methods of use thereof
SA111320455B1 (en) 2010-05-17 2014-10-16 Envivo Pharmaceuticals Inc Acrystalline form of (R)-7-chloro-N-(quinuclidin-3-‎Yl)benzo[B]thiophene-2-carboxamide hydrochloride ‎monohydrate
FR2966827B1 (en) 2010-10-27 2014-08-22 Kimonella Ventures Ltd PEPTIDE COMPOUND USEFUL FOR INHIBITING AMYLOID PLATE FORMATION
EP2808679A4 (en) * 2012-01-27 2015-01-14 Panasonic Healthcare Co Ltd Diagnosis method and diagnosis system for alzheimer's disease
EP3666272A1 (en) 2012-05-08 2020-06-17 Forum Pharmaceuticals Inc. Use of encenicline in the treatment of cognitive impairment, alzheimer's disease, memory deficit
DK2882428T3 (en) * 2012-07-13 2019-04-15 Pain Therapeutics Inc PROCEDURE TO INHIBIT TAU PHOSPHORIZATION
US9717710B2 (en) 2012-10-05 2017-08-01 Vtv Therapeutics Llc Treatment of mild and moderate Alzheimer's disease
GB201303507D0 (en) * 2013-02-27 2013-04-10 Hollfelder Florian Assays
WO2018067662A1 (en) * 2016-10-04 2018-04-12 University Of Miami Protein amyloidogenesis and related methods
WO2019190822A1 (en) 2018-03-28 2019-10-03 Vtv Therapeutics Llc Crystalline forms of [3-(4- {2-butyl-1-[4-(4-chloro-phenoxy)-phenyl]-1h-imidazol-4-yl} -phenoxy)-propyl]-diethyl-amine
WO2019190823A1 (en) 2018-03-28 2019-10-03 Vtv Therapeutics Llc Pharmaceutically acceptable salts of [3-(4- {2-butyl-1-[4-(4-chlorophenoxy)-phenyl]-1h-imidazol-4-yl} -phenoxy)-propyl]-diethyl-amine
WO2020076668A1 (en) 2018-10-10 2020-04-16 Vtv Therapeutics Llc Metabolites of [3-(4-{2-butyl-l-[4-(4-chloro-phenoxy)-phenyl]-lh-imidazol-4-yl } -phen ox y)-prop yl] -diethyl-amine
CU24626B1 (en) 2019-12-26 2022-11-07 Centro Nac De Biopreparados PHARMACEUTICAL COMPOSITION BASED ON PROTEINS WITH NEUROPROTECTIVE, IMMUNOMODULATORY, ANTI-INFLAMMATORY AND ANTIMICROBIAL ACTIVITIES

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH637363A5 (en) * 1977-11-24 1983-07-29 Sandoz Ag Process for preparing novel 2-aminotetralins
EP0360077A2 (en) * 1988-09-20 1990-03-28 Troponwerke GmbH &amp; Co. KG Use of serotonin antagonists in the treatment of apoplexia cerebri

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5554601A (en) 1993-11-05 1996-09-10 University Of Florida Methods for neuroprotection
US6689574B1 (en) * 1997-10-07 2004-02-10 Merck & Co., Inc. Assays for nuclear receptor agonists and antagonists using fluorescence resonance energy transfer
US6329159B1 (en) * 1999-03-11 2001-12-11 Millennium Pharmaceuticals, Inc. Anti-GPR-9-6 antibodies and methods of identifying agents which modulate GPR-9-6 function
US6689578B2 (en) * 2001-12-06 2004-02-10 Paradigm Genetics, Inc. Methods for the identification of inhibitors of 5-aminolevulinate synthase as antibiotics

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH637363A5 (en) * 1977-11-24 1983-07-29 Sandoz Ag Process for preparing novel 2-aminotetralins
EP0360077A2 (en) * 1988-09-20 1990-03-28 Troponwerke GmbH &amp; Co. KG Use of serotonin antagonists in the treatment of apoplexia cerebri

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
AULD ET AL.: "Beta-Amyloid peptides as direct cholinergic neuromodulatorys : a missing link ? " TRENDS NEUROSCI., vol. 21, no. 1, January 1998 (1998-01), pages 43-49, XP000853850 cited in the application *
ITOH ET AL.: "Dysfunction of cholinergic and dopaminergic neuronal systems in beta-amyloid protein-infused rats" J. NEUROCHEM., vol. 66, 1996, pages 1113-1117, XP002916315 *
KIHARA ET AL.: "Neuroprotective effect of nicotine-related substances against beta-amyloid cytotoxicity" NEUROSCI. RES., vol. suppl., no. 21, 1997, page s158 XP002128692 *
KIHARA ET AL.: "Nicotinic receptor stimulation protects neurons against beta-amyloid toxicity" ANN. NEUROL., vol. 42, no. 2, August 1997 (1997-08), pages 159-163, XP000853838 cited in the application *
MAURICE ET AL.: "Amnesia induced in mice by centrally administered beta-amyloid peptides involves cholinergic dysfunction" BRAIN RES., vol. 706, no. 2, 1996, pages 181-193, XP002128693 *
MCDERMED ET AL.: "Synthesis and pharmacology of some 2-aminotetralins. Dopamine receptor agonists" J. MED. CHEM. , vol. 18, no. 4, 1975, pages 362-367, XP002067369 *
SALOMON ET AL.: "Nicotine inhibits amyloid formation by the beta-peptide" BIOCHEMISTRY, vol. 35, no. 42, 1996, pages 13568-13578, XP000857787 *
SJ\BERG ET AL.: "Neuronal nicotinic receptor activation : a promising strategy for the treatment of Alzheimer's disease ?" INT. J. GERIATRIC PSYCHOPHARMACOLOGY, vol. 1, no. 3, 1998, pages 145-149, XP002128695 *
ZAMANI ET AL.: "Nicotine modulates the neurtoxic effect of beta-amyloid protein(25-35) in hippocampal cultures" NEUROREPORT, vol. 8, no. 2, 20 January 1997 (1997-01-20), pages 513-517, XP002128694 *

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7214686B2 (en) 1997-06-30 2007-05-08 Targacept, Inc. Pharmaceutical compositions and methods for effecting dopamine release
WO2001085093A3 (en) * 1999-12-23 2002-08-29 Neurochem Inc Compounds and methods for modulating cerebral amyloid angiopathy
WO2001085093A2 (en) * 1999-12-23 2001-11-15 Neurochem, Inc. Compounds and methods for modulating cerebral amyloid angiopathy
US6670399B2 (en) 1999-12-23 2003-12-30 Neurochem (International) Limited Compounds and methods for modulating cerebral amyloid angiopathy
WO2001070733A2 (en) * 2000-03-21 2001-09-27 Suntory Limited Di-substituted iminoheterocyclic compounds
US6900202B2 (en) 2000-03-21 2005-05-31 Daiichi Suntory Pharma Co., Ltd. Di-substituted imineheterocyclic compounds
WO2001070733A3 (en) * 2000-03-21 2002-05-02 Suntory Ltd Di-substituted iminoheterocyclic compounds
WO2001073446A1 (en) * 2000-03-29 2001-10-04 Synaptica Limited Alpha 7 nicotinic receptor screening assays
US7049337B2 (en) * 2000-05-19 2006-05-23 Wayne State University Derivatives of 2-aminotetralins and pharmaceutical analogs thereof exhibiting differential CNS receptor activity and behavior
WO2002014351A2 (en) * 2000-08-14 2002-02-21 Ortho-Mcneil Pharmaceutical, Inc. α7 NICOTINIC RECEPTOR PEPTIDES AS LIGANDS FOR β AMYLOID P EPTIDES
WO2002014351A3 (en) * 2000-08-14 2003-08-14 Ortho Mcneil Pharm Inc α7 NICOTINIC RECEPTOR PEPTIDES AS LIGANDS FOR β AMYLOID P EPTIDES
US7067261B2 (en) 2001-12-14 2006-06-27 Targacept, Inc. Methods and compositions for treatment of central nervous system disorders
US8884017B2 (en) 2001-12-27 2014-11-11 Bayer Intellectual Property Gmbh 2-heteroarylcarboxylic acid amides
EP2305664A1 (en) 2002-08-30 2011-04-06 Memory Pharmaceuticals Corporation Anabaseine derivatives useful in the treatment of neurodegeneratives diseases
EP2298759A1 (en) 2002-08-30 2011-03-23 Memory Pharmaceuticals Corporation Anabaseine derivatives useful in the treatment of neurodegenerative diseases
EP2298758A1 (en) 2002-08-30 2011-03-23 Memory Pharmaceuticals Corporation Anabaseine derivatives useful in the treatment of neurodegenerative diseases
EP2305675A1 (en) 2002-09-25 2011-04-06 Memory Pharmaceuticals Corporation Indazoles, Benzothiazoles, and Benzoisothiazoles, and preparation and uses thereof
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US7897611B2 (en) 2003-10-15 2011-03-01 Targacept, Inc. Pharmaceutical compositions and methods for relieving pain and treating central nervous system disorders
US7402592B2 (en) 2003-10-15 2008-07-22 Targacept, Inc. Pharmaceutical compositions and methods for relieving pain and treating central nervous system disorders
WO2005058800A1 (en) * 2003-12-11 2005-06-30 Newron Pharmaceuticals S.P.A. Hydroxylamine derivatives
US7763751B2 (en) 2003-12-11 2010-07-27 Newron Pharmaceuticals S.P.A. Hydroxylamine derivatives
EP1541547A1 (en) * 2003-12-11 2005-06-15 Newron Pharmaceuticals S.p.A. Hydroxylamine derivatives
WO2005092890A2 (en) 2004-03-25 2005-10-06 Memory Pharmaceuticals Corporation Indazoles, benzothiazoles, benzoisothiazoles, benzisoxazoles, and preparation and uses thereof
EP2336128A1 (en) 2004-03-25 2011-06-22 Memory Pharmaceuticals Corporation Benzoisothiazoles and preparation and uses thereof
US7732607B2 (en) 2005-08-22 2010-06-08 Anatoly Mazurov Heteroaryl-substituted diazatricycloalkanes and methods of use thereof
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
FR2905009A1 (en) * 2006-08-18 2008-02-22 Servier Lab METHOD OF SCREENING COMPOUNDS WITH ANTI-AMYLOID PROPERTIES
WO2008020131A1 (en) * 2006-08-18 2008-02-21 Les Laboratoires Servier Method of screening for compounds with anti-amyloid properties
US8987453B2 (en) 2006-11-06 2015-03-24 Abbvie Inc. Azaadamantane derivatives and methods of use
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US8163729B2 (en) 2007-01-16 2012-04-24 Wyeth Modulators of α7 nicotinic acetylcholine receptors and therapeutic uses thereof
WO2008087529A1 (en) 2007-01-16 2008-07-24 Siena Biotech S.P.A. Nicotinic acetylcholine receptor modulators
US8119659B2 (en) 2007-08-02 2012-02-21 Targacept, Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofuran-2-carboxamide, novel salt forms, and methods of use thereof
US8541446B2 (en) 2007-08-02 2013-09-24 Targacept, Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofurn-2-carboxamide, novel salt forms, and methods of use thereof
EP2465501A1 (en) 2007-08-02 2012-06-20 Targacept Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofuran-2-carboxamide, novel salt forms, and methods of preparation
EP2431037A1 (en) 2007-08-02 2012-03-21 Targacept, Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofuran-2-carboxamide, novel salt forms, and methods of use thereof
US8846715B2 (en) 2007-08-02 2014-09-30 Targacept, Inc. (2S,3R)-N-(2((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofuran-2-carboxamide novel salt forms, and methods of use thereof
US7981906B2 (en) 2007-08-02 2011-07-19 Targacept, Inc. (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl-benzofuran-2-carboxamide, novel salt forms, and methods of use thereof
US8114891B2 (en) 2007-10-01 2012-02-14 Comentis, Inc. 4-substituted quinuclidine derivatives, methods of production, pharmaceutical uses thereof
EP2633868A1 (en) 2008-02-13 2013-09-04 Targacept, Inc. Combination of alpha 7 nicotinic agonists and antipsychotics
US8901151B2 (en) 2009-01-26 2014-12-02 Targacept, Inc. Preparation and therapeutic applications of (2S, 3R)-N-2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]OCT-3-yl)-3,5-difluorobenzamide
US9173876B2 (en) 2009-01-26 2015-11-03 Targacept, Inc. Preparation and therapeutic applications of (2S,3R)-N-2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)-3,5-difluorobenzamide
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
DE102010020553A1 (en) 2010-05-14 2011-11-17 Bayer Schering Pharma Aktiengesellschaft Substituted 8-alkoxy-2-aminotetralin derivatives and their use
WO2011141409A1 (en) 2010-05-14 2011-11-17 Bayer Pharma Aktiengesellschaft Substituted 8-alkoxy-2-aminotetralin derivatives, and use thereof
US8673903B2 (en) 2010-05-14 2014-03-18 Bayer Intellectual Property Gmbh Substituted 8-alkoxy-2-aminotetralin derivatives, and use thereof
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9464078B2 (en) 2010-09-23 2016-10-11 Abbvie Inc. Monohydrate of azaadamantane derivatives
EP2872899A4 (en) * 2012-07-13 2015-12-30 Pain Therapeutics Inc Alzheimer's disease assay in a living patient
WO2014012054A1 (en) 2012-07-13 2014-01-16 Pain Therapeutics, Inc. Alzheimer's disease assay in a living patent
US9688636B2 (en) 2012-07-20 2017-06-27 Bayer Pharma Aktiengesellschaft 5-aminotetrahydroquinoline-2-carboxylic acids and their use
US10053428B2 (en) 2012-07-20 2018-08-21 Bayer Pharma Aktiengesellschaft 5-aminotetrahydroquinoline-2-carboxylic acids and their use
US9387203B2 (en) 2012-07-20 2016-07-12 Bayer Pharma Aktiengesellschaft Substituted aminoindane- and aminotetralinecarboxylic acids and the use thereof
US8981104B2 (en) 2012-07-20 2015-03-17 Bayer Pharma Aktiengesellschaft 5-aminotetrahydroquinoline-2-carboxylic acids and their use
US9434724B2 (en) 2014-07-11 2016-09-06 Alpharmagen, Llc Quinuclidines for modulating alpha 7 activity
US9724340B2 (en) 2015-07-31 2017-08-08 Attenua, Inc. Antitussive compositions and methods

Also Published As

Publication number Publication date
DK1083889T3 (en) 2004-04-13
EP1083889A2 (en) 2001-03-21
CA2333951A1 (en) 1999-12-09
ES2212570T3 (en) 2004-07-16
JP4637351B2 (en) 2011-02-23
ATE255888T1 (en) 2003-12-15
US7018797B2 (en) 2006-03-28
PT1083889E (en) 2004-04-30
EP1083889B1 (en) 2003-12-10
US6441049B2 (en) 2002-08-27
WO1999062505A3 (en) 2000-04-06
US20020013374A1 (en) 2002-01-31
AR018431A1 (en) 2001-11-14
DE69913520T2 (en) 2004-11-25
AU4543399A (en) 1999-12-20
AU765142B2 (en) 2003-09-11
CA2333951C (en) 2012-02-28
US20030130165A1 (en) 2003-07-10
JP2002516853A (en) 2002-06-11
DE69913520D1 (en) 2004-01-22

Similar Documents

Publication Publication Date Title
EP1083889B1 (en) Tetrahydronaphtalene compounds and their use for the treatment of neurodegenerative diseases
US9708309B2 (en) Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
KR101486026B1 (en) Derivatives of fluorene, anthracene, xanthene, dibenzosuberone and acridine and uses thereof
UA76571C2 (en) Derivatives of 1-alkyl-1-azoniabicyclo [2.2.2.]octanecarbamate and use thereof as antagonists of muscarin receptor
AU5448890A (en) Pcp receptor ligands and the use thereof
PT85379B (en) PROCESS FOR THE PREPARATION OF AMIDA DERIVATIVES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
AU2018216040A1 (en) OGA inhibitor compounds
WO2008109840A1 (en) Tricyclic compounds useful in treating iron disorders
US20220106330A1 (en) Solid forms of a promoter of spinogenesis
US6498196B1 (en) Compounds useful in pain management
JP2014511373A (en) Compounds isolated from turmeric oil and methods of use thereof
KR20090019847A (en) Pyrrolidine derivatives having activity at the glyt1 transporter
JP2007534616A (en) Tacrine derivatives as inhibitors of acetylcholinesterase
MXPA00011847A (en) Method for treating neurodegenerative disorders
US5688789A (en) PCP receptor ligands and the use thereof
US5132313A (en) Non-competitive NMDA receptor antagonists and methods for their use
CA3102903A1 (en) Oga inhibitor compounds
CN116531381A (en) Sulfur-containing compound and pharmaceutical composition thereof
EP1140815A1 (en) Novel thio-aminotetralin compounds useful in pain management
JP2023507180A (en) OGA inhibitor compounds
CA3103910A1 (en) Oga inhibitor compounds
EP0587670A1 (en) Pcp receptor ligands and the use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM HR HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM HR HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 45433/99

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2000 551761

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2333951

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2000/011847

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1999928342

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999928342

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1999928342

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 45433/99

Country of ref document: AU