US20020013315A1 - Gamma secretase inhibitors - Google Patents

Gamma secretase inhibitors Download PDF

Info

Publication number
US20020013315A1
US20020013315A1 US09/896,296 US89629601A US2002013315A1 US 20020013315 A1 US20020013315 A1 US 20020013315A1 US 89629601 A US89629601 A US 89629601A US 2002013315 A1 US2002013315 A1 US 2002013315A1
Authority
US
United States
Prior art keywords
compound according
alkyl
pharmaceutically acceptable
alkoxycarbonyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US09/896,296
Other versions
US6448229B2 (en
Inventor
Martin Teall
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Organon Pharma UK Ltd
Zimmer Spine Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Assigned to SULZER SPINE-TECH INC. reassignment SULZER SPINE-TECH INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SEME, STEVEN J., HANSON, DAVID A., LONGHINI, ROSS A.
Publication of US20020013315A1 publication Critical patent/US20020013315A1/en
Assigned to MERCK SHARP & DOHME LTD. reassignment MERCK SHARP & DOHME LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TEALL, MARTIN RICHARD
Application granted granted Critical
Publication of US6448229B2 publication Critical patent/US6448229B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • 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
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06191Dipeptides containing heteroatoms different from O, S, or N
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to compounds, their salts, pharmaceutical compositions comprising them, processes for making them and their use in treating Alzheimer's Disease.
  • AD Alzheimer's Disease
  • the rate of amyloid accumulation is a combination of the rates of formation, aggregation and egress from the brain. It is generally accepted that the main constituent of amyloid plaques is the 4 kD amyloid protein ( ⁇ A4, also referred to as A ⁇ , ⁇ -protein and ⁇ AP) which is a proteolytic product of a precursor protein of much larger size.
  • ⁇ A4 also referred to as A ⁇ , ⁇ -protein and ⁇ AP
  • the ragged NH 2 - and COOH-termini of the native A ⁇ amyloid indicates that a complex mechanism of proteolysis is involved in its biogenesis.
  • amyloid precursor protein (APP or A ⁇ PP) has a receptor-like structure with a large ectodomain, a membrane spanning region and a short cytoplasmic tail.
  • Different isoforms of APP result from the alternative splicing of three exons in a single gene and have 695,751 and 770 amino acids respectively.
  • the A ⁇ domain encompasses parts of both extra-cellular and transmembrane domains of APP, thus its release implies the existence of two distinct proteolytic events to generate its NH 2 - and COOH-termini. At least two secretory mechanisms exist which release APP from the membrane and generate the soluble, COOH-truncated forms of APP (APPs). Proteases which release APP and its fragments from the membrane are termed “secretases”. Most APPs is released by a putative ⁇ -secretase which cleaves within the A ⁇ domain (between residues Lys 16 and Leu 17 ) to release ( ⁇ -APPs and precludes the release of intact A ⁇ .
  • a minor portion of APPs is released by a ⁇ -secretase, which cleaves near the NH 2 -terminus of A ⁇ and produces COOH-terminal fragments (CTFs) which contain the whole A ⁇ domain. Finding these fragments in the extracellular compartment suggests that another proteolytic activity ( ⁇ -secretase) exists under normal conditions which can generate the COOH-terminus of A ⁇ .
  • ⁇ secretase inhibitors to inhibit the functioning of presinilins, which have been proposed as candidates ⁇ secretases, has raised questions over their suitability.
  • signalling through the Notch pathway important during embryonic development and in haematopoeisis, requires the presenilin-dependent proteolytic release of the Notch intracellular domain (NICD).
  • Notch intracellular domain NBD
  • the present invention provides a compound of formula I or a pharmaceutically acceptable salt thereof:
  • X is CH 2 , oxygen or sulphur
  • Ar is phenyl optionally substituted by one, two or three substituents chosen from halogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, cyano, nitro, NR 1 R 2 where R 1 and R 2 are independently hydrogen or C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyloxy, C 2-6 alkynyloxy, thiol, C 1-6 alkylthio, C 2-6 alkenylthio, C 2-6 alkynylthio, C 1-6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 haloalkyl, C 2-6 haloalkenyl and C 2-6 haloalkynyl.
  • X is oxygen
  • Ar is optionally substituted by one, two or three substituents chosen from halogen, C 1-6 alkyl, hydroxy, amino, C 1-6 alkoxy, thiol, C 1-6 alkoxycarbonyl and C 1-6 haloalkyl.
  • Ar is optionally substituted by one substituent chosen from halogen, C 1-4 alkyl, hydroxy, amino, C 1-4 alkoxy, thiol, C 1-4 alkoxycarbonyl and C 1-4 haloalkyl.
  • Ar is unsubstituted.
  • a specific Example of the present invention is: 2-[2-(3,5-difluorophenyl)acetylamino]-N- ⁇ phenyl[(4-phenylmorpholin-2-ylmethyl)carbamoyl]methyl ⁇ propionamide and the pharmaceutically acceptable salts thereof.
  • C 1-6 alkyl includes methyl and ethyl groups, and straight-chained and branched propyl, butyl, pentyl and hexyl groups. Particular alkyl groups are methyl, ethyl, n-propyl, isopropyl and t-butyl. Derived expressions such as “C 1-4 alkyl”, “C 2-6 alkenyl” and “C 2-6 alkynyl” are to be construed in an analogous manner.
  • halogen as used herein includes fluorine, chlorine, bromine and iodine, of which fluorine and chlorine are preferred.
  • C 1-6 alkoxy includes methoxy and ethoxy groups, and straight-chained, branched and cyclic propoxy and butoxy groups, including cyclopropylmethoxy.
  • C 2-6 alkynyloxy “C 2-6 alkenyloxy”, “C 1-6 alkylthio”, “C 2-6 alkenylthio”, “C 2-6 alkynylthio” and “C 1-4 alkoxy” are to be construed in analogous manner.
  • C 1-6 alkoxycarbonyl includes methoxycarbonyl and alkoxycarbonyl groups and straight-chained, branched and cyclic propoxycarbonyl and butoxycarbonyl groups, including cyclopropylmethoxycarbonyl. “C 1-4 alkoxycarbonyl” is to be construed in analogous manner.
  • Examples of pharmaceutically acceptable salts are hydrochlorides, sulfates, citrates, tartrates, acetates, methanesulfonates, phosphates, oxalates and benzoates.
  • the compounds of the present invention have an activity as inhibitors of ⁇ secretase.
  • the compounds of the invention inhibit proteolysis of PS-1.
  • compositions comprising one or more compounds of this invention and a pharmaceutically acceptable carrier.
  • these compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, transdermal patches, auto-injector devices or suppositories; for oral, parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation.
  • a pharmaceutical carrier e.g.
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • Typical unit dosage forms contain from 1 to 100 mg, for example 1, 2, 5, 10, 25, 50 or 100 mg, of the active ingredient.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • the present invention also provides a compound of the invention or a pharmaceutically acceptable salt thereof for use in a method of treatment of the human body.
  • the treatment is for a condition associated with the deposition of ⁇ -amyloid.
  • the condition is a neurological disease having associated ⁇ -amyloid deposition such as Alzheimer's disease.
  • this treatment occurs without inhibiting Notch signalling.
  • the present invention further provides the use of a compound of the present invention or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating or preventing Alzheimer's disease, preferably without inhibiting Notch signalling.
  • This method of treatment preferably does not inhibit Notch signalling.
  • a suitable dosage level is about 0.01 to 250 mg/kg per day, preferably about 0.01 to 100 mg/kg per day, and especially about 0.01 to 5 mg/kg of body weight per day.
  • the compounds may be administered on a regimen of 1 to 4 times per day. In some cases, however, dosage outside these limits may be used.
  • X and Ar are as defined above, generally in the presence of a coupling agent such as 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, and hydroxybenzotriazole, generally in the presence of a base such as triethylamine, and a solvent such as tetrahydrofuran, for about 18 h at room temperature.
  • a coupling agent such as 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, and hydroxybenzotriazole
  • a typical assay which can be used to determine the level of activity of compounds of the present invention is as follows:
  • redox dye reduction To determine if compounds are cytotoxic cell viability following compound administration is assessed by the use of redox dye reduction.
  • a typical example is a combination of redox dye MTS (Promega) and the electron coupling reagent PES. This mixture is made up according to the manufacturer's instructions and left at room temperature.
  • the Example of the present invention had an ED 50 of less than 500 nM in the above assay.
  • Xenopus embryos were cultured in ⁇ fraction (1/10) ⁇ NAM [Beck & Slack, Development, 126, 1611-1620, 1999] supplemented with protease inhibitors (200 ⁇ stock in DMSO) from stage 15 [P. D. Nieuwkoop and J. Faber, Normal table of Xenopus laevis, Daudin 1967, Reprinted Garland 1994] (24 hours), to stage 40 (3 days).
  • Control siblings were incubated in ⁇ fraction (1/10) ⁇ NAM containing 0.05% DMSO. At 3 days they were fixed in batches of 20 and measured using a graduated eyepiece on a Wild dissecting microscope set to 25 ⁇ .
  • Example of the present invention did not significantly inhibit tail bud formation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides compounds of formula (I) and pharmaceutically acceptable salts thereof:
Figure US20020013315A1-20020131-C00001
wherein X is CH2, O or S. The compounds inhibit gamma secretase without affecting Notch signalling, and hence find use in the treatment or prevention of Alzheimer's disease.

Description

  • The present invention relates to compounds, their salts, pharmaceutical compositions comprising them, processes for making them and their use in treating Alzheimer's Disease. [0001]
  • Alzheimer's Disease (AD) is characterized by the abnormal deposition of amyloid in the brain in the form of extra-cellular plaques and intra-cellular neurofibrillary tangles. The rate of amyloid accumulation is a combination of the rates of formation, aggregation and egress from the brain. It is generally accepted that the main constituent of amyloid plaques is the 4 kD amyloid protein (βA4, also referred to as Aβ,β-protein and βAP) which is a proteolytic product of a precursor protein of much larger size. The ragged NH[0002] 2- and COOH-termini of the native Aβ amyloid indicates that a complex mechanism of proteolysis is involved in its biogenesis.
  • The amyloid precursor protein (APP or AβPP) has a receptor-like structure with a large ectodomain, a membrane spanning region and a short cytoplasmic tail. Different isoforms of APP result from the alternative splicing of three exons in a single gene and have 695,751 and 770 amino acids respectively. [0003]
  • The Aβ domain encompasses parts of both extra-cellular and transmembrane domains of APP, thus its release implies the existence of two distinct proteolytic events to generate its NH[0004] 2- and COOH-termini. At least two secretory mechanisms exist which release APP from the membrane and generate the soluble, COOH-truncated forms of APP (APPs). Proteases which release APP and its fragments from the membrane are termed “secretases”. Most APPs is released by a putative α-secretase which cleaves within the Aβdomain (between residues Lys16 and Leu17) to release (α-APPs and precludes the release of intact Aβ. A minor portion of APPs is released by a β-secretase, which cleaves near the NH2-terminus of Aβ and produces COOH-terminal fragments (CTFs) which contain the whole Aβ domain. Finding these fragments in the extracellular compartment suggests that another proteolytic activity (γ-secretase) exists under normal conditions which can generate the COOH-terminus of Aβ.
  • It is believed that γ secretase itself depends for its activity on the presence of presenilin-1. In a manner that is not fully understood presenilin-1 appears to undergo autocleavage. [0005]
  • The potential of γ secretase inhibitors to inhibit the functioning of presinilins, which have been proposed as candidates γ secretases, has raised questions over their suitability. In particular, signalling through the Notch pathway, important during embryonic development and in haematopoeisis, requires the presenilin-dependent proteolytic release of the Notch intracellular domain (NICD). Using a novel Xenopus developmental assay for Notch activity it has surprisingly been found that the present γ secretase inhibitors do not prevent Notch signalling in-vivo. [0006]
  • The present compounds are structurally related to those disclosed in WO-A-9822494. However there is no discussion in that document of the problem of interference of Notch signalling nor any suggestion of how the provision of γ-secretase inhibitors that do not inhibit Notch signalling may be achieved. [0007]
  • Accordingly, the present invention provides a compound of formula I or a pharmaceutically acceptable salt thereof: [0008]
    Figure US20020013315A1-20020131-C00002
  • wherein: X is CH[0009] 2, oxygen or sulphur; and
  • Ar is phenyl optionally substituted by one, two or three substituents chosen from halogen, C[0010] 1-6alkyl, C2-6alkenyl, C2-6alkynyl, hydroxy, cyano, nitro, NR1R2 where R1 and R2 are independently hydrogen or C1-6alkyl, C1-6alkoxy, C2-6alkenyloxy, C2-6alkynyloxy, thiol, C1-6alkylthio, C2-6alkenylthio, C2-6alkynylthio, C1-6alkylcarbonyl, C1-6alkoxycarbonyl, C1-6haloalkyl, C2-6haloalkenyl and C2-6haloalkynyl.
  • Preferably X is oxygen. [0011]
  • Preferably Ar is optionally substituted by one, two or three substituents chosen from halogen, C[0012] 1-6alkyl, hydroxy, amino, C1-6alkoxy, thiol, C1-6alkoxycarbonyl and C1-6haloalkyl.
  • More preferably Ar is optionally substituted by one substituent chosen from halogen, C[0013] 1-4alkyl, hydroxy, amino, C1-4alkoxy, thiol, C1-4alkoxycarbonyl and C1-4haloalkyl.
  • In one embodiment Ar is unsubstituted. [0014]
  • A specific Example of the present invention is: 2-[2-(3,5-difluorophenyl)acetylamino]-N-{phenyl[(4-phenylmorpholin-2-ylmethyl)carbamoyl]methyl}propionamide and the pharmaceutically acceptable salts thereof. [0015]
  • As used herein, the expression “C[0016] 1-6alkyl” includes methyl and ethyl groups, and straight-chained and branched propyl, butyl, pentyl and hexyl groups. Particular alkyl groups are methyl, ethyl, n-propyl, isopropyl and t-butyl. Derived expressions such as “C1-4alkyl”, “C2-6alkenyl” and “C2-6alkynyl” are to be construed in an analogous manner.
  • The term “halogen” as used herein includes fluorine, chlorine, bromine and iodine, of which fluorine and chlorine are preferred. [0017]
  • As used herein the term “C[0018] 1-6alkoxy” includes methoxy and ethoxy groups, and straight-chained, branched and cyclic propoxy and butoxy groups, including cyclopropylmethoxy. “C2-6alkynyloxy”, “C2-6alkenyloxy”, “C1-6alkylthio”, “C2-6alkenylthio”, “C2-6alkynylthio” and “C1-4alkoxy” are to be construed in analogous manner.
  • As used herein the term “C[0019] 1-6alkoxycarbonyl” includes methoxycarbonyl and alkoxycarbonyl groups and straight-chained, branched and cyclic propoxycarbonyl and butoxycarbonyl groups, including cyclopropylmethoxycarbonyl. “C1-4alkoxycarbonyl” is to be construed in analogous manner.
  • Examples of pharmaceutically acceptable salts are hydrochlorides, sulfates, citrates, tartrates, acetates, methanesulfonates, phosphates, oxalates and benzoates.[0020]
  • The compounds of the present invention have an activity as inhibitors of γsecretase. In a preferred embodiment the compounds of the invention inhibit proteolysis of PS-1. [0021]
  • The invention also provides pharmaceutical compositions comprising one or more compounds of this invention and a pharmaceutically acceptable carrier. Preferably these compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, transdermal patches, auto-injector devices or suppositories; for oral, parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation. For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums or surfactants such as sorbitan monooleate, polyethylene glycel, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. Typical unit dosage forms contain from 1 to 100 mg, for example 1, 2, 5, 10, 25, 50 or 100 mg, of the active ingredient. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. [0022]
  • The present invention also provides a compound of the invention or a pharmaceutically acceptable salt thereof for use in a method of treatment of the human body. Preferably the treatment is for a condition associated with the deposition of β-amyloid. Preferably the condition is a neurological disease having associated β-amyloid deposition such as Alzheimer's disease. Preferably this treatment occurs without inhibiting Notch signalling. [0023]
  • The present invention further provides the use of a compound of the present invention or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating or preventing Alzheimer's disease, preferably without inhibiting Notch signalling. [0024]
  • Also disclosed is a method of treatment of a subject suffering from or prone to Alzheimer's disease which comprises administering to that subject an effective amount of a compound according to the present invention or a pharmaceutically acceptable salt thereof. This method of treatment preferably does not inhibit Notch signalling. [0025]
  • The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin. [0026]
  • For treating or preventing Alzheimer's Disease, a suitable dosage level is about 0.01 to 250 mg/kg per day, preferably about 0.01 to 100 mg/kg per day, and especially about 0.01 to 5 mg/kg of body weight per day. The compounds may be administered on a regimen of 1 to 4 times per day. In some cases, however, dosage outside these limits may be used. [0027]
  • There is also provided a process for producing a compound of formula I or a pharmaceutically acceptable salt thereof which comprises reacting a compound of formula II with a compound of formula III: [0028]
    Figure US20020013315A1-20020131-C00003
  • wherein X and Ar are as defined above, generally in the presence of a coupling agent such as 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, and hydroxybenzotriazole, generally in the presence of a base such as triethylamine, and a solvent such as tetrahydrofuran, for about 18 h at room temperature. [0029]
  • Compounds of formulae II and III are commercially available or can be made by known methods from commercially available compounds. [0030]
  • It will be understood that any compound of formula I initially obtained from the above process may, where appropriate, subsequently be elaborated into a further compound of formula I by techniques known from the art. [0031]
  • It will also be appreciated that where more than one isomer can be obtained from a reaction then the resulting mixture of isomers can be separated by conventional means. [0032]
  • Where the above-described process for the preparation of the compounds according to the invention gives rise to mixtures of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography. The novel compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. [0033]
  • During any of the above synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in [0034] Protective Groups in Organic Chemistry, ed. J. F. W. McOmie, Plenum Press, 1973; and T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • A typical assay which can be used to determine the level of activity of compounds of the present invention is as follows: [0035]
  • (1) Mouse neuroblastoma neuro 2[0036] a cells expressing human app695 are cultured at 50-70% confluency in the presence of sterile 10 mM sodium butyrate.
  • (2) Cells are placed in 96-well plates at 30,000/well/100 μL in minimal essential medium (MEM) (phenol red-free)+10% foetal bovine serum (FBS), 5mM HEPES buffer (pH7.3), 1% glutamine, 0.2 mg/mL G418 antibiotic, 10 mM sodium butyrate. [0037]
  • (3) Make dilutions of the compound plate. Dilute stock solution to 5.5% DMSO/110 μM compound. Mix compounds vigorously and store at 4° C. until use. [0038]
  • (4) Add 10 μL compound/well. Mix plate briefly, and leave for 18 h in 37° C. incubator. [0039]
  • (5) Remove 90 μL of culture supernatant and dilute 1:1 with ice-cold 25mM HEPES (pH.3), 0.1% BSA, 1.0 mM EDTA (+broad spectrum protease inhibitor cocktail; pre-aliquotted into a 96-well plate). Mix and keep on ice or freeze at -80° C. [0040]
  • (6) Add back 100 μL of warm MEM +10% FBS, 50 mM HEPES (pH7.3), 1% glutamine, 0.2 mg/mL G418, 10 mM sodium butyrate to each well, and return plate to 37° C. incubator. [0041]
  • (7) Prepare reagents necessary to determine amyloid peptide levels, for example by ELISA assay [0042]
  • (8) To determine if compounds are cytotoxic cell viability following compound administration is assessed by the use of redox dye reduction. A typical example is a combination of redox dye MTS (Promega) and the electron coupling reagent PES. This mixture is made up according to the manufacturer's instructions and left at room temperature. [0043]
  • (9) Quantitate amyloid beta 40 and 42 peptides using an appropriate volume of diluted culture medium by standard ELISA techniques. [0044]
  • (10) Add 15 μL/well MTS/PES solution to the cells; mix and leave at 37° C. [0045]
  • (11) Read plate when the absorbance values are approximately 1.0 (mix briefly before reading to disperse the reduced formazan product). [0046]
  • The Example of the present invention had an ED[0047] 50 of less than 500 nM in the above assay.
  • The following assay was used to determine the effect of compounds on Notch signalling. [0048]
  • Xenopus embryos were cultured in {fraction (1/10)} NAM [Beck & Slack, Development, 126, 1611-1620, 1999] supplemented with protease inhibitors (200× stock in DMSO) from stage 15 [P. D. Nieuwkoop and J. Faber, Normal table of Xenopus laevis, Daudin 1967, Reprinted Garland 1994] (24 hours), to stage 40 (3 days). Control siblings were incubated in {fraction (1/10)} NAM containing 0.05% DMSO. At 3 days they were fixed in batches of 20 and measured using a graduated eyepiece on a Wild dissecting microscope set to 25×. Each tadpole was measured from head to proctodaeum (head and trunk) and from proctodaeum to tail tip (tail). Tail buds were extirpated at stage 30 and cultured for 24 hours as described previously [Tucker & Slack, Development, 121, 249-262, 1995]. Somites were detected by staining with monoclonal antibody 12/101 [Kintner & Brockes, Nature, 308, 67-69, 1984], as described elsewhere [Tucker & Slack, 1995]. In situ hybridisation for Xhox3 has been previously described [Beck & Slack, Mech. Dev., 72, 41-52, 1998]. [0049]
  • The Example of the present invention did not significantly inhibit tail bud formation. [0050]
  • The following Examples illustrate the present invention. [0051]
  • EXAMPLE 1
  • 2-[2-(3,5-Difluorophenyl)acetylamino]-N-{phenyl[(4-phenylmorpholin-2-ylmethyl)carbamoyl]methyl}propionamide [0052]
  • A mixture of [(3,5-Difluorophenyl)acetylamino]-1-ethylcarbamoyl-1-phenyl ethanoic acid (20 mg, 0.053 mmol), 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide (11.2 mg, 0.058 mmol) and hydroxybenzotriazole (7.8 mg, 0.058 mmol) in tetrahydrofuran was treated with N-phenyl 2-aminomethyl morpholine (15.2 mg, 0.079 mmol) and triethylamine (16 mg, 0.159 mmol) and the reaction stirred for 18 hours at room temperature. Evaporation of the solvent and purification on a bond elute cartridge gave the product (6 mg). Mixture of diastereoisomers: ([0053] 1H, DMSO) 8.45-8.34 (3 H, m), 7.43-6.76 (10 H, m), 5.44 (1 H, t), 4.44 (1 H, t), 3.91 (1 H, br), 3.59-3.16 (11 H, m), 2.61 (1 H, dd), 2.26 (1 H,m), 1.22 (3 H, m) MS (CI+) MH+551.

Claims (9)

1. A compound of formula I or a pharmaceutically acceptable salt thereof:
Figure US20020013315A1-20020131-C00004
wherein: X is CH2, oxygen or sulphur; and
Ar is phenyl optionally substituted by one, two or three substituents chosen from halogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, hydroxy, cyano, nitro, NR1R2 where R1 and R2 are independently hydrogen or C1-6alkyl, C1-6alkoxy, C2-6alkenyloxy, C2-6alkynyloxy, thiol, C1-6alkylthio, C2-6alkenylthio, C2-6alkynylthio, C1-6alkylcarbonyl, C1-6alkoxycarbonyl, C1-6haloalkyl, C2-6haloalkenyl and C2-6haloalkynyl:
2. A compound according to claim 1 wherein X is oxygen.
3. A compound according to claim 1 wherein Ar is optionally substituted by one, two or three substituents chosen from halogen, C1-6alkyl, hydroxy, amino, C1-6alkoxy, thiol, C1-6alkoxycarbonyl and C1-6haloalkyl.
4. A compound according to claim 3 wherein Ar is optionally substituted by one substituent chosen from halogen, C1-4alkyl, hydroxy, amino, C1-4alkoxy, thiol, C1-4alkoxycarbonyl and C1-4haloalkyl.
5. A compound according to claim 1 wherein Ar is unsubstituted.
6. A compound according to claim 1 selected from 2-[2-(3,5 -difluorophenyl)acetylamino]-N-{phenyl[(4-phenylmorpholin-2-ylmethyl)carbamoyl]methyl}propionamide and the pharmaceutically acceptable salts thereof.
7. A pharmaceutical composition comprising one or more compounds according to claim 1 and a pharmaceutically acceptable carrier.
8. A method of treatment of a subject suffering from or prone to Alzheimer's disease which comprises administering to that subject an effective amount of a compound according to claim 1.
9. A method of treatment according to claim 8 which does not inhibit Notch signalling.
US09/896,296 2000-07-06 2001-06-29 Gamma secretase inhibitors Expired - Fee Related US6448229B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB0016681.9A GB0016681D0 (en) 2000-07-06 2000-07-06 Therapeutic compounds
GB0016681 2000-07-06
GB0016681.9 2000-07-06

Publications (2)

Publication Number Publication Date
US20020013315A1 true US20020013315A1 (en) 2002-01-31
US6448229B2 US6448229B2 (en) 2002-09-10

Family

ID=9895191

Family Applications (1)

Application Number Title Priority Date Filing Date
US09/896,296 Expired - Fee Related US6448229B2 (en) 2000-07-06 2001-06-29 Gamma secretase inhibitors

Country Status (2)

Country Link
US (1) US6448229B2 (en)
GB (1) GB0016681D0 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020133159A1 (en) * 2000-12-08 2002-09-19 Jackson Roger P. Closure for open-headed medical implant
US20040167526A1 (en) * 2002-09-06 2004-08-26 Roger P. Jackson Closure for rod receiving orthopedic implant having left handed thread removal
US20040172032A1 (en) * 2002-09-06 2004-09-02 Jackson Roger P. Anti-splay medical implant closure with multi-surface removal aperture
US20040199164A1 (en) * 2002-09-06 2004-10-07 Jackson Roger P. Helical wound mechanically interlocking mating guide and advancement structure
US20050182410A1 (en) * 2002-09-06 2005-08-18 Jackson Roger P. Helical guide and advancement flange with radially loaded lip
US20050232927A1 (en) * 2004-02-03 2005-10-20 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
US20060009773A1 (en) * 2002-09-06 2006-01-12 Jackson Roger P Helical interlocking mating guide and advancement structure
US20060241603A1 (en) * 2003-06-18 2006-10-26 Jackson Roger P Polyaxial bone screw assembly with fixed retaining structure
US20060252073A1 (en) * 2005-04-18 2006-11-09 Regents Of The University Of Michigan Compositions and methods for the treatment of cancer
US20070078099A1 (en) * 2003-02-27 2007-04-05 Mclaurin Joanne Method of preventing, treating and diagnosing disorders of protein aggregation
US20080019961A1 (en) * 2006-02-21 2008-01-24 Regents Of The University Of Michigan Hedgehog signaling pathway antagonist cancer treatment
US20080178305A1 (en) * 2000-08-03 2008-07-24 The Regents Of The University Of Michigan Isolation And Use Of Solid Tumor Stem Cells
US20090163498A1 (en) * 2002-11-18 2009-06-25 Chemocentryx, Inc. Aryl Sulfonamides
US20100016904A1 (en) * 2003-06-18 2010-01-21 Jackson Roger P Upload shank swivel head bone screw spinal implant
US7723112B2 (en) 2005-10-31 2010-05-25 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20100211114A1 (en) * 2003-06-18 2010-08-19 Jackson Roger P Polyaxial bone anchor with shelf capture connection
US20100318136A1 (en) * 2003-06-18 2010-12-16 Jackson Roger P Polyaxial bone screw assembly
EP2369014A1 (en) 2004-02-03 2011-09-28 The Regents Of The University Of Michigan Office Of Technology Transfer Compositions and methods for characterizing, regulating, diagnosing and treating cancer

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6191166B1 (en) * 1997-11-21 2001-02-20 Elan Pharmaceuticals, Inc. Methods and compounds for inhibiting β-amyloid peptide release and/or its synthesis
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
WO2003032994A2 (en) * 2001-10-17 2003-04-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel tri-substituted pyrimidines, method for production and use thereof as medicament
PT2500360E (en) 2005-10-31 2015-10-15 Oncomed Pharm Inc Compositions and methods for diagnosing and treating cancer
US7723477B2 (en) 2005-10-31 2010-05-25 Oncomed Pharmaceuticals, Inc. Compositions and methods for inhibiting Wnt-dependent solid tumor cell growth
US9567396B2 (en) 2006-03-07 2017-02-14 Evonik Degussa Gmbh Notch inhibition in the prevention of vein graft failure
WO2011053822A2 (en) 2009-11-01 2011-05-05 The Brigham And Women's Hospital, Inc. Notch inhibition in the treatment and prevention of obesity and metabolic syndrome
US8133857B2 (en) * 2006-03-07 2012-03-13 The Brigham and Women's FHospital, Inc. NOTCH inhibition in the treatment of atherosclerosis
JP5379692B2 (en) 2006-11-09 2013-12-25 プロビオドルグ エージー 3-Hydroxy-1,5-dihydro-pyrrol-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcers, cancer and other diseases
JP5523107B2 (en) 2006-11-30 2014-06-18 プロビオドルグ エージー Novel inhibitors of glutaminyl cyclase
US8148147B2 (en) 2007-01-24 2012-04-03 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing pancreatic cancer
ZA200905537B (en) 2007-03-01 2010-10-27 Probiodrug Ag New use of glutaminyl cyclase inhibitors
JP5667440B2 (en) 2007-04-18 2015-02-12 プロビオドルグ エージー Thiourea derivatives as glutaminyl cyclase inhibitors
SG190568A1 (en) 2008-09-26 2013-06-28 Oncomed Pharm Inc Frizzled-binding agents and uses thereof
US20120129189A1 (en) * 2009-07-17 2012-05-24 Chulan Kwon Methods of Controlling Cell Proliferation
US8486940B2 (en) 2009-09-11 2013-07-16 Probiodrug Ag Inhibitors
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
KR101790806B1 (en) 2010-03-10 2017-11-20 프로비오드룩 아게 Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
JP2013530929A (en) 2010-04-01 2013-08-01 オンコメッド ファーマシューティカルズ インコーポレイテッド Frizzled binders and uses thereof
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
ES2570167T3 (en) 2011-03-16 2016-05-17 Probiodrug Ag Benzimidazole derivatives as glutaminyl cyclase inhibitors
US9629891B2 (en) 2011-10-17 2017-04-25 Nationwide Children's Hospital, Inc. Products and methods for aortic abdominal aneurysm
CA2887711A1 (en) 2012-10-23 2014-05-01 Oncomed Pharmaceuticals, Inc. Methods of treating neuroendocrine tumors using wnt pathway-binding agents
US9359444B2 (en) 2013-02-04 2016-06-07 Oncomed Pharmaceuticals Inc. Methods and monitoring of treatment with a Wnt pathway inhibitor
US9168300B2 (en) 2013-03-14 2015-10-27 Oncomed Pharmaceuticals, Inc. MET-binding agents and uses thereof
DK3461819T3 (en) 2017-09-29 2020-08-10 Probiodrug Ag GLUTAMINYL CYCLASE INHIBITORS
US20210290633A1 (en) 2018-07-19 2021-09-23 INSERM (Insstitut National de la Santé et de la Recherche Médicale) Combination for treating cancer
EP4244391A1 (en) 2020-11-16 2023-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR016751A1 (en) 1996-11-22 2001-08-01 Athena Neurosciences Inc METHOD FOR INHIBITING THE RELEASE OF THE BETA-AMYLOID PEPTIDE IN A CELL, PHARMACEUTICAL COMPOSITION AND USEFUL COMPOUNDS IN THIS METHOD

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8044259B2 (en) 2000-08-03 2011-10-25 The Regents Of The University Of Michigan Determining the capability of a test compound to affect solid tumor stem cells
US8420885B2 (en) 2000-08-03 2013-04-16 The Regents Of The University Of Michigan Determining the capability of a test compound to affect solid tumor stem cells
US20080178305A1 (en) * 2000-08-03 2008-07-24 The Regents Of The University Of Michigan Isolation And Use Of Solid Tumor Stem Cells
US20020133159A1 (en) * 2000-12-08 2002-09-19 Jackson Roger P. Closure for open-headed medical implant
US20050182410A1 (en) * 2002-09-06 2005-08-18 Jackson Roger P. Helical guide and advancement flange with radially loaded lip
US20080039848A1 (en) * 2002-09-06 2008-02-14 Jackson Roger P Anti-splay medical implant closure with multi-surface removal aperture
US20060009773A1 (en) * 2002-09-06 2006-01-12 Jackson Roger P Helical interlocking mating guide and advancement structure
US20040199164A1 (en) * 2002-09-06 2004-10-07 Jackson Roger P. Helical wound mechanically interlocking mating guide and advancement structure
US20040167526A1 (en) * 2002-09-06 2004-08-26 Roger P. Jackson Closure for rod receiving orthopedic implant having left handed thread removal
US20090259259A1 (en) * 2002-09-06 2009-10-15 Jackson Roger P Helical wound mechanically interlocking mating guide and advancement structure
US20040172032A1 (en) * 2002-09-06 2004-09-02 Jackson Roger P. Anti-splay medical implant closure with multi-surface removal aperture
US9890148B2 (en) 2002-11-18 2018-02-13 Chemocentryx, Inc. Aryl sulfonamides
US20090163498A1 (en) * 2002-11-18 2009-06-25 Chemocentryx, Inc. Aryl Sulfonamides
US10364240B2 (en) 2002-11-18 2019-07-30 ChemoCentryx. Inc. Aryl sulfonamides
US9833420B2 (en) 2003-02-27 2017-12-05 JoAnne McLaurin Methods of preventing, treating, and diagnosing disorders of protein aggregation
US8859628B2 (en) 2003-02-27 2014-10-14 JoAnne McLaurin Method for preventing, treating and diagnosing disorders of protein aggregation
US20090227686A1 (en) * 2003-02-27 2009-09-10 Mclaurin Joanne Methods of preventing, treating and diagnosing disorders of protein aggregation
US20070078099A1 (en) * 2003-02-27 2007-04-05 Mclaurin Joanne Method of preventing, treating and diagnosing disorders of protein aggregation
US20060241603A1 (en) * 2003-06-18 2006-10-26 Jackson Roger P Polyaxial bone screw assembly with fixed retaining structure
US20100211114A1 (en) * 2003-06-18 2010-08-19 Jackson Roger P Polyaxial bone anchor with shelf capture connection
US20100318136A1 (en) * 2003-06-18 2010-12-16 Jackson Roger P Polyaxial bone screw assembly
US20100030280A1 (en) * 2003-06-18 2010-02-04 Jackson Roger P Upload shank swivel head bone screw spinal implant
US20100016904A1 (en) * 2003-06-18 2010-01-21 Jackson Roger P Upload shank swivel head bone screw spinal implant
EP2369014A1 (en) 2004-02-03 2011-09-28 The Regents Of The University Of Michigan Office Of Technology Transfer Compositions and methods for characterizing, regulating, diagnosing and treating cancer
US20080260734A1 (en) * 2004-02-03 2008-10-23 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
US20050232927A1 (en) * 2004-02-03 2005-10-20 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
US20060252073A1 (en) * 2005-04-18 2006-11-09 Regents Of The University Of Michigan Compositions and methods for the treatment of cancer
US7723112B2 (en) 2005-10-31 2010-05-25 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20080019961A1 (en) * 2006-02-21 2008-01-24 Regents Of The University Of Michigan Hedgehog signaling pathway antagonist cancer treatment

Also Published As

Publication number Publication date
US6448229B2 (en) 2002-09-10
GB0016681D0 (en) 2000-08-23

Similar Documents

Publication Publication Date Title
US6448229B2 (en) Gamma secretase inhibitors
US6984626B2 (en) Gamma-secretase inhibitors
US6756511B2 (en) Gamma-secretase inhibitors
US7157478B2 (en) Oxadiazole derivatives for inhibition of gamma secretase
US7049296B2 (en) Gamma-secretase inhibitors
TOMIYAMA et al. Rifampicin inhibits the toxicity of pre-aggregated amyloid peptides by binding to peptide fibrils and preventing amyloid-cell interaction
JP2004531517A (en) Sulfone regulating the action of γ-secretase
AU2017272151B2 (en) Benzylideneguanidine derivatives and therapeutic use for the treatment of protein misfolding diseases
WO2003093253A1 (en) Alkynyl-substituted spirocyclic sulfamides for the treatment of alzheimer's disease
US20030078240A1 (en) Phosphinylmethyl and phosphorylmethyl succinic and glutaric acid analogs as beta-secretase inhibitors
HUT70408A (en) 1-benzene-sulfonyl-1,3-dihydro-indol-2-one derivatives, process for producing them and pharmaceutical compositions containing them
KR20120112351A (en) Opsin-binding ligands, compositions and methods of use
KR20110028554A (en) O-substituted hydroxyaryl derivatives
EP1309585B1 (en) Novel indole derivatives and their use as medicaments
DE60318317T2 (en) ACYLAMINOTHIAZOL DERIVATIVES AND THEIR UTILIZATION AS BETA-AMYLOID INHIBITORS
IL92870A (en) Substituted semicarbazones and their use as arthropodicides
US20110021471A1 (en) REDUCING Abeta42 LEVELS AND Abeta AGGREGATION
EP0595183B1 (en) Hydantoin derivatives, salts thereof and maillard reaction inhibitors comprising the same
EP3535239A1 (en) Inhibitors of mtor-deptor interactions and methods of use thereof
PT809635E (en) ALPHA-SUBSTITUTED HYDRAZINES WITH CALPAIN INHIBITING ACTIVITY
US5252592A (en) Tetrazoleacetic acid derivatives having aldose reductase inhibitory activity
KR20010092424A (en) Novel type condensed pyridazinone compounds
US20040248877A1 (en) Polycyclic diazodioxide-based Bcl-2 protein antagonists and use thereof
Attia et al. Research Article Anticonvulsant Potential of Certain New (2E)-2-[1-Aryl-3-(1H-imidazol-1-yl) propylidene]-N-(aryl/H) hydrazinecarboxamides
KR20080006384A (en) Compounds for inhibiting p25 and process for preparing same

Legal Events

Date Code Title Description
AS Assignment

Owner name: SULZER SPINE-TECH INC., MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HANSON, DAVID A.;LONGHINI, ROSS A.;SEME, STEVEN J.;REEL/FRAME:012449/0688;SIGNING DATES FROM 20011016 TO 20011022

AS Assignment

Owner name: MERCK SHARP & DOHME LTD., ENGLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TEALL, MARTIN RICHARD;REEL/FRAME:013116/0080

Effective date: 20010621

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Expired due to failure to pay maintenance fee

Effective date: 20140910