EP2054414A2 - Imidazole amines as inhibitors of beta-secretase - Google Patents

Imidazole amines as inhibitors of beta-secretase

Info

Publication number
EP2054414A2
EP2054414A2 EP07840855A EP07840855A EP2054414A2 EP 2054414 A2 EP2054414 A2 EP 2054414A2 EP 07840855 A EP07840855 A EP 07840855A EP 07840855 A EP07840855 A EP 07840855A EP 2054414 A2 EP2054414 A2 EP 2054414A2
Authority
EP
European Patent Office
Prior art keywords
compound
formula
optionally substituted
phenyl
group
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.)
Withdrawn
Application number
EP07840855A
Other languages
German (de)
French (fr)
Inventor
Michael Sotirios Malamas
Keith Douglas Barnes
Matthew Robert Johnson
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.)
Wyeth LLC
Original Assignee
Wyeth LLC
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 Wyeth LLC filed Critical Wyeth LLC
Publication of EP2054414A2 publication Critical patent/EP2054414A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61P25/28Drugs 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • AD Alzheimer's disease
  • ⁇ -amyloid angiopathy cerebral blood vessels
  • Amyloidogen ⁇ c plaques and vascular amyloid angiopathy also characterize the brains of patients with Trisomy 21 (Down's Syndrome), Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), and other neurodegenerative disorders. Neurofibrillary tangles also occur in other dementia- inducing disorders.
  • A-amyloid The family of proteins known as ⁇ -amyloid are thought to be causal for the pathology and subsequent cognitive decline in Alzheimer's disease.
  • Proteolytic processing of the amyloid precursor protein (APP) generates amyloid ⁇ (A-beta) peptide; specifically, A-beta is produced by the cleavage of APP at the N-terminus by ⁇ -secretase and at the C-terminus by one or more y-secretases.
  • Aspartyl protease enzyme, or ⁇ -secretase enzyme (BACE), activity is correlated directly to the generation of A-beta peptide from APP (Sinha, et al, Nature, 1999, 402, 537-540), Increasingly, studies indicate that the inhibition of the ⁇ -secretase enzyme, inhibits the production of A-beta peptide.
  • the inhibition of ⁇ -secretase and consequent lowering of A-beta peptide may lead to the reduction of ⁇ -amyloid deposits in the brain and ⁇ -amylo ⁇ d levels in the cerebral blood vessels and to an effective treatment of a disease or disorder caused thereby.
  • the compounds provided may also be useful to further study and elucidate the ⁇ -secretase enzyme.
  • the present invention provides an imidazole amine of formula I
  • Q is O, S or CH 2 ;
  • VV is O 1 S or CH 2 ;
  • X is N, NO, SO m , O or CH;
  • Y is N, NO, SO 01 , O or CR 10 ;
  • Z is N, NO 1 SO m , O or CRn with the proviso that when X is CH, Y is CR 10 and Z is CR 11 then one of Q or W must be O or S; m is 0, 1 or 2; n is 0 or 1; R 1 and R 2 are each independently H or an optionally substituted Ci-
  • R 3 and R 4 are each independently H, or an optionafly substituted C 1 -C 4 alky j group or R 3 and R 4 may be taken together to form a 4- to 7- membered ring optionally containing one or two heteroatoms selected from O, N or S;
  • R 5 and R 6 are each independently H, halogen, NO 2 , CN, OR 12 , CO 2 R 13 ,
  • R 7 and R 8 are each independently H, halogen, NO 2 , CN, OR 15 , NR 17 R 18 or a CrC 6 alky!, C r C 6 haloaikyl, C 2 -C 6 alkenyl, C 2 -C 6 aikyny(,
  • R 7 and R 8 may be taken together with the atoms to which they are attached to form an optionally substituted 5- to 7-membered ring optionally containing one or two heteroatoms selected from O, N or S;
  • R 9 is H, halogen, NO 2 , CN, OR 15 , NR 17 R 18 or a Ci-C 6 alkyl, d-Cghaloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted;
  • R 10 and Rn are each independently H or a C r C 6 alkyl, C r C 6 haloalkyl,
  • R 12 , Ri3, Ru and R 15 are each independently H or a C r C 6 alkyl, C 1 -
  • Ri7, Ri8, Rig and R 20 are each independently H, CrC 4 alkyl, C 3 -
  • Cscycloatkyl or R 17 and Ri S or R 19 and R 20 may be taken together with the atom to which they are attached to form a 5- to 7- membered ring optionally containing an additional heteroatom selected from O, N or S; and p is 0, 1 or 2; or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
  • the present invention also provides therapeutic methods and pharmaceutical compositions useful for the treatment, prevention or amelioration of a disease or disorder characterized by increased ⁇ -amyloid deposits or increased ⁇ -amyloid levels in a patient.
  • AD Alzheimer's disease
  • A-beta amyloid beta peptide piays a central role in the pathogenesis of the disease.
  • AD Alzheimer's disease
  • ⁇ -amyloid deposits and vascular ⁇ -amyloid angiopathy also characterize individuals with Downs Syndrome, Hereditary Cerebral Hemmorhage with Amyloidosis of the Dutch type and other neurodegenreative and dementia- inducing disorders.
  • BACE1 amyloid precursor protein
  • BACE2 a second homologous aspartyl protease named BACE2 was found to have ⁇ -secretase activity in vitro.
  • Low molecular weight, non-peptide, non-substrate-related inhibitors of BACE1 or ⁇ - secretase are earnestly sought both as an aid in the study of the ⁇ -secretase enzyme and as potential therapeutic agents.
  • imidazole amine compounds of formula I demonstrate inhibition of ⁇ -secretase and the selective inhibition of BACE1.
  • sard imidazole amine compounds may be used as effective therapeutic agents for the treatment, prevention or amelioration of a disease or disorder characterized by elevated ⁇ -amyloid deposits or ⁇ -amyloid levels in a patient.
  • the present invention provides an imidazole amine compound of formula I
  • Q is O, S or CH 2 ;
  • W is O, S or CH 2 ;
  • X is N 1 NO, SO m , O or CH;
  • Y is N, NO, SO m , O or CR 10 ;
  • Z is N, NO, SO m , O or CR 11 with the proviso that when X is CH, Y is CR 10 and Z is CRn then one of Q or W must be O or S; m is 0, 1 or 2; n is O or 1; R 1 and R 2 are each independently H or an optionally substituted C 1 -
  • R 3 and R 4 are each independently H, or an optionally substituted C 1 -C 4 alkyi group or R 3 and R 4 may be taken together to form a 4- to 7- membered ring optionally containing one or two heteroatoms selected from O, N or S;
  • R 6 and R 6 are each independently H, halogen, NO 2 , CN, OR 12 , CO 2 R 13 , COR 14 , NR 17 Ri 8 , SOpNR 19 R 20 or a d-Cealkyl, C r C 6 haioaikyl r C 2 - C 6 alkenyL C 2 -C 6 alkynyl or C 3 -C 8 cycloalkyl group each optionally substituted;
  • R 7 and R 8 are each independently H, halogen, NO 2 , CN, OR 15 , NR 17 R 18 or a C ⁇ Cealkyl, C r C 6 haloalkyl ( C 2 -C 6 alkenyl, C 2 -C 6 alkyny!, C 3 ⁇ C 8 cycloalkyl or cycloheteroalkyl group each optionally substituted or when attached to adjacent carbon atoms R 7 and R 8 may be taken together with the atoms to which they are attached to form an optionally substituted 5- to 7-membered ring optionally containing one or two heteroatoms selected from O, N or S;
  • R 9 is H, halogen, NO 2 , CN, OR 15 , NR 17 R 18 or a C r C 6 alkyl, C r C 6 haloalky ⁇ , C 2 -C 6 alkenyl, Ca-Cealkynyl, C 3 -C 8 cycloalkyl, cyclo
  • R 10 and R 11 are each independently H or a C ⁇ Cealkyl, CrCehaloalkyl, C z -C 6 alkeny!, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, cycloheteroalkyl or aryl group each optionally substituted;
  • R 12 , Ri3, R- I4 and Ri 5 are each independently H or a CrC ⁇ alkyl, C 1 - C ⁇ haloalkyi, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyi, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted;
  • R 17 , R 18 , R 19 and R 20 are each independently H, C r C 4 alkyl, C 3 -
  • Cgcycloalkyf or R 17 and R 18 or R 19 and R 20 may be taken together with the atom to which they are attached to form a 5- to 7- membered ring optionally containing an additional heteroatom selected from O 5 N or S; and p is O, 1 or 2; or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
  • halogen designates F, Cl, Br or 1
  • cycloheteroalkyi designates a five- to seven-membered cycloalkyl ring system containing 1 or 2 heteroatoms, which may be the same or different, selected from N, O or S and optionally containing one double bond.
  • exemplary of the cycloheteroalkyi ring systems included in the term as designated herein are the following rings wherein X 1 is NR, O or S; and R is H or an optional substituent as described hereinbeiow:
  • heteroaryl designates a five- to ten-membered aromatic ring system containing 1 , 2 or 3 heteroatoms, which may be the same or different, selected from N, O or S.
  • heteroaryl ring systems include pyrrolyl, azolyl, oxazolyl, thiazolyl, imidazolyl, furyl, thienyl, qu ⁇ nolinyi, isoquinoiinyl, indolyl, benzothienyl, benzofuranyl, benzisoxazolyl or the like.
  • aryl designates a carbocyclic aromatic ring system, e.g., of 6-14 carbon atoms, such as phenyl, naphthy!, anthracenyl or the like.
  • aryl ⁇ Ci- C 4 )alkyl designates an aryl group as defined hereinabove attached to a Ci-C 4 alkyl group which may be straight or branched.
  • Said aryl(C r C 4 )alkyl groups include benzyl, phenethyl, naphthylmethyl, or the like.
  • haloalkyl as used herein designates a C n H 2n+ !
  • haloalkyl designates CF 3 and the term haloalkoxy designates OCF 3 .
  • substituent groups which are optionally present may be one or more of those customarily employed tn the development of pharmaceutical compounds or the modification of such compounds to influence their structure/activity, persistence, absorption, stability or other beneficial property
  • substituents include halogen atoms, ⁇ itro, cyano, thiocyanato, cyanato, hydroxyl, alky!, haloaikyl, alkoxy, haloalkoxy, amino, alkylamino, dialkylamino, formyl, alkoxycarbonyl, carboxyl, alkanoyl, alkylthio, al
  • Pharmaceutically acceptable salts may be any acid addition salt formed by a compound of formula I and a pharmaceutically acceptable acid such as phosphoric, sulfuric, hydrochloric, hydrobromic, citric, maleic, malonic, mandehc, succinic, fumaric, acetic, lactic, nitric, sulfonic, p-toluene sulfonic, methane sulfonic acid or the like
  • a pharmaceutically acceptable acid such as phosphoric, sulfuric, hydrochloric, hydrobromic, citric, maleic, malonic, mandehc, succinic, fumaric, acetic, lactic, nitric, sulfonic, p-toluene sulfonic, methane sulfonic acid or the like
  • compounds of the invention include esters, carbamates or other conventional prodrug forms, which in general, are functional derivatives of the compounds of the invention and which are readily converted to the inventive active moiety in vivo
  • the method of the invention embraces the treatment of the various conditions described heremabove with a compound of formula I or with a compound which is not specifically disclosed but which, upon administration, converts to a compound of formula I in vivo
  • metabolites of the compounds of the present invention defined as active species produced upon introduction of these compounds into a biological system
  • Tautomers often exist in equilibrium with each other. As these tautomers interconvert under environmental and physiological conditions, they provide the same useful biological effects.
  • the present invention includes mixtures of such tautomers as well as the individual tautomers of Formula I and Formula It.
  • the compounds of the invention may contain one or more asymmetric carbon atoms or one or more asymmetric (chiral) centers and may thus give rise to optica! isomers and diastereomers.
  • the invention includes such optical isomers and disastereomers; as well as the racemic and resolved, enantiomerically pure stereoisomers; as well as other mixtures of the R and S stereoisomers.
  • one stereoisomer may be more active or may exhibit beneficial effects when enriched relative to the other stereoisomers) or when separated from the other stereoisomerfs). Additionally, the skilled artisan knows how to separate, enrich or selectively prepare said stereoisomers.
  • the present invention comprises compounds of Formula 1, the stereoisomers thereof, the tautomers thereof and the pharmaceutically acceptable salts thereof.
  • the compounds of the invention may be present as a mixture of stereoisomers, individual stereoisomers, or as an optically active or enantiomerically pure form.
  • the point of attachment may be via Z in which case to fulfil valency requirements R 11 is absent.
  • an example of the five membered ring is pyrazoly! such as pyrazol-4-yJ (i.e., X and Y are N), which ring may be optionally substituted, e.g., 1- ethylpyrazol-4-yl or 1 -(2,2,2 ⁇ trifluoroethy)pyrazol-4-yl.
  • n 1
  • an example of the six membered ring is pyridyl such as pyrid-4- yl, or phenyl, which rings may be optionally substituted, e.g., 2,6-diethylpyrid-4-yl or 4-trifluromethoxyphenyl.
  • Preferred compounds of the invention are those compounds of formula ! wherein R 1 and R 2 are H. Another group of preferred compounds of the invention are those compounds of formula I wherein R 9 is an optionally substituted heteroaryl group. Also preferred are those formula I compounds wherein X is N. A further group of preferred compounds of the invention are those compounds of formula I wherein R 9 is an optionally substituted heteroaryl group and is attached to the phenyl ring in the 3-position of the phenyl ring.
  • More preferred compounds of the invention are those compounds of formula 1 wherein R 1 and R 2 are H and Rg is an optionally substituted heteroaryl group.
  • Another group of more preferred compounds of the invention are those compounds of formula I wherein R 1 and R 2 are H; R 9 is an optionally substituted heteroaryl group; and X is N.
  • a further group of more preferred compounds of the invention are those compounds of formula i wherein R 1 and R 2 are H and R 9 is an optionally substituted heteroaryl group and is attached to the phenyl ring in the 3-position of the phenyl ring.
  • Preferred compounds of formula I include:
  • the present invention provides a process for the preparation of a compound of formula i wherein R 9 is an optionally substituted aryl or heteroaryf group (Ia) which comprises reacting a compound of formula Il wherein Hal is Cl or Br with an optionally substituted aryl or heteroaryl group having a leaving group selected from B(OH) 2 , Sn(Bu) 3 or Sn(CH 3 ) 3 in the presence of a palladium catalyst and an inorganic base optionally in the presence of a solvent.
  • A represents an optionally substituted aryl or heteroaryl group
  • W is B(OH) 2 , Sn(Bu) 3 or Sn(CH 3 J 3
  • Hal is Cl or Br.
  • Palladium catalysts suitable for use in the process of the invention include
  • Pd(O) or Pd(H) catalysts such as dichlorobis(tri-o-tolyfphosphine)palladium(ll), Pd(OCOCH 3 ) 2 /tri-o-tolylphosphine, tetrakis(triphenylphosphine)pa!ladium(0), tris(dibenzylideneacetone)dipallad!um(0)triphenylphosphine, or the like.
  • Inorganic bases suitable for use in the inventive process include Na or K hydroxides, carbonates or bicarbonates, preferably Na 2 CO 3 Or K 2 CO 3 .
  • Solvents suitable for use in the inventive process include polar or non-polar organic solvents such as toluene, diethoxy ethyl ether, dioxane, ethyleneglycol dimethyl ether or any non-reactive organic solvent which is capable of solubiiizing the formula Il or heteroaryl compounds.
  • polar or non-polar organic solvents such as toluene, diethoxy ethyl ether, dioxane, ethyleneglycol dimethyl ether or any non-reactive organic solvent which is capable of solubiiizing the formula Il or heteroaryl compounds.
  • Compounds of formula Il may be prepared using conventional synthetic methods and, if required, standard separation or isolation techniques.
  • compounds of formula Il wherein R t and R 2 are H and Q and W are CH 2 (Ha) may be prepared by reacting a compound of formula IM with a diamine of formula IV to give the bicyclic compound of formula V and reacting said formula V compound with t- butyl hydroperoxide and ammonium hydroxide to give the desired formula Ma compound.
  • the reaction is shown in flow diagram Il wherein Hal is Cl or Br,
  • compounds of formula Il wherein R t and R 2 are H; R 3 and R 4 are H; Q is CH 2 ; and W is O (lib) may be prepared by reacting the formula III compound with 2-(aminooxy)ethanamine dihydrochloride in the presence of a base such as triethylamine and a solvent to give the bicyclic compound of formula Vl and reacting said formula Vl compound with t-butyl hydroperoxide and ammonium hydroxide to give the desired formula Mb compound.
  • a base such as triethylamine and a solvent
  • the reaction is shown in flow diagram III wherein Hal is Cl or Br.
  • R 1 and R 2 are other than H
  • Compounds of formula 1 wherein R 1 and R 2 are other than H may be prepared using standard aikylation techniques such as reacting the compound of formula ! wherein R 1 and R 2 are H with an alkyl halide, R 1 -HaI 1 to give the compound of formula I wherein R 2 is H (Id) and optionally reacting said formula Id compound with a second aikyi halide, R 2 -HaI, to give the desired formula I compound wherein R 1 and R 2 are other than H.
  • the compounds of the invention are useful for the treatment, prevention or amelioration of a disease or disorder characterized by elevated ⁇ -amyloid deposits or ⁇ -amyloid levels in a patient, including Alzheimer's disease, Downs Syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch type or other neurodegenerative or dementia-inducing disorders.
  • the present invention provides a method for the treatment, prevention or amelioration of a disease or disorder characterized by elevated ⁇ -amyloid deposits or ⁇ -amyloid levels in a patient which comprises providing said patient with a therapeutically effective amount of a compound of formula I as described hereinabove.
  • the compound may be provided by oral or parenteral administration or in any common manner known to be an effective administration of a therapeutic agent to a patient in need thereof.
  • providing designates either directly administering such a compound or substance, or administering a prodrug, derivative or analog which forms an equivalent amount of the compound or substance within the body.
  • CNS disorder may vary according to the specific condition(s) being treated, the size, age and response pattern of the patient, the severity of the disorder, the judgment of the attending physician and the like.
  • effective amounts for daily oral administration may be about 0.01 to 1,000 mg/kg, preferably about 0.5 to 500 mg/kg and effective amounts for parenteral administration may be about 0.1 to 100 mg/kg, preferably about 0.5 to 50 mg/kg.
  • the compounds of the invention are provided by administering the compound or a precursor thereof in a solid or liquid form, either neat or in combination with one or more conventional pharmaceutical carriers or excipients. Accordingly, the present invention provides a pharmaceutical composition which comprises a pharmaceuticaliy acceptable carrier and an effective amount of a compound of formula I as described hereinabove.
  • Solid carriers suitable for use in the composition of the invention include one or more substances which may also act as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aides, binders, tablet-disintegrating agents or encapsulating materials.
  • the carrier may be a finely divided solid which is in admixture with a finely divided compound of formula I.
  • the formula I compound may be mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired. Said powders and tablets may contain up to 99% by weight of the formula I compound.
  • Solid carriers suitable for use in the composition of the invention include calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, polyvinylpyrroiidine, low melting waxes and ion exchange resins.
  • any pharmaceutically acceptable liquid carrier suitable for preparing soiutions, suspensions, emulsions, syrups and elixirs may be employed in the composition of the invention.
  • Compounds of formula f may be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, or a pharmaceutically acceptable oil or fat, or a mixture thereof.
  • Said liquid composition may contain other suitable pharmaceuticai additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, coloring agents, viscosity regulators, stabilizers, osmo-regulators, or the like.
  • liquid carriers suitable for oral and parenteral administration include water (particularly containing additives as above, e.g., cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) or their derivatives, or oils (e.g., fractionated coconut oil and arachis oil).
  • the carrier may also be an oily ester such as ethyl ofeate or isopropyl myristate.
  • compositions of the invention which are sterile solutions or suspensions are suitable for intramuscular, intraperitoneal or subcutaneous injection. Sterile solutions may also be administered intravenously.
  • Inventive compositions suitable for oral administration may be in either liquid or solid composition form.
  • sustained delivery is defined as delaying the release of an active agent, i.e., a compound of the invention, until after placement in a delivery environment, followed by a sustained release of the agent at a later time.
  • active agent i.e., a compound of the invention
  • sustained delivery devices include, e.g., hydrogels (see, e.g., US Patent Nos. 5,266,325; 4,959,217; and 5,292,515), an osmotic pump, such as described by Alza (US Patent Nos. 4,295,987 and 5,273,752) or Merck (European Patent No.
  • hydrophobic membrane materials such as ethyienemethacryiate (EMA) and ethylenevinylacetate (EVA); bioresorbable polymer systems (see, e.g., International Patent Publication No. WO 98/44964, Bioxid and Celiomeda; US Patent Nos. 5,756,127 and 5,854,388); other bioresorbable impiant devices have been described as being composed of, for example, polyesters, poiyanhydrides, or lactic acid/g!ycoiic acid copolymers (see, e.g., US Patent No. 5,817,343 (Alkermes Inc.) ⁇ .
  • the compounds of the invention may be formulated as described herein.
  • the invention provides a pharmaceutical kit for delivery of a product.
  • the kit contains packaging or a container with the compound formulated for the desired delivery route.
  • the kit may contain a suspension containing a compound of the invention formulated for aerosol or spray delivery of a predetermined dose by inhalation.
  • the kit may further contain instructions for monitoring circulating levels of product and materials for performing such assays including, e.g., reagents, well plates, containers, markers or labels, and the like.
  • Such kits are readily packaged in a manner suitable for treatment of a desired indication.
  • the kit may also contain instructions for use of the spray pump or other delivery device.
  • kits will be readily apparent to one of skill in the art, taking into consideration the desired indication and the delivery route.
  • the doses may be repeated daily, weekly, or monthly, for a predetermined length of time or as prescribed.
  • a mixture of potassium terf-butoxide (0.355 g, 3.16 mrnol) in tetrahydrofuran at -78 0 C was treated dropwise with a solution of 1 (0.665 g, 2.53 mmol) in tetrahydrofuran, stirred for 10 mtn, treated with carbon disulfide (0.635 g, 8,34 mmol), allowed to warm to room temperature slowly and stirred for 1 h at room temperature.
  • the reaction mixture was cooled to -78 0 C 1 treated with di-2-pyridyl-thiocarbonate (0.880 g, 3.79 mmol), allowed to warm to room temperature, stirred overnight at room temperature and concentrated in vacuo.
  • reaction mixture was treated with anhydrous methanol, cooled to 0 0 C, treated with sodium borohydride (0.991 g, 26.2 mmol), warmed to room temperature, stirred for 1 h at to room temperature, cooled to 0 0 C and quenched by the careful addition of saturated ammonium chloride until gas evolution had ceased and ail precipitates had dissolved.
  • the reaction mixture was diluted with methylene chloride and water. The phases were separated. The organic phase was washed with brine, dried over sodium sulfate and concentrated in vacuo.
  • a mixture of potassium f-butoxtde (0.04 g, 0.37 mmo! in tetrahydrofuran at -78 0 C was treated dropwise over a period of 2 min. with a solution of 2 (0.11 g, 0.34 mmol) and carbon disulfide (0.04 g, 0.51 mmol) in tetrahydrofuran, stirred at -78 0 C for 0.5 h, slowly warmed to room temperature, stirred at room temperature for 1 h and diluted with methylene chloride and water. The phases were separated.
  • Assay Conditions 10 nM human BACE1 (or 10 nM Murine BACE1 , 1.5 nM human BACE2) 25 ⁇ M substrate (WABC-6, MW 1549.6, from AnaSpec); final buffer conditions:50 mM Na-Acetate, pH 4.5, 0.05% CHAPS, 25% PBS; temperature: room temperature; reagent information: Na-Acetate: Aldrich, Cat. # 24,124-5 CHAPS: Research Organics, Cat.
  • Fluorescence Readings Readings at ⁇ ex 320 nm and ⁇ em 420 nm are taken every 40-sec for 30 min at room temperature to determine the linear slope for substrate cleavage rate (v,).
  • Fluorescent Kinetic Assay for human recombinant BACE 2 This assay is used to provide kinetic and selectivity parameters for the analyses of the tested compounds.
  • final assay conditions 10 nM human BACE1 (or 10 nM Murine BACE1 , 1.5 nM human BACE2) 25 ⁇ M Substrate (WABC-6, MW 1549.6, from AnaSpec).
  • Final buffer conditions 50 mM Na-Acetate, pH 4.5, 0.05% CHAPS, 25% PBS. Temperature: room temperature.
  • Reagent Information Na- Acetate: Aldrich, Cat.# 24,124-5 CHAPS: Research Organics, Cat. # 1304C 1X PBS: Mediatech (Cellgro), Cat# 21 -031 -CV Peptide Substrate AbzSEVNLDAEFRDpa: AnaSpec, Peptide Name: WABC-6
  • V 0 substrate cleavage rate in the absence of inhibitor
  • IC 5O value represents the value obtained at 100% inhibition.
  • the compounds of the invention are potent and selective inhibitors of BACE1.

Landscapes

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

Abstract

The present invention provides a compound of formula (I) and the use thereof for the therapeutic treatment, prevention or amelioration of a disease or disorder characterized by elevated ß-amyloid deposits or ß-amyloid levels in a patient (I).

Description

IMIDAZOLE AMINES AS INHIBITORS OF β-SECRETASE
BACKGROUND OF THE INVENTION
Alzheimer's disease (AD), a progressive degenerative disease of the brain primarily associated with aging, is a serious healthcare problem. Clinically, AD is characterized by the of Joss of memory, cognition, reasoning, judgment, and orientation. Also affected, as the disease progresses, are motor, sensory, and linguistic abilities until global impairment of multiple cognitive functions occurs. These cognitive losses take place gradually, but typically lead to severe impairment and eventual death in 4-12 years. Patients with AD display characteristic β-amyloid deposits in the brain and in cerebral blood vessels (β-amyloid angiopathy) as well as neurofibrillary tangles. Amyloidogenϊc plaques and vascular amyloid angiopathy also characterize the brains of patients with Trisomy 21 (Down's Syndrome), Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), and other neurodegenerative disorders. Neurofibrillary tangles also occur in other dementia- inducing disorders.
The family of proteins known as β-amyloid are thought to be causal for the pathology and subsequent cognitive decline in Alzheimer's disease. Proteolytic processing of the amyloid precursor protein (APP) generates amyloid β (A-beta) peptide; specifically, A-beta is produced by the cleavage of APP at the N-terminus by β-secretase and at the C-terminus by one or more y-secretases. Aspartyl protease enzyme, or β-secretase enzyme (BACE), activity is correlated directly to the generation of A-beta peptide from APP (Sinha, et al, Nature, 1999, 402, 537-540), Increasingly, studies indicate that the inhibition of the β-secretase enzyme, inhibits the production of A-beta peptide. The inhibition of β-secretase and consequent lowering of A-beta peptide may lead to the reduction of β-amyloid deposits in the brain and β-amyloϊd levels in the cerebral blood vessels and to an effective treatment of a disease or disorder caused thereby.
Therefore, it is an object of this invention to provide compounds which are inhibitors of β-secretase and are useful as therapeutic agents in the treatment, prevention or amelioration of a disease or disorder characterized by elevated β- amyjoid deposits or β-amyloid levels in a patient.
It is another object of this invention to provide therapeutic methods and pharmaceutical compositions useful for the treatment, prevention or amefioration of a disease or disorder characterized by elevated β-amyfoid deposits or β-amyloid levels in a patient.
It is a feature of this invention that the compounds provided may also be useful to further study and elucidate the β-secretase enzyme.
These and other objects and features of the invention will become more apparent by the detailed description set forth hereinbelow.
SUMMARY OF THE INVENTION
The present invention provides an imidazole amine of formula I
(I)
wherein
Q is O, S or CH2;
VV is O1 S or CH2; X is N, NO, SOm, O or CH;
Y is N, NO, SO01, O or CR10;
Z is N, NO1 SOm, O or CRn with the proviso that when X is CH, Y is CR10 and Z is CR11 then one of Q or W must be O or S; m is 0, 1 or 2; n is 0 or 1; R1 and R2 are each independently H or an optionally substituted Ci-
C4alkyi group;
R3 and R4 are each independently H, or an optionafly substituted C1-C4 alkyj group or R3 and R4 may be taken together to form a 4- to 7- membered ring optionally containing one or two heteroatoms selected from O, N or S; R5 and R6 are each independently H, halogen, NO2, CN, OR12, CO2R13,
COR14, NR17Ri8, SOpNR19R20 or a CrC6alkyl, CrC6haloalkyl, C2- Cealkenyl, C2-C6alkynyl or C3-C8cycloalkyl group each optionally substituted; R7 and R8 are each independently H, halogen, NO2, CN, OR15, NR17R18 or a CrC6alky!, CrC6haloaikyl, C2-C6alkenyl, C2-C6aikyny(,
C3-Cecycloalkyl or cycloheteroalkyl group each optionafly substituted or when attached to adjacent carbon atoms R7 and R8 may be taken together with the atoms to which they are attached to form an optionally substituted 5- to 7-membered ring optionally containing one or two heteroatoms selected from O, N or S; R9 is H, halogen, NO2, CN, OR15, NR17R18 or a Ci-C6alkyl, d-Cghaloalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted; R10 and Rn are each independently H or a CrC6alkyl, CrC6haloalkyl,
C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, cycloheteroalkyl or aryl group each optionally substituted; R12, Ri3, Ru and R15 are each independently H or a CrC6alkyl, C1-
C6haloalkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyl, cycioheteroalkyl, aryl or heteroaryl group each optionally substituted; Ri7, Ri8, Rig and R20 are each independently H, CrC4alkyl, C3-
Cscycloatkyl or R17 and RiSor R19 and R20 may be taken together with the atom to which they are attached to form a 5- to 7- membered ring optionally containing an additional heteroatom selected from O, N or S; and p is 0, 1 or 2; or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
The present invention also provides therapeutic methods and pharmaceutical compositions useful for the treatment, prevention or amelioration of a disease or disorder characterized by increased β-amyloid deposits or increased β-amyloid levels in a patient.
DETAILED DESCRIPTION OF THE INVENTION
Alzheimer's disease (AD) is a major degenerative disease of the brain which presents clinically by progressive loss of memory, cognition, reasoning, judgement and emotional stability and gradually leads to profound mental deteoriation and death. The exact cause of AD is unknown, but increasing evidence indicates that amyloid beta peptide (A-beta) piays a central role in the pathogenesis of the disease. (D. B. Schenk; R. E. Rydel ef a/, Journal of Medicinal Chemistry, 1995. 21 ,4141 and D. J. Seikoe, Physiology Review, 2001. 81 , 741). Patients with AD exhibit characteristic neuropathological markers such as neuritic plaques (and in β-amyioid angiopathy, deposits in cerebral blood vessels) as wefl as neurofibrillary tangles detected in the brain at autopsy, A-beta is a major component of neuritic plaques in AD brains. In addition, β-amyloid deposits and vascular β-amyloid angiopathy also characterize individuals with Downs Syndrome, Hereditary Cerebral Hemmorhage with Amyloidosis of the Dutch type and other neurodegenreative and dementia- inducing disorders. Over expression of the amyloid precursor protein (APP), altered cleavage of APP to A-beta or a decrease in the clearance of A-beta from a patient's brain may increase the levels of soluble or fibrullar forms of A-beta in the brain. The β-site APP cleaving enzyme, BACE1 , also called memapsin-2 or Asp-2, was identified in 1999 (R. Vassar, B. D. Bennett, er a/, Nature, 1999, 402, 537). BACE1 is a membrane-bound aspartic protease with all the known functional properties and characteristics of β-secretase. Parallel to BACE1 , a second homologous aspartyl protease named BACE2 was found to have β-secretase activity in vitro. Low molecular weight, non-peptide, non-substrate-related inhibitors of BACE1 or β- secretase are earnestly sought both as an aid in the study of the β-secretase enzyme and as potential therapeutic agents.
Surprisingly, it has now been found that imidazole amine compounds of formula I demonstrate inhibition of β-secretase and the selective inhibition of BACE1. Advantageously, sard imidazole amine compounds may be used as effective therapeutic agents for the treatment, prevention or amelioration of a disease or disorder characterized by elevated β-amyloid deposits or β-amyloid levels in a patient. Accordingly, the present invention provides an imidazole amine compound of formula I
(D
wherein
Q is O, S or CH2;
W is O, S or CH2;
X is N1 NO, SOm, O or CH;
Y is N, NO, SOm, O or CR10;
Z is N, NO, SOm, O or CR11 with the proviso that when X is CH, Y is CR10 and Z is CRn then one of Q or W must be O or S; m is 0, 1 or 2; n is O or 1; R1 and R2 are each independently H or an optionally substituted C1-
C4alky) group;
R3 and R4 are each independently H, or an optionally substituted C1-C4 alkyi group or R3 and R4 may be taken together to form a 4- to 7- membered ring optionally containing one or two heteroatoms selected from O, N or S;
R6 and R6 are each independently H, halogen, NO2, CN, OR12, CO2R13, COR14, NR17Ri8, SOpNR19R20 or a d-Cealkyl, CrC6haioaikylr C2- C6alkenyL C2-C6alkynyl or C3-C8cycloalkyl group each optionally substituted;
R7 and R8 are each independently H, halogen, NO2, CN, OR15, NR17R18 or a C^Cealkyl, CrC6haloalkyl( C2-C6alkenyl, C2-C6alkyny!, C3~C8cycloalkyl or cycloheteroalkyl group each optionally substituted or when attached to adjacent carbon atoms R7 and R8 may be taken together with the atoms to which they are attached to form an optionally substituted 5- to 7-membered ring optionally containing one or two heteroatoms selected from O, N or S; R9 is H, halogen, NO2, CN, OR15, NR17R18 or a CrC6alkyl, CrC6haloalkyϊ, C2-C6alkenyl, Ca-Cealkynyl, C3-C8cycloalkyl, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted;
R10 and R11 are each independently H or a C^Cealkyl, CrCehaloalkyl, Cz-C6alkeny!, C2-C6alkynyl, C3-C8cycloalkyl, cycloheteroalkyl or aryl group each optionally substituted;
R12, Ri3, R-I4 and Ri5 are each independently H or a CrCεalkyl, C1- Cβhaloalkyi, C2-C6alkenyl, C2-C6alkynyl, C3-C8cycloalkyi, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted; R17, R18, R19 and R20 are each independently H, CrC4alkyl, C3-
Cgcycloalkyf or R17 and R18 or R19 and R20 may be taken together with the atom to which they are attached to form a 5- to 7- membered ring optionally containing an additional heteroatom selected from O5 N or S; and p is O, 1 or 2; or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
As used in the specification and claims, the term halogen designates F, Cl, Br or 1 and the term cycloheteroalkyi designates a five- to seven-membered cycloalkyl ring system containing 1 or 2 heteroatoms, which may be the same or different, selected from N, O or S and optionally containing one double bond. Exemplary of the cycloheteroalkyi ring systems included in the term as designated herein are the following rings wherein X1 is NR, O or S; and R is H or an optional substituent as described hereinbeiow:
Similarly, as used in the specification and claims, the term heteroaryl designates a five- to ten-membered aromatic ring system containing 1 , 2 or 3 heteroatoms, which may be the same or different, selected from N, O or S. Such heteroaryl ring systems include pyrrolyl, azolyl, oxazolyl, thiazolyl, imidazolyl, furyl, thienyl, quϊnolinyi, isoquinoiinyl, indolyl, benzothienyl, benzofuranyl, benzisoxazolyl or the like. The term aryl designates a carbocyclic aromatic ring system, e.g., of 6-14 carbon atoms, such as phenyl, naphthy!, anthracenyl or the like. The term aryl{Ci- C4)alkyl designates an aryl group as defined hereinabove attached to a Ci-C4alkyl group which may be straight or branched. Said aryl(CrC4)alkyl groups include benzyl, phenethyl, naphthylmethyl, or the like. The term haloalkyl as used herein designates a CnH2n+! group having from one to 2n+1 halogen atoms which may be the same or different and the term haloalkoxy as used herein designates an OCnH2n+I group having from one to 2n+1 halogen atoms which may be the same or different. Preferably the term haloalkyl designates CF3 and the term haloalkoxy designates OCF3.
In the specification and claims, when terms such as d-Cβalkyl, C2-C6alkenyl, C2-C6aikynyi, C3-C8cycloalkyl, cycloheteroalkyi, aryl, aryl(Ci-C4)alkyl or heteroaryl are designated as being optionally substituted, the substituent groups which are optionally present may be one or more of those customarily employed tn the development of pharmaceutical compounds or the modification of such compounds to influence their structure/activity, persistence, absorption, stability or other beneficial property Specific examples of such substituents include halogen atoms, πitro, cyano, thiocyanato, cyanato, hydroxyl, alky!, haloaikyl, alkoxy, haloalkoxy, amino, alkylamino, dialkylamino, formyl, alkoxycarbonyl, carboxyl, alkanoyl, alkylthio, alkylsuphinyl, alkylsuiphonyl, carbamoyl, alkylamido, phenyl, phenoxy, benzyl, benzyloxy, heterocyclyl (e g , heteroaryl, cycloheteroalkyl) or cycloalkyl groups, preferably halogen atoms or lower alkyl or lower aikoxy groups, where lower denotes 1-6 carbon atoms Typically, 0-3 substituents may be present When any of the foregoing substituents represents or contains an alkyl substituent group, this may be linear or branched and may contain up to 12, preferably up to 6, more preferably up to 4 carbon atoms
Pharmaceutically acceptable salts may be any acid addition salt formed by a compound of formula I and a pharmaceutically acceptable acid such as phosphoric, sulfuric, hydrochloric, hydrobromic, citric, maleic, malonic, mandehc, succinic, fumaric, acetic, lactic, nitric, sulfonic, p-toluene sulfonic, methane sulfonic acid or the like
Compounds of the invention include esters, carbamates or other conventional prodrug forms, which in general, are functional derivatives of the compounds of the invention and which are readily converted to the inventive active moiety in vivo Correspondingly, the method of the invention embraces the treatment of the various conditions described heremabove with a compound of formula I or with a compound which is not specifically disclosed but which, upon administration, converts to a compound of formula I in vivo Also included are metabolites of the compounds of the present invention defined as active species produced upon introduction of these compounds into a biological system
Compounds of the invention may exist as one or more tautomers One skilled m the art will recognize that compounds of formula I may also exist as the tautomer It as shown below
_g_
(It)
Tautomers often exist in equilibrium with each other. As these tautomers interconvert under environmental and physiological conditions, they provide the same useful biological effects. The present invention includes mixtures of such tautomers as well as the individual tautomers of Formula I and Formula It.
The compounds of the invention may contain one or more asymmetric carbon atoms or one or more asymmetric (chiral) centers and may thus give rise to optica! isomers and diastereomers. Thus, the invention includes such optical isomers and disastereomers; as well as the racemic and resolved, enantiomerically pure stereoisomers; as well as other mixtures of the R and S stereoisomers. One skilled in the art will appreciate that one stereoisomer may be more active or may exhibit beneficial effects when enriched relative to the other stereoisomers) or when separated from the other stereoisomerfs). Additionally, the skilled artisan knows how to separate, enrich or selectively prepare said stereoisomers. Accordingly, the present invention comprises compounds of Formula 1, the stereoisomers thereof, the tautomers thereof and the pharmaceutically acceptable salts thereof. The compounds of the invention may be present as a mixture of stereoisomers, individual stereoisomers, or as an optically active or enantiomerically pure form. The point of attachment may be via Z in which case to fulfil valency requirements R11 is absent.
When n is 0, an example of the five membered ring is pyrazoly! such as pyrazol-4-yJ (i.e., X and Y are N), which ring may be optionally substituted, e.g., 1- ethylpyrazol-4-yl or 1 -(2,2,2~trifluoroethy)pyrazol-4-yl.
.9- When n is 1 , an example of the six membered ring is pyridyl such as pyrid-4- yl, or phenyl, which rings may be optionally substituted, e.g., 2,6-diethylpyrid-4-yl or 4-trifluromethoxyphenyl.
Preferred compounds of the invention are those compounds of formula ! wherein R1 and R2 are H. Another group of preferred compounds of the invention are those compounds of formula I wherein R9 is an optionally substituted heteroaryl group. Also preferred are those formula I compounds wherein X is N. A further group of preferred compounds of the invention are those compounds of formula I wherein R9 is an optionally substituted heteroaryl group and is attached to the phenyl ring in the 3-position of the phenyl ring.
More preferred compounds of the invention are those compounds of formula 1 wherein R1 and R2 are H and Rg is an optionally substituted heteroaryl group. Another group of more preferred compounds of the invention are those compounds of formula I wherein R1 and R2 are H; R9 is an optionally substituted heteroaryl group; and X is N. A further group of more preferred compounds of the invention are those compounds of formula i wherein R1 and R2 are H and R9 is an optionally substituted heteroaryl group and is attached to the phenyl ring in the 3-position of the phenyl ring. Preferred compounds of formula I include:
8-E3-(2-fluoropyridin-3-yl)-pheny!]-8-pyridin-4-yl-2,3»4,8-tetrahydroimidazo[1 ,5- a]pyrimidin-6-amine; 8-(2,6-diethylpyridin-4-yl)-8-[3-(2-fiuoropyridin-3-yl)-phenyl)-2,3,4,8-tetrahydro- imidazo[1 ,5-a]pyrimidin-6-amine; 8-{1-ethyI-1 H-pyrazol-4-yl)-8-t3-(2-fluoropyridin-3-yl)phenyl3-2,3,4!8-tetrahydro- imidazo[1 ,5-a]pyrimidin-6-amine; 8-[3-(2-fluoropyridin-3-yl}phenyl]-8-[1-(2,2,2-trif!uoroethyl)-1H-pyrazol-4-yl]-2,3,4,8- tetrahydroimidazo[1 ,5-a]pyrimidin-6-amine;
8-[3-(2-fIuoropyridin-3-yt)phenyl]-8-[4-(trifluoromethoxy)phenyl]-3,4-dihydro-8H- imidazo[5, 1 -c][1 ,2,4]oxadiazin-6-amine;
8-[3-(5-fluoropyridin-3-yI)phenyl]-8-[4-(trifluoromethoxy)phenyl]-3,4-dihydro-8H- imidazo[5, 1 -cj[1 ,2,4]oxadiazin-6-amine; the tautomers thereof; the stereoisomers thereof; or the pharmaceutically acceptable salts thereof.
Advantageously, the present invention provides a process for the preparation of a compound of formula i wherein R9 is an optionally substituted aryl or heteroaryf group (Ia) which comprises reacting a compound of formula Il wherein Hal is Cl or Br with an optionally substituted aryl or heteroaryl group having a leaving group selected from B(OH)2, Sn(Bu)3 or Sn(CH3)3 in the presence of a palladium catalyst and an inorganic base optionally in the presence of a solvent. The process is shown in flow diargram I, wherein A represents an optionally substituted aryl or heteroaryl group; W is B(OH)2, Sn(Bu)3 or Sn(CH3J3; and Hal is Cl or Br.
Flow Diagram I
(H)
(Ia) Palladium catalysts suitable for use in the process of the invention include
Pd(O) or Pd(H) catalysts such as dichlorobis(tri-o-tolyfphosphine)palladium(ll), Pd(OCOCH3)2/tri-o-tolylphosphine, tetrakis(triphenylphosphine)pa!ladium(0), tris(dibenzylideneacetone)dipallad!um(0)triphenylphosphine, or the like.
Inorganic bases suitable for use in the inventive process include Na or K hydroxides, carbonates or bicarbonates, preferably Na2CO3 Or K2CO3.
Solvents suitable for use in the inventive process include polar or non-polar organic solvents such as toluene, diethoxy ethyl ether, dioxane, ethyleneglycol dimethyl ether or any non-reactive organic solvent which is capable of solubiiizing the formula Il or heteroaryl compounds. Compounds of formula Il may be prepared using conventional synthetic methods and, if required, standard separation or isolation techniques. For example, compounds of formula Il wherein Rt and R2 are H and Q and W are CH2 (Ha), may be prepared by reacting a compound of formula IM with a diamine of formula IV to give the bicyclic compound of formula V and reacting said formula V compound with t- butyl hydroperoxide and ammonium hydroxide to give the desired formula Ma compound. The reaction is shown in flow diagram Il wherein Hal is Cl or Br,
FLOW DIAGRAM Il
(Ha)
Similarly, compounds of formula Il wherein Rt and R2 are H; R3 and R4 are H; Q is CH2; and W is O (lib) may be prepared by reacting the formula III compound with 2-(aminooxy)ethanamine dihydrochloride in the presence of a base such as triethylamine and a solvent to give the bicyclic compound of formula Vl and reacting said formula Vl compound with t-butyl hydroperoxide and ammonium hydroxide to give the desired formula Mb compound. The reaction is shown in flow diagram III wherein Hal is Cl or Br. Flow Diagram III
(Hb)
Compounds of formula Il wherein R1 and R2 are H; R3 and R4 are H; Q is O; and W is CH2 (lie) may be prepared by reacting the formula ill compound with Boc- protected 2-(aminooxy)ethanamine give the protected amine compound of formula VIi; deprotecting said formula VII compound in the presence of an acid such as trifluoroacetic acid to give the corresponding free amine of formula VIII; and cyclizing the formula VIiI compound to give the bicycltc compound of formula IX and reacting said formula IX compound with t-butyl hydroperoxide and ammonium hydroxide to give the desired formula Hc compound. The reaction is shown in flow diagram IV wherein Boc is t-butylcarbonyioxy and Hal is Cl or Br.
Flow Diagram IV
(VOl) (IX)
(lie)
Compounds of formula Ha, lib and ilc may be converted to the corresponding compounds of formula Ia wherein R1 and R2 are H using the procedure described hereϊnabove in flow diagram I.
Compounds of formula Hi may be readily prepared by reacting a heteroaryi halide compound of formula X with a benzonitriie compound of formula Xl in the presence of a base such as t-butyl lithium to give the methylamϊne of formula XM: reacting said formula XII amine with thiophosgene in the presence of a base such as NaHCO3 to give the thiocyanate compound of formula XlII; and reacting said formula XIlI thiocyanate with carbon disulfide in the presence of a base such as potassium t- butoxide to give the desired compound of formula IEI. The reaction is shown in flow diagram V wherein Hal represents Cl or Br. FLOW DIAGRAM V
(aq) 3. MeOH, NaBH4
(XIII)
OH)
Compounds of formula I wherein R9 is other than an optionally substituted aryl or heteroary! group may be prepared by the reactions shown heretnabove in flow diagrams il through IV and employing the corresponding formula EII compound wherein Hal is replaced by the desired R9 substituent.
Compounds of formula 1 wherein R1 and R2 are other than H may be prepared using standard aikylation techniques such as reacting the compound of formula ! wherein R1 and R2 are H with an alkyl halide, R1-HaI1 to give the compound of formula I wherein R2 is H (Id) and optionally reacting said formula Id compound with a second aikyi halide, R2-HaI, to give the desired formula I compound wherein R1 and R2 are other than H. Advantageously, the compounds of the invention are useful for the treatment, prevention or amelioration of a disease or disorder characterized by elevated β-amyloid deposits or β-amyloid levels in a patient, including Alzheimer's disease, Downs Syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch type or other neurodegenerative or dementia-inducing disorders. Accordingly, the present invention provides a method for the treatment, prevention or amelioration of a disease or disorder characterized by elevated β-amyloid deposits or β-amyloid levels in a patient which comprises providing said patient with a therapeutically effective amount of a compound of formula I as described hereinabove. The compound may be provided by oral or parenteral administration or in any common manner known to be an effective administration of a therapeutic agent to a patient in need thereof.
The term "providing" as used herein with respect to providing a compound or substance embraced by the invention, designates either directly administering such a compound or substance, or administering a prodrug, derivative or analog which forms an equivalent amount of the compound or substance within the body. The therapeutically effective amount provided in the treatment of a specific
CNS disorder may vary according to the specific condition(s) being treated, the size, age and response pattern of the patient, the severity of the disorder, the judgment of the attending physician and the like. In general, effective amounts for daily oral administration may be about 0.01 to 1,000 mg/kg, preferably about 0.5 to 500 mg/kg and effective amounts for parenteral administration may be about 0.1 to 100 mg/kg, preferably about 0.5 to 50 mg/kg.
In actual practice, the compounds of the invention are provided by administering the compound or a precursor thereof in a solid or liquid form, either neat or in combination with one or more conventional pharmaceutical carriers or excipients. Accordingly, the present invention provides a pharmaceutical composition which comprises a pharmaceuticaliy acceptable carrier and an effective amount of a compound of formula I as described hereinabove.
Solid carriers suitable for use in the composition of the invention include one or more substances which may also act as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aides, binders, tablet-disintegrating agents or encapsulating materials. In powders, the carrier may be a finely divided solid which is in admixture with a finely divided compound of formula I. In tablets, the formula I compound may be mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired. Said powders and tablets may contain up to 99% by weight of the formula I compound. Solid carriers suitable for use in the composition of the invention include calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, polyvinylpyrroiidine, low melting waxes and ion exchange resins.
Any pharmaceutically acceptable liquid carrier suitable for preparing soiutions, suspensions, emulsions, syrups and elixirs may be employed in the composition of the invention. Compounds of formula f may be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, or a pharmaceutically acceptable oil or fat, or a mixture thereof. Said liquid composition may contain other suitable pharmaceuticai additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, coloring agents, viscosity regulators, stabilizers, osmo-regulators, or the like. Examples of liquid carriers suitable for oral and parenteral administration include water (particularly containing additives as above, e.g., cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) or their derivatives, or oils (e.g., fractionated coconut oil and arachis oil). For parenteral administration the carrier may also be an oily ester such as ethyl ofeate or isopropyl myristate.
Compositions of the invention which are sterile solutions or suspensions are suitable for intramuscular, intraperitoneal or subcutaneous injection. Sterile solutions may also be administered intravenously. Inventive compositions suitable for oral administration may be in either liquid or solid composition form.
Alternatively, the use of sustained delivery devices may be desirable, in order to avoid the necessity for the patient to take medications on a daily basis, "Sustained delivery" is defined as delaying the release of an active agent, i.e., a compound of the invention, until after placement in a delivery environment, followed by a sustained release of the agent at a later time. Those of skill in the art know suitable sustained delivery devices. Examples of suitable sustained delivery devices include, e.g., hydrogels (see, e.g., US Patent Nos. 5,266,325; 4,959,217; and 5,292,515), an osmotic pump, such as described by Alza (US Patent Nos. 4,295,987 and 5,273,752) or Merck (European Patent No. 314,206), among others; hydrophobic membrane materials, such as ethyienemethacryiate (EMA) and ethylenevinylacetate (EVA); bioresorbable polymer systems (see, e.g., International Patent Publication No. WO 98/44964, Bioxid and Celiomeda; US Patent Nos. 5,756,127 and 5,854,388); other bioresorbable impiant devices have been described as being composed of, for example, polyesters, poiyanhydrides, or lactic acid/g!ycoiic acid copolymers (see, e.g., US Patent No. 5,817,343 (Alkermes Inc.)}. For use in such sustained delivery devices, the compounds of the invention may be formulated as described herein. In another aspect, the invention provides a pharmaceutical kit for delivery of a product. Suitably, the kit contains packaging or a container with the compound formulated for the desired delivery route. For example, if the kit is designed for administration by inhalation, it may contain a suspension containing a compound of the invention formulated for aerosol or spray delivery of a predetermined dose by inhalation. Suitably, the kit contains instructions on dosing and an insert regarding the active agent. Optionally, the kit may further contain instructions for monitoring circulating levels of product and materials for performing such assays including, e.g., reagents, well plates, containers, markers or labels, and the like. Such kits are readily packaged in a manner suitable for treatment of a desired indication. For example, the kit may also contain instructions for use of the spray pump or other delivery device.
Other suitable components to such kits will be readily apparent to one of skill in the art, taking into consideration the desired indication and the delivery route. The doses may be repeated daily, weekly, or monthly, for a predetermined length of time or as prescribed.
For a more clear understanding, and in order to illustrate the invention more clearly, specific examples thereof are set forth hereinbelow. The following examples are merely illustrative and are not to be understood as limiting the scope and underlying principles of the invention in any way. Indeed, various modifications of the invention, in addition to those shown and described herein, will become apparent to those skilled in the art from the examples set forth hereinbelow and the foregoing description. Such modifications are also intended to fall within the scope of the appended claims.
Unless otherwise noted, all parts are parts by weight. The term NMR designates nuclear magnetic resonance. The terms DME and DMF designate ethylene glycol dimethyl ether and dimethyl formamide, respectively. EXAMPLE 1
Preparation of 843-f2-Fluoropyridin-3-yl)phenvπ-8-pyridin-4-yl-2.3,4,8- tetrahvdrotmidazori,5-a1pyrimidin-6-amine
J-BuOOH
5: 1 MeOHz1NH4OH
A) Preparation of Compound 1
A solution of te/f-butyl lithium (30.0 mL of 1.7 M in pentane, 51.0 mmol) in diethyl ether, at -78 0C, was treated dropwise with a solution of 4-bromopyridine (25.7 mmol) in diethyl ether, stirred at -78 0C for 40 min, allowed to warm to 0 0C, treated sequentially with methanol and sodium borohydride (1.94 g( 51.0 mmol), stirred overnight at room temperature, diluted with saturated aqueous ammonium chloride and concentrated under reduced pressure to remove most of the methanol and diethyl ether. The resultant aqueous residue was extracted with methylene chloride. The extracts were combined, washed with brine, dried over sodium sulfate and concentrated in vacuo. Purification of this residue by flash chromatography (silica, methylene chloride to 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded compound 1 as a yellow syrup, 4.21 g (69% yield), 1H NMR (300 MHz1 CDCI3) δ 8.54 (dd, J = 4.5, 1 ,5 Hz, 2H), 7.36-7.28 (m, 6H)1 5.14 (s, 1 H), 1.25 (s, 2H, br); ESl MS m/z 264 [Ci2H11BrN2+ H]+, B) Preparation of Compound 2
A mixture of potassium terf-butoxide (0.355 g, 3.16 mrnol) in tetrahydrofuran at -78 0C was treated dropwise with a solution of 1 (0.665 g, 2.53 mmol) in tetrahydrofuran, stirred for 10 mtn, treated with carbon disulfide (0.635 g, 8,34 mmol), allowed to warm to room temperature slowly and stirred for 1 h at room temperature. The reaction mixture was cooled to -78 0C1 treated with di-2-pyridyl-thiocarbonate (0.880 g, 3.79 mmol), allowed to warm to room temperature, stirred overnight at room temperature and concentrated in vacuo. The resultant residue was purified by flash chromatography (silica, 97.5:2.5 methylene chloride/methanol) to give compound 2 as a pink oil, 0.310 g (32% yield), 1H NMR (300 MHz, CDCf3) δ 8.66 (dd, J = 4.5, 1.5 Hz, 2H), 7.57 (dt, J = 6.5, 2.0 Hz, 1 H), 7.47 (d, J = 2.0 Hz, 1 H), 7.34 (ddr J = 4.5, 1.5 Hz, 2H), 7.32-7.26 (m, 2H); ESI MS m/z 381 [C14H9BrN2S3 + H]+. C) Preparation of Compound 3
A mixture of 2 (0.31 O g, 0.810 mmoi) and 1 ,3-diaminopropane (0.181 g, 2.44 mmol) in ethanoi was heated at 70 0C for 1 h, cooled to room temperature and concentrated to dryness, Purification of the resultant residue by flash chromatography (silica, 97.5:2.5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded compound 3 as a white solid, 0.260 g (83% yield), 1H NMR (500 MHz, CD3OD) δ 8.57 (dd, J = 4.5, 1.5 Hz, 2H), 7.56-7.52 (m, 2H), 7.41 (dd, J = 4.5, 1.5 Hz, 2H), 7.36-7.33 (m, 2H), 3.86 (t, J = 6.0 Hz1 2H)r 3.56 (t, J = 5.5 Hz, 2H), 1.88 (tt, J = 6.0, 5.5 Hz, 2H); ESI MS m/z 387 [C17H15BrN4S + H]+. D) Preparation of Compound 4 A mixture of 3 (0.260 g, 0.670 mmol) and t-butyl hydroperoxide (1.73 g of a
70% solution in water, 13.4 mmol) in methanol and concentrated aqueous ammonium hydroxide was stirred overnight at room temperature and concentrated in vacuo. Purification of the resultant residue by flash chromatography (sifica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded compound 4 as a white solid, 0.21O g (84% yield), 1H NMR (300 MHz, CDCI3) δ 8.48 (dd, J = 4.5, 1.5 Hz, 2H), 7.52 (t, J = 1.5 Hz, 1 H), 7.46 (dt, J = 7.5, 1.5 Hz1 1 H), 7.40 (dd, J = 4.5, 1.5 Hz, 2H), 7.32 (dt, J = 7.5, 1.5 Hz, 1 H), 7.25 (t, J = 7.5 Hz, 1 H), 3.69 (t, J = 6.0 Hz, 2H), 3.50 (t, J = 5.1 Hz, 2H)1 1.86 (tt, J = 6.0, 5.1 Hz, 2H); ESI MS m/z
370 [C17H16BrN5 + H]+.
D) Preparation of 8-[3-(2-Fluoropyridin-3-yl}phenyl]-8-pyridin-4-yi-2,3,4,8- tetrahydroimidazo[1,5-a]pyrimidin-6-amine A mixture of 4 (0.10O g, 0.27 mrnoi), 2-fluoropyridine-3-boronic acid (0.076 g,
0.540 mmoi), bis(triphenylphosphino)pafladium(ll) chloride (0.010 g, 0.014 mrnoi), triphenylphosphine (0.0071 g, 0.027 mmof) and sodium carbonate (0,086 g, 0.810 mmo!) in 3:1 DME/water was heated at reflux temperature for 2 h, cooled to room temperature and diluted with ethyl acetate and water. The phases were separated. The organic phase was washed with brine, dried over magnesium sulfate and concentrated in vacuo. Purification of the resultant residue by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded a white solid. This material was freeze dried from 2:1 acetonitrile/water to afford the title product as a white solid, 0.082 g (79% yield), mp 125-131 0C; 1H NMR (500 MHz, CDCI3) δ 8.16 (dd, J = 4.5, 1.5 Hz, 2H), 8.16 (dt, J = 5.0, 1.5 Hz, 1H), 7.83 (ddd, J = 9.5, 7.5, 2.0 Hz, 1 H), 7.70 (d, J = 1.5 Hz, 1 H), 7.52 (dt, J = 8.0, 1.5 Hz, 1 H), 7.47 (dd, J = 4.5, 1.5 Hz1 2H), 7.47-7.46 (m, 1 H), 7.41 (t, J = 7.5 Hz, 1 H), 7.24 (ddd, J = 7.5, 5.0, 2.0 Hz1 1H), 3.62-3.57 (m, 4H), 1.88 (m, 2H); ESI MS m/z 387 [C22H19FN6 + H]+.
EXAMPLE 2
Preparation of 8-(2,6-Diethylpyridin-4-v0-8-r3~(2-fluoropyridin-3-v0phenvπ- 2,3,4,8-tetrahvdroimidazof1,5-a1pyrimidin-6-amine
PPh3
Using essentially the same procedure described in Example 1 and employing 4-Bromo~2,6~diethylpyrid!ne in Step A, the title product is obtained as a white solid, 0.095 g, mp 174-176 0C; 1H NMR (500 MHz, CDCI3) δ 8.16 (d, J = 4.2 Hz, 1 H), 8.04- 7.98 (m, 1 H), 7.54-7.37 (m, 5H), 7.13 (S1 2H), 3.70 (t, J = 6.0 Hz, 2H), 3.50 (t, J = 6.0 Hz, 2H), 2.75 (q, J = 7.8 Hz, 4H), 1.87 (t, J = 5.7 Hz, 2H), 1.23 (t, J = 7.8 Hz, 6H); ESI MS m/z 443 [C26H27FN6 + H]+
EXAMPLE 3
Preparation of 8-(1-EtHyH H-pyrazol-4-vπ-β-r3-(2-fluoropyridin-3-yl)phenvn- 2,3,4,8-tetrahvdroimidazof1,5-a1pyrimidin-β-amine
?-BuOOH NH4OH PPh3
A) Preparation of Compound 1 A mixture of f-butyl lithium (16.2 mL of a 1.7 M solution in pentane, 27.5 mmol) and diethyl ether was cooied to -78 0C1 treated dropwise over a 15 min. period with a solution of 4-bromo-1 -ethyl pyrazole (2.3 g, 13.1 mmol) in diethyl ether, stirred at -78 0C for 10 min, treated dropwise with a solution of 3-bromobenzonitrile (2.58 g, 14.1 mmol) in ether, stirred at -78 0C for 45 min and allowed to warm to room temperature for1 h. The reaction mixture was treated with anhydrous methanol, cooled to 0 0C, treated with sodium borohydride (0.991 g, 26.2 mmol), warmed to room temperature, stirred for 1 h at to room temperature, cooled to 0 0C and quenched by the careful addition of saturated ammonium chloride until gas evolution had ceased and ail precipitates had dissolved. The reaction mixture was diluted with methylene chloride and water. The phases were separated. The organic phase was washed with brine, dried over sodium sulfate and concentrated in vacuo. Purification of the resultant residue by flash chromatography (silica, methylene chloride/methanol 95:5) afforded compound 1 as a colorless oil, 1.91 g (52% yield), 1H NMR (300 MHz, CDCI3) δ 7.54 (br s, 1H), 7.41-7.35 (m , 2H), 7.32^7.27 (m, 1H), 7.24-7,16 (m, 2H), 5.11 (s, 1 H), 4.10 (q, J = 7.3 Hz, 2H), 1.89 (br s, 2H), 1.44 (t, J = 7.3 Hz, 3H); ESI MS m/z 263 [(C12H14BrN3 - NH2) + H]+.
B) Preparation of Compound 2 A mixture of 1 (0.112 g, 0.40 rnmoi) in methylene chloride and saturated aqueous sodium bicarbonate was cooled with an ice bath, treated with thtophosgene (0.05 g, 0.44 mmol), stirred vigorously for 30 min and diluted with methylene chloride. The phases were separated, The organic phase was washed with brine, dried over sodium sulfate and concentrated to afford compound 2 as a yellow syrup, 0.1 1 g (84% yield), 1H NMR (300 MHz, CDCI3) δ 7.51-7.45 (m, 2H), 7.37 (s, 1H), 7.31-7.24 (m, 3H), 5.93 (S1 1 H), 4.14 (q, J = 7.3 Hz, 2H), 1.47 (t, J = 7.3 Hz, 3H).
C) Preparation of Compound 3
A mixture of potassium f-butoxtde (0.04 g, 0.37 mmo!) in tetrahydrofuran at -78 0C was treated dropwise over a period of 2 min. with a solution of 2 (0.11 g, 0.34 mmol) and carbon disulfide (0.04 g, 0.51 mmol) in tetrahydrofuran, stirred at -78 0C for 0.5 h, slowly warmed to room temperature, stirred at room temperature for 1 h and diluted with methylene chloride and water. The phases were separated. The organic phase was washed with brine, dried over sodium sulfate and concentrated to afford compound 3 as a yellow solid, 0.089 g (66% yield), 1H NMR (300 MHz, DMSO- Of6) δ 7.88-7.26 (m, 6H)1 4.15, 4.06 (2q, J = 7.3 Hz, 2H), 1.41-1.39 (m, 3H); ESI MS m/z 398 [C14H12BrN3S3 + H]+.
D) Preparation of Compound 4
A mixture of 3 (0.50 g, 1.25 mmol) and 1 ,3-diaminopropane (0.28 g, 3.75 mmol) in ethanol was heated at 70 0C for 1 h, cooled to room temperature and evaporated under reduced pressure. The resultant residue was partitioned between ethyl acetate and water. The organic phase was separated, washed with brine, dried over sodium sulfate and concentrated in vacuo to afford compound 4 as a pale yellow oil, 0.38 g (75% yield), 1H NMR (300 MHz, CDCI3) δ 7.63 (s, 1 H), 7.53-7.47 (m, 2H), 7.43-7.23 (m, 3H), 4.15 (q, J = 7.3 Hz, 2H), 3.83 (t, J = 6.0 Hz, 2H), 3.53- 3.45 (m, 2H), 1.90-1.83 (m, 2H), 1.43 (t, J = 7.3 Hz, 3H); ESI MS m/z 404 [C17H18BrN5S + H]+.
E) Preparation of Compound 5 A mixture of compound 4 (0.38 g, 0,94 mmol) and f-butyl hydroperoxide (3.6 g of a 70% solution in water, 28.2 mmol) in methanol and concentrated aqueous ammonium hydroxide was stirred overnight at room temperature, treated with 10% aqueous sodium thiosulfate (30 ml_) and concentrated under reduced pressure to remove most of the methanol. The resultant aqueous mixture was extracted with methylene chloride. The extracts were combined, washed sequentially with water and brine, dried over sodium sulfate and concentrated in vacuo. Purification of this residue by flash chromatography (silica, 95:5:0,25 methylene chloride/methanol/ concentrated ammonium hydroxide) afforded compound 5 as a colorless syrup, 0.09 g (25% yield), 1H NMR (300 MHz, CDCI3) δ 7.66 (s, 1H), 7.48-7.34 (m, 4H), 7.17 (t, J = 7.8 Hz, 1 H), 4.1 1 (q, J = 7.3 Hz, 2H), 3.59 (t. J = 5.8 Hz5 2H), 3.58-3.52 (m, 2H), 1.91-1.80 (m, 2H), 1.46 (t, J = 7.3 Hz, 3H); ESI MS m/z 387 [C17H19BrN6 + H]+. Preparation of 8-(1-Ethyl-1 H-pyrazol-4-yl)-8-[3-{2-fluoropyridin-3-yl)phenyl]- 2,3,4,8-tetrahydroimidazo[1,5-a]pyrimidin-6-amine A mixture of 5 (0.090 g, 0.230 mmol), 2-fluoropyridine-3-boronic acid (0.065 g, 0.460 mmol), bis(triphenylphosphino)palladium(ll) chloride (0.008 g, 0.011 mmol), triphenylphosphine (0.006 g, 0.022 mmol) and sodium carbonate (0.073 g, 0.690 mmol) in 3:1 DME/water was heated at reflux temperature for 1.5 h, cooled to room temperature and diluted with ethyl acetate and water. The phases were separated. The organic phase was washed with brine, dried over magnesium sulfate and concentrated in vacuo. Purification of the resultant residue by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded 0.035 g of an off-white solid. This material was freeze dried from 2:1 acetonitrile/water to afford a white solid, which was contaminated with dimethylformamide. Purification of this solid by flash chromatography {silica,
95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) to afford a white solid, which was freeze dried from 2:1 acetonitrile/water to afford the title product as a white solid, 0.034 g (35% yield), mp 91-115 0C; 1H NMR (300 MHz, CDCI3) δ 8.20-8.12 (m, 1H), 7.90-7.81 (m, 1 H), 7.71 (br s, 1 H), 7.59-7.40 (m, 6H), 4.12 (q, J = 7,3 Hz, 2H), 3.70 (t, J = 6.0 Hz, 2H), 3.57 (t, J = 4.9 Hz, 2H), 1.95-1.82 (m, 2H), 1.46 (t, J = 7.3 Hz, 3H); ESI MS m/z 404 [C22H22FN7 + H]+. EXAMPLE 4
Preparation of 8-[3-f2-Fluoropyridin-3-yl)phenvn-8-ri-f2.2.2-trifluoroethyl)-1 H- pyrazol-4-vn-2,3,4,8-tetrahvdroimidazori,5-a1pyrimidin-6-amine
Using essentially the same procedure described in Example 3 and employing 1(4-bromophenyl)-1-[(2s2,2-trifIuoroethyl)pyrazol-4-yl]methylamine in Step B, the title product was obtained as a white solid, mp 106-116 °C, 1H NMR (300 MHz, CDCI3) δ 8.17 (dd, J = 1.7, 1.6 Hz, 1 H), 7.82 (m, 1H), 7.68 (d, J = 1.4 Hz, 1H), 7.63 (s, 1 H), 7.59 (s, 1 H), 7.57-7.41 (m, 3H), 7.23 (m, 1 H)1 4.68 (q, J = 8.4 Hz5 2H), 3.60 (t, J = 5.9 Hz, 2H), 3.54 (m, 2H), 1.85 (m, 2H); ESi MS m/z 458 [C22H19F4N7 + H]+.
EXAMPLE S
Preparation of 843-(2-Fluoropyridin-3-v0ρhenvπ-8-r4-(triftuoromethoxy)- phenvπ-3.4-dihvdro-8H-imidazof5.1-ciri,2,41oxadiazin-6-amine
A} Preparation of Compound 1
A mixture of 4-(3-bromophenyl)-4-[4-(trifluorornethoxy)phenyl]-1,3- thiazolidine-2,5-dithione (0.50 g, 1.08 mmol), 2-(aminooxy)ethanamine (0.48 g, 3.23 mmoi, prepared as described in J, Med. Chem. 2000, 43(12), 2347) and triethylamSne (0.71 g, 7.00 mmoi) in ethanol was stirred at ice bath temperatures for 2 h, warmed to room temperature, stirred at room temperature for 24 h, heated to 70 0C, stirred at 70 0C for 2 h, cooled to room temperature and concentrated under reduced pressure. The resultant residue was partitioned between ethyl acetate and water. The organic phase was washed sequentially with 1 N HCI and brine, dried over magnesium sulfate and concentrated in vacuo. Purification of this residue by flash chromatography (silica, 1 :5 ethyl acetate/hexanes) afforded compound 1 as a white solid, 0.277 g (54% yield), 1H NMR (300 MHz, CDCI3) δ 7.53 (m, 2H), 7,39 (d, J = 8.9 Hz, 2H), 7.38-7.22 (m, 4H), 4.11 (m, 2H), 4.03 (m, 2H); ESi MS m/z 472 [Ct8H13BrF3N3O2S + H]+. B) Preparation of Compound 2
A mixture of compound 1 (0.27 g, 0.571 mmol) and (--butyl hydroperoxide (1.47 g of a 70% solution in water, 11.4 mmol) in methanol and concentrated aqueous ammonium hydroxide was stirred overnight at room temperature, treated with 10% aqueous sodium thiosulfate and concentrated to remove most of the methanol. The resultant aqueous mixture was extracted with methylene chloride. The extracts were combined, washed with brine, dried over magnesium sulfate and concentrated in vacuo. Purification of this concentrate by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded compound 2 as a white solid, 0.166 g (64% yield), 1H NMR (300 MHz, CDCI3) δ 7.70 (t, J = 1.8 Hz, 1H), 7.56 (m, 1H). 7.45-7.37 (m, 3H), 7.20-7.13 (m, 3H), 3.99 (m, 2H), 3.77 (m, 2H); ESI MS m/z 456 [C18H14BrF3N4O2 + H]*. C) Preparation of 8-|3-(2-FIuoropyridin-3-yl)phenyl]-8-[4-(trifIuoromethoxy)- pheny[I-3,4-dihydro-8H-imidazo[5,1"C][1,2,4]oxadia2in-6-amine
A mixture of 2 (0.16 g, 0.351 mmoi), 2-fluoropyridine»3-boronic acid (0.089 g, 0.633 mmoi), bis(triphenylphosphino)palladium(ll) chloride (0.012 g, 0,018 mmol), triphenylphosphine (0-0092 g, 0.035 mmol) and sodium carbonate (0.112 g, 1.05 mmol) in 3:1 DME/water was heated at reflux for 3 h, cooled to room temperature and diluted with ethyl acetate and water. The organic phase was separated, washed with brine, dried over magnesium sulfate and concentrated. Purification of the resuntant concentrate by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded 0.12 g of a white solid. This material was freeze dried from 2:1 acetonitrile/water (6 m!_) to afford the title product as a white solid, 0.109 g (66% yield), mp 102-117 0C; 1H NMR (300 MHz1 CDCI3) δ 8.16 (m, 1 H), 7.83 (m, 1 H), 7.71 <m, 1 H), 7.61-7.56 (m, 3H), 7.46- 7.41 (m, 2H), 7.25 (m, 1H), 7.15 (m, 2H), 4.00 (m, 2H), 3.78 (m, 2H); ESI MS m/z 472 [C23H17F4N5O2 + Hf.
EXAMPLE 6
Preparation of 8-r3-(5-Fluoropyridin-3-yl)phenvπ-844-(triftuoromethoxy)phenvπ- 2,8-dihvdro-3H-imidazof 1 ,5-bif 1 ,2,4]oxadiazin-β-amine
MBuOOH
McOHZKH4OH
A) Preparation of Compound 1
A mixture of 4-(3-bromophenyl)-4-[4-(trifluoromethoxy)phenyl]-1 ,3- thiazoiidine-2,5-dithione (1 ,32 g, 2.84 mmoi) and Boc-protected 2-
(aminooxy)ethanamine (1.49 g, 8.53 mmo!) in ethanoi was stirred at 70 0C for 1.5 h, cooled to room temperature and concentrated under reduced pressure. The resultant residue was partitioned between ethyl acetate and water. The organic phase was separated, washed with brine, dried over sodium sulfate and concentrated in vacuo. Purification of this residue by flash chromatography ( silica, 1 :4 ethyl acetate/hexanes) afforded compound 1 as a white solid, 1.12 g (65% yield), 1H NMR (SOO MHz, CDCl3) δ 7.72 (bs, 1H), 7.56 (dt, J = 4.1 , 1.6 Hz, 1H), 7.49 (d, J = 1.8 Hz1 1H), 7.38 (d, J = 8.7 Hz, 2H), 7.30 (m, 3H), 4.68 (bs, 1 H)1 4.21 (t, J = 5.1 Hz1 2H), 3.37 (bs, 2H), 1.43 (s, 9H); ESI MS m/z 607 [C23H23BrF3N3O4S2 + H]+. B) Preparation of Compound 2
A mixture of compound 1 (1.12 g, 1.85 mmol), trifluoroacetic acid (6.0 mL) and methylene chloride was stirred at ambient temperatures for 1 h and concentrated under reduced pressure. The concentrate was basified with 10% aqueous potassium carbonate to pH 9 and extracted with methylene chloride. The extracts were combined, dried over sodium sulfate and concentrated to afford compound 2 as an off-white solid, 0.842 g (90% yield), 1H NMR (500 MHz, CDCI3) δ 7.21™7.60 (m, 6H)1 7.11-7.25 (m, 2H), 4.22 (t, J = 5.3 Hz5 2H), 2.94 (t, J = 5.2 Hzt 2H), 1.62 (s, 2H); ESI MS m/z 507 [C18H15BrF3N3O2S2 + H]+.
C) Preparation of Compound 3 A solution of compound 2 (0.842 g, 1.66 mmol) in ethanol was heated at reflux temperature for 1 h, cooled to room temperature and concentrated in vacuo. The resultant residue was purified by flash chromatography (silica, 3:1 hexanes/ethy! acetate) to afford compound 3 as an off-white solid, 0.437 g (56% yield), 1H NMR (500 MHz, CDCI3) δ 7.63 (m, 2H), 7.39 (dd, J = 4.6, 2.1 Hz, 2H), 7.28 (m, 2H), 7.23 (d, J = 8.0 Hz, 2H), 4.11 (t, J = 2.9 Hz1 2H), 4.02 (t, J = 3.2 Hz, 2H); ESI MS m/z 473 [C18H13BrF3N3O2S t H]+;
D) Preparation of Compound 4
A mixture of compound 3 (0.434 g, 0.920 mmol) and f-butyl hydroperoxide (3.55 g of a 70% solution in water, 27.6 mmol) in methanol and concentrated aqueous ammonium hydroxide was stirred overnight at room temperature, treated with 10% aqueous sodium thiosulfate and concentrated to remove most of the methanol. The resultant aqueous mixture was extracted with methylene chloride. The extracts were combined, dried over sodium sulfate and concentrated to dryness. Purification of this residue by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/ concentrated ammonium hydroxide) afforded compound 4 as an off white solid, 0.284 g (68% yield), 1H NMR (500 MHz, CDCI3) δ 7.69 (t, J = 1.8 Hz, 1 H), 7.55 (dd, J = 4.7, 2.1 Hz, 2H), 7.42 (dt, J = 5.2, 1.0 Hz, 1 H), 7.40 (dt, J = 6.0, 1.0 Hz, 1 H), 7.18 (m, 3H), 4.00 (t, J = 4.5 Hz, 2H), 3.79 (t, J = 6.1 Hz, 2H); ESl MS m/z 456 [C18H14BrF3N4O2 + H]+. E) Preparation of 8-[3-(5-Fluoropyridin-3-yl)phenyl]-8-[4-{trifluoromethoxy)- phenyl]-2,8-dihydro-3H-imidazo[1 ,5-b][1 ,2,4]oxadiazi n-6-amiπe
A mixture of compound 4 (0.095 g, 0.209 mmol), compound 5 (0.122 g, 0.313 mmol), and dich!orobis(triphenylphosphine)palladium(ll) (0,007 g, 0.011 mmol) in DMF was degassed then heated at 150 °C in a sealed tube for 1.5 h. The mixture was cooled to room temperature and diluted with ethyl acetate and 5% aqueous LiCL The organic phase was separated, washed with 5% aqueous LiCI, dried over sodium sulfate and concentrated in vacuo. Purification of the resultant residue by flash chromatography (silica, 95:5:0.25 methylene chloride/methanol/concentrated ammonium hydroxide) afforded the title product as a white solid, 0.050 g (38% yield), mp 120-135 0C, 1 H NMR (SOO MHZ, CDCl3) § 8.67 (bs, 1 H)1 8.38 (d, J = 2.3 Hz, 1H)5 7.78 (bs, 1 H), 7.45-7.62 (m, 6H), 7.18 (d, J = 8.3 Hz, 2H), 4.05 (bs, 2H), 3.99 (bs, 2H); ESI MS m/z 472 [C23Hi7F4N5O2+ H]+.
EXAMPLE 7
Evaluation of the Enzyme Activity of Test Compounds and the Inhibition of hBACEL MuBACEI and hBACE2 by Test Compounds
Assay Conditions: 10 nM human BACE1 (or 10 nM Murine BACE1 , 1.5 nM human BACE2) 25 μM substrate (WABC-6, MW 1549.6, from AnaSpec); final buffer conditions:50 mM Na-Acetate, pH 4.5, 0.05% CHAPS, 25% PBS; temperature: room temperature; reagent information: Na-Acetate: Aldrich, Cat. # 24,124-5 CHAPS: Research Organics, Cat. # 1304C 1X PBS: Mediatech (Cellgro), Cat# 21-031 -CV; peptide substrate AbzSEVNLDAEFRDpa: AnaSpec, Peptide Name: WABC-6; determination of stock substrate (AbzSEVNLDAEFRDpa) concentration: a 25 mM stock solution in dimethyl sulfoxide (DMSO) is prepared using the peptide weight and MW and diluted to 25 μM. The concentration is determined by absorbance at 354 nm using an extinction coefficient ε of 18172 M"1cm"\ The substrate stock is stored in small aiiquots at -80° C. [Substrate Stock] = ABS 354 nm * 106 / 18172 (in mM)
Determination of Stock Enzyme Concentration: The stock concentration of each enzyme by ABS at 280 nm using i of 64150 M'1 cm'1 for hBACEI and MuBACEI , 62870 M*1 cm"1 for hBACE2 in 6 M Guanidinium Hydrochloride (from Research Organics, Cat. # 5134G-2), pH 6.
(The extinction coefficient ε 280 πm for each enzyme was calculated based on known amino acid composition and published extinction coefficients for Trp (5.69 M"1 cm"1) and Tyr (1.28 M"1 cm'1) residues (Anal. Biochem. 182, 319-326).
Dilution and mixing steps: total reaction volume: 100 μl_ 1. 2X inhibitor dilutions in buffer A(66.7 mM Na-Acetate, pH 4.5, 0.0667% CHAPS) are prepared,
2. 4X enzyme dilution in buffer A(66.7 mM Na-Acetate, pH 4.5, 0.0667% CHAPS) are prepared, 3. 100 μM substrate dilution in 1X PBS is prepared,
4. 50 μL 2X Inhibitor and 25 μL 100 μM substrate are added to each well of 96-well plate (from DYNEX Technologies, VWR #: 1 1311-046), the immediately 25 μL 4X enzyme are added to the inhibitor and substrate mixer, the fluorescence readings are initiated.
Fluorescence Readings: Readings at λex 320 nm and λem 420 nm are taken every 40-sec for 30 min at room temperature to determine the linear slope for substrate cleavage rate (v,).
Calculation of % inhibition: % inhibition = 100 * (1- v, / V0) (V1 = substrate cleavage rate in the presence of inhibitor, V0 = substrate cleavage rate in the absence of inhibitor) IC50 Determination: % Inhibition = [(B * IC50") + (100 * lo n)] / (IC50" + I0"),
Fluorescent Kinetic Assay for human recombinant BACE 2 This assay is used to provide kinetic and selectivity parameters for the analyses of the tested compounds.
Materials and methods: final assay conditions: 10 nM human BACE1 (or 10 nM Murine BACE1 , 1.5 nM human BACE2) 25 μM Substrate (WABC-6, MW 1549.6, from AnaSpec). Final buffer conditions: 50 mM Na-Acetate, pH 4.5, 0.05% CHAPS, 25% PBS. Temperature: room temperature. Reagent Information: Na- Acetate: Aldrich, Cat.# 24,124-5 CHAPS: Research Organics, Cat. # 1304C 1X PBS: Mediatech (Cellgro), Cat# 21 -031 -CV Peptide Substrate AbzSEVNLDAEFRDpa: AnaSpec, Peptide Name: WABC-6
Determination of stock substrate (AbzSEVNLDAEFRDpa) concentration: A 25 mM stock sofution in DMSO is prepared using the peptide weight and MW, and diluted to 25 μM. The concentration is determined by absorbance at 354 nm using an extinction coefficient ε of 18172 M~1cm*1. The substrate stock is stored in small aliquots at -80° C. [Substrate Stock] = ABS 3S4 nm * 106 / 18172 (in mM)
Determination of stock enzyme concentration: The stock concentration of each enzyme is determined by ABS at 280 nm using ε of 64150 M"1crrf1 for hBACEI and MuBACEI , 62870 M"1 cm 1 for hBACE2 in 6 M guanidinium hydrochloride (from Research Organics, Cat. # 5134G-2), pH 6. (The extinction coefficient ε28O rim for each enzyme is calculated based on known amino acid composition and published extinction coefficients for Trp (5.69 M'1 cm"1) and Tyr (1.28 M'1 cm"1) residues (Anal. Biochem. 182, 319-326).)
Dilution and Mixing Steps: Total Reaction Voume.: 100 μL
1. 2X inhibitor dilutions in buffer A(66.7 mM Na-Acetate, pH 4.5, 0.0667% CHAPS) are prepared,
2. 4X enzyme dilution in buffer A(66.7 mM Na-Acetate, pH 4.5, 0.0667% CHAPS) are prepared,
3. 100 μM substrate dilution in 1X PBS, 50 μL 2X inhibitor and 25 μL 100 μM substrate are added to each well of 96-we!l plate (from DYNEX Technologies, VWR #: 11311-046), then immediately 25 μL 4X enzyme is added to the inhibitor and substrate mixer and the fluorescence readings are initiated.
Fluorescence Readings: Readings at λex 320 nm, λem 420 nm are taken every 40-sec for 30 min at room temperature and to determine the linear slope for substrate cleavage rate (v,). Analysis of calculation of % inhibition: % Inhibition = 100 * (1- v, / v0) v, = substrate cleavage rate in the presence of inhibitor,
V0 = substrate cleavage rate in the absence of inhibitor)
IC50 Determination:
% Inhibition = ((B * IC5O n) + (100 * I0")) / (IC5O n + I0"),
The data obtained are shown in Table I below. Unless otherwise noted, the
IC5O value represents the value obtained at 100% inhibition.
TABLE I
Example BACE1 BACE2 Number IC50 IC50
1 0.52 51 % at 25 μM
2 0.01 38 % at 25 μM
3 0.09 3.52
4 0.08 5.12
5 0.03 2.02
6 0 05 1.19
Results and Discussion:
As can be seen from the data shown on Table I hereinabove, the compounds of the invention are potent and selective inhibitors of BACE1.

Claims

What is claimed is:
1. A compound of formula I
(D
wherein
Q is O, S or CH2;
W is O, S or CH2;
X is N, NO, SOm, O or CH;
Y is N, NO, SOmi O or CR10;
Z is N, NO, SOm, O or CRn with the proviso that when X is CH, Y is CR10 and Z is CRn then one of Q or W must be O or S; m is 0, 1 or 2; n is 0 or 1 ; R1 and R2 are each independently H or an optionaily substituted C1-
C4alkyl group; R3 and R4 are each independently H, or an optionally substituted C1-C4 alky! group or R3 and R4 may be taken together to form a 4- to 7- membered ring optionally containing one or two heteroatoms selected from O, N or S; R5 and R6 are each independently H, halogen, NO2, CN, ORi2, CO2R13,
COR14, NR17Ri3, SOpNR19R20 or a CrCβalkyt, CrC6haloalkyl, C2-
C6a!kenyl, C2-C6alkynyl or C3-C8cycloalkyl group each optionally substituted; R7 and R8 are each independently H, halogen, NO2, CN, OR15, NR17R18 or a Ci-C6alkyl, CrC6haioaikyf, C2-C6alkenyl, C2-C6alkynyl, C3-Cacycioalkyl or cycloheteroalkyl group each optionally substituted or when attached to adjacent carbon atoms R7 and R8 may be taken together with the atoms to which they are attached to form an optionally substituted 5- to 7-membered ring optionally containing one or two heteroatoms selected from O, N or S; R9 is H, halogen, NO2, CN, ORi5, NR17R18 or a CrC6aIkyl, CrC6ha!oaikyl, C2-C6alkenyi[ C2-C6alkynyl; C3-C8cycloalkyl, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted;
R10 and R11 are each independently H or a CrC6aIkyl, Ct-C6haloalkyl, C2-C6alkenyl, C2-C6aikynyl, C3-C8cycloalkyl, cycloheteroalkyl or aryl group each optionally substituted;
Ri2, Ri3, Ru and R15 are each independently H or a Ci-C6aikyl, Ci- Cehatoalkyl, C2-C6alkeny!, C2-C6aikynyl, C3-C8cycloalkyl, cycloheteroalkyl, aryl or heteroaryl group each optionally substituted; R17, R18, Rig and R2o are each independently H, CrC4alkyl, C3-
Cgcycioalkyl or R17 and R18 or R19 and R20 may be taken together with the atom to which they are attached to form a 5- to 7- membered ring optionally containing an additional heteroatom selected from O, N or S; and p is 0, 1 or 2; or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1 wherein R1 and R2 are H.
3. The compound according to claim 1 or claim 2 wherein R9 is an optionally substituted heteroaryl group.
4. The compound according to claim 3 wherein R9 is attached to the phenyl ring in the 3-position of the phenyl ring.
5 The compound according to any one of claims 1 to 4 wherein X is N
6 The compound according to any one of claims 1 to 5 wherein R3 and R4 are H
7 The compound according to any one of claims 1 to 6 wherein n is 0 and X and Y are N
8 The compound according to any one of claims 1 to 6 wherein n is 1 , and X is CH or N and Y is CRi0
9 The compound according to ciaim 1 which is one of the following 8-[3-(2-fiuoropyridιn-3-yl)-phenyl]-8-pyridιn-4-yI-2,3,4,8-tetrahydroιmιdazo[1 ,5- a]pyπmιdin-6-amιne,
8-(2,6-diethyipyπdin-4-yl)-8-[3-(2-fluoropyrιdιn-3-yl)-phenyl]-2,3!4,8-tetrahydro- imidazoli 8-(1 -ethyl-1 H-pyrazol-4-yl)-8-[3-(2-fluoropyndιn-3-yf)phenyl]-2,3,4!8-tetrahydro- imιdazo[1 ,5-a]pyπmιdιn-6-amιne, 8-ϊ3-(2-fluoropyπdιn-3-yl)phenyl]-8-[1 -(2,2,2-tπfluoroethyl)-1 H-pyrazol-4-yl]-2,3 ,4,8- tetrahydroιmιdazo[1 ,5-a]pyrιmιdιn-6-amιne, 8-[3-(2-fluoropyrιdin-3-yl)phenyi3-8-[4-(tπf!uoromethoxy)phenyl]-3,4-dihydro-8H- ιmιdazo[5, 1 -c][1 ,2,4]oxadtazm-6-amιne,
8-[3-(5-fluoropyπdιn-3-yl)phenyl]-8-[4-(tπfluoromethoxy)phenyl]-3,4-dιhydro-8H- ιmιdazo[5, 1 -c][1 ,2,4]oxadιazιn-6-amιne, a tautomer thereof, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof
10 A method for the treatment, prevention or amelioration of a disease or disorder characterized by elevated β-amyioid deposits or β-amyloid levels in a patient which comprises providing said patient with a therapeutically effective amount of a compound of formula I as claimed in any one of claims 1 to 9 or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
11. The method according to claim 10 wherein said disease or disorder is selected from Alzheimer's disease; cognitive impairment; Down's Syndrome;
HCHWA-D; cognitive decline; senile dementia; cerebral amyloid angiopathy; and a neurodegenerative disorder.
12. The method according to claim 10 wherein said disease or disorder is characterized by the production of β-amyloid deposits or neurofibrillary tangles.
13. A method for modulating the activity of BACE which comprises contacting a receptor thereof with an effective amount of a compound as claimed in any one of claims 1 to 9.
14. A method for the treatment of Alzheimer's disease in a patient in need thereof which comprises providing to said patient an effective amount of a compound as claimed in any one of claims 1 to 9.
15. A pharmaceutical composition which comprises a pharmaceutically acceptable carrier and a compound of formula I as claimed in any one of claims 1 to 9 or a tautomer thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof.
16. A process for the preparation of a compound of formula I as defined in claim 1 wherein R9 is an optionally substituted aryl or heteroaryl group; which process comprises reacting a compound of formula Il
(H)
wherein Hal is C! or Br and R1, R2, R3, R4, R5, Re, R7, Rs, n, Q, W, X, Y and Z are as defined for formula I hereinabove with a compound of formula A-W wherein A is an optionally substituted aryl or heteroaryl group and W is a leaving group selected from B(OH)2, Sn(n Bu}3 or Sn(CH3)3 in the presence of a palladium catalyst optionally in the presence of a solvent.
EP07840855A 2006-08-17 2007-08-10 Imidazole amines as inhibitors of beta-secretase Withdrawn EP2054414A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US83835706P 2006-08-17 2006-08-17
PCT/US2007/075690 WO2008022024A2 (en) 2006-08-17 2007-08-10 Imidazole amines as inhibitors of beta-secretase

Publications (1)

Publication Number Publication Date
EP2054414A2 true EP2054414A2 (en) 2009-05-06

Family

ID=38988046

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07840855A Withdrawn EP2054414A2 (en) 2006-08-17 2007-08-10 Imidazole amines as inhibitors of beta-secretase

Country Status (9)

Country Link
EP (1) EP2054414A2 (en)
JP (1) JP2010500999A (en)
AR (1) AR062409A1 (en)
CA (1) CA2660441A1 (en)
CL (1) CL2007002288A1 (en)
MX (1) MX2009001699A (en)
PE (1) PE20080744A1 (en)
TW (1) TW200817406A (en)
WO (1) WO2008022024A2 (en)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7700603B2 (en) 2003-12-15 2010-04-20 Schering Corporation Heterocyclic aspartyl protease inhibitors
US7592348B2 (en) 2003-12-15 2009-09-22 Schering Corporation Heterocyclic aspartyl protease inhibitors
US7763609B2 (en) 2003-12-15 2010-07-27 Schering Corporation Heterocyclic aspartyl protease inhibitors
EP1896478B1 (en) 2005-06-14 2014-05-21 Merck Sharp & Dohme Corp. Aspartyl protease inhibitors
AR056865A1 (en) 2005-06-14 2007-10-31 Schering Corp NITROGEN HETEROCICLES AND ITS USE AS INHIBITORS OF PROTEASES, PHARMACEUTICAL COMPOSITIONS
RU2416603C9 (en) 2005-10-25 2012-06-20 Сионоги Энд Ко., Лтд. Aminodihydrothiazine derivatives
AR061264A1 (en) 2006-06-12 2008-08-13 Schering Corp ASPARTILE-PROTEASES INHIBITORS DERIVED FROM PYRIMIDINE, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND USES TO TREAT COGNITIVE OR NEURODEGENERATIVE DISEASES, AND AS HIV VIRUS INHIBITORS.
WO2008133273A1 (en) 2007-04-24 2008-11-06 Shionogi & Co., Ltd. Pharmaceutical composition for treatment of alzheimer's disease
ES2476605T3 (en) 2007-04-24 2014-07-15 Shionogi & Co., Ltd. Aminohydrotiazine derivatives substituted with cyclic groups
JP2011518224A (en) 2008-04-22 2011-06-23 シェーリング コーポレイション Thiophenyl substituted 2-imino-3-methylpyrrolopyrimidinone compounds, compositions, and their use as BACE-1 inhibitors
NZ589590A (en) 2008-06-13 2012-05-25 Shionogi & Co Sulfur-containing heterocyclic derivative having beta-secretase-inhibiting activity
WO2010047372A1 (en) 2008-10-22 2010-04-29 塩野義製薬株式会社 2-aminopyridin-4-one and 2-aminopyridine derivative both having bace1-inhibiting activity
UA108363C2 (en) 2009-10-08 2015-04-27 IMINOTIADIASIADIOXIDE OXIDES AS BACE INHIBITORS, COMPOSITIONS THEREOF AND THEIR APPLICATIONS
RU2012129168A (en) 2009-12-11 2014-01-20 Сионоги Энд Ко. Лтд. OXASINE DERIVATIVES
AU2011321427A1 (en) 2010-10-29 2013-05-02 Shionogi & Co., Ltd. Naphthyridine derivative
EP2634188A4 (en) * 2010-10-29 2014-05-07 Shionogi & Co Fused aminodihydropyrimidine derivative
EP2694521B1 (en) 2011-04-07 2015-11-25 Merck Sharp & Dohme Corp. Pyrrolidine-fused thiadiazine dioxide compounds as bace inhibitors, compositions, and their use
WO2012138734A1 (en) 2011-04-07 2012-10-11 Merck Sharp & Dohme Corp. C5-c6 oxacyclic-fused thiadiazine dioxide compounds as bace inhibitors, compositions, and their use
EP2703399A4 (en) 2011-04-26 2014-10-15 Shionogi & Co Oxazine derivative and bace 1 inhibitor containing same
EP2747769B1 (en) 2011-08-22 2017-08-02 Merck Sharp & Dohme Corp. 2-spiro-substituted iminothiazines and their mono-and dioxides as bace inhibitors, compositions and their use
EP2912035A4 (en) 2012-10-24 2016-06-15 Shionogi & Co Dihydrooxazine or oxazepine derivatives having bace1 inhibitory activity

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1758907B1 (en) * 2004-06-16 2009-02-18 Wyeth Diphenylimidazopyrimidine and -imidazole amines as inhibitors of b-secretase
BRPI0512213A (en) * 2004-06-16 2008-02-19 Wyeth Corp method for treating, preventing or ameliorating a disease or disorder; pharmaceutical composition; process for the preparation of a compound; and use of a compound
JP2008526966A (en) * 2005-01-14 2008-07-24 ワイス Amino-imidazolones for the inhibition of β-secretase
AU2006266167A1 (en) * 2005-06-30 2007-01-11 Wyeth Amino-5-(6-membered)heteroarylimidazolone compounds and the use thereof for beta-secretase modulation
TW200738683A (en) * 2005-06-30 2007-10-16 Wyeth Corp Amino-5-(5-membered)heteroarylimidazolone compounds and the use thereof for β-secretase modulation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2008022024A2 *

Also Published As

Publication number Publication date
JP2010500999A (en) 2010-01-14
WO2008022024A3 (en) 2008-05-22
CL2007002288A1 (en) 2008-02-08
WO2008022024A2 (en) 2008-02-21
PE20080744A1 (en) 2008-05-24
MX2009001699A (en) 2009-02-25
CA2660441A1 (en) 2008-02-21
TW200817406A (en) 2008-04-16
AR062409A1 (en) 2008-11-05

Similar Documents

Publication Publication Date Title
WO2008022024A2 (en) Imidazole amines as inhibitors of beta-secretase
US7700606B2 (en) Imidazole amines as inhibitors of β-secretase
MXPA06014792A (en) Diphenylimidazopyrimidine and -imidazole amines as inhibitors of b-secretase.
US7563796B2 (en) Diphenylimidazopyrimidines as inhibitors of β-secretase
AU2003214414B2 (en) Azaindoles as inhibitors of c-Jun N-terminal kinases
CA2629512C (en) Bicyclic cinnamide compound and prophylactic or therapeutic uses thereof
US20070203116A1 (en) Dihydrospiro[dibenzo[a,d][7]annulene-5,4&#39;-imidazol] compounds for the inhibition of beta-secretase
AU2017382029A1 (en) Bicyclic dihydropyrimidine-carboxamide derivatives as Rho-kinase inhibitors
KR20090130319A (en) Spiroindolinone derivatives
JP2019520396A (en) Spiro fused ring ureas as ROCK inhibitors
CA2566094A1 (en) Cinnamide compound
CA2116068A1 (en) Cyclic derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
CA2724294A1 (en) Glucagon receptor antagonists, compositions, and methods for their use
KR101710197B1 (en) Substituted n-phenyl-1-(4-pyridinyl)-1h-pyrazol-3-amines
CA2870062A1 (en) Pyrrolopyrazone inhibitors of tankyrase
US6710064B2 (en) Hydantoin compounds useful as anti-inflammatory agents
WO2015066697A1 (en) Fused morpholinopyrimidines and methods of use thereof
WO2015066696A1 (en) Fused morphlinopyrimidines and methods of use thereof
CA2436943A1 (en) Hydantoin compounds useful as anti-inflammatory agents
KR20070030938A (en) A method for preparing pyrrolotriazine compounds

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090211

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: MALAMAS, MICHAEL, SOTIRIOS

Inventor name: BARNES, KEITH, DOUGLAS

Inventor name: JOHNSON, MATTHEW, ROBERT

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20100315

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: WYETH LLC

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120301