WO2004054988A1 - Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses - Google Patents

Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses Download PDF

Info

Publication number
WO2004054988A1
WO2004054988A1 PCT/CA2003/001962 CA0301962W WO2004054988A1 WO 2004054988 A1 WO2004054988 A1 WO 2004054988A1 CA 0301962 W CA0301962 W CA 0301962W WO 2004054988 A1 WO2004054988 A1 WO 2004054988A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
aralkyl
aryl
alkenyl
Prior art date
Application number
PCT/CA2003/001962
Other languages
French (fr)
Inventor
Bruce P. Connop
David D. MACDONALD
Parimal Nathwani
Michael G. Organ
Original Assignee
Active Pass Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Active Pass Pharmaceuticals, Inc. filed Critical Active Pass Pharmaceuticals, Inc.
Priority to AU2003287840A priority Critical patent/AU2003287840A1/en
Publication of WO2004054988A1 publication Critical patent/WO2004054988A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/56One oxygen atom and one sulfur atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/52Two oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/60Three or more oxygen or sulfur atoms

Definitions

  • the invention relates to compounds, compositions and methods for regulating the production and/or release of ⁇ -amyloid in cells, and provides for alleviation and prevention of ⁇ -amyloid production, release and/or plaque development such as occurs in, e.g., Alzheimer's disease.
  • Alzheimer's disease is a common brain disorder of the elderly and is associated with progressive dementia.
  • the key features of the disease include progressive memory impairment, loss of language and visuospatial skills, and behavior deficits. These changes in cognitive function are the result of degeneration of neurons in the cerebral cortex, hippocampus, basal forebrain, and other regions of the brain.
  • Neuropathological analyses of postmortem Alzheimer's diseased brains consistently reveal the presence of large numbers of neurofibrillary tangles in degenerated neurons and neuritic plaques in the extracellular space and walls of the cerebral microvasculature.
  • the neurofibrillary tangles are composed of bundles of paired helical filaments containing hyperphosphorylated tau protein (Lee, V. M and Trojanowski, J.
  • the neuritic plaques consist of deposits of proteinaceous material surrounding a ⁇ -amyloid core (Selkoe, D. J., Annu. Rev. Neurosci. 77:489-517, 1994).
  • a ⁇ ⁇ -amyloid peptide
  • a ⁇ -amyloid precursor protein APP
  • Alternative splicing generates several different isoforms of APP; in neurons, the predominant isoform is 695 amino acids in length (APP695).
  • APP traverses the endoplasmic reticulum (ER) and trans- Golgi network (TGN), it becomes N- and O-glycosylated and tyrosine-sulfated.
  • Mature holoprotein can be catabolized in several compartments to produce both non- and amyloidogenic APP fragments.
  • APP is expressed and constitutively catabolized in most cells.
  • APP The dominant catabolic pathway appears to be cleavage of APP within the A ⁇ sequence by an enzyme provisionally termed ⁇ -secretase, leading to release of a soluble ectodomain fragment known as APPs ⁇ .
  • APP can also be cleaved by enzymes known as ⁇ - and ⁇ -secretase at the N- and C-termini of the A ⁇ , respectively, followed by release of A ⁇ into the extracellular space.
  • BACE1 ⁇ -site APP cleaving enzyme
  • BACE2 ⁇ -site APP cleaving enzyme
  • a ⁇ 40 is the predominant form of A ⁇ produced by sequential cleavage of APP
  • 5-7% of total A ⁇ exists as A ⁇ 42 (Cappai et al., Int. J. Biochem. Cell Biol.
  • a ⁇ peptide appears to dramatically alter its biochemical and biophysical properties. Specifically, the additional two amino acids at the C-terminus of A ⁇ 42 are hydrophobic, presumably increasing the propensity of A ⁇ 42 to aggregate. For example, Jarrett et al. demonstrated that A ⁇ 42 aggregates very rapidly in vitro compared to A ⁇ 40, suggesting that the longer forms of A ⁇ may be the important pathological proteins that are involved in the initial seeding of the neuritic plaques in AD (Jarrett et al., Biochemistry 32:4693-4697, 1993; Jarrett et al., Ann. NY Acad. Sci. 695: 144-148, 1993).
  • a ⁇ A ⁇ 42
  • a ⁇ 42 A ⁇ -amyloid precursor protein
  • FAD familial forms of AD
  • APP ⁇ -amyloid precursor protein
  • This aggressive form of Alzheimer's disease has been shown to be caused by missense mutations in (at least) three genes: the ⁇ -amyloid precursor protein (APP) gene itself (Goate, A. et al., Nature 349:704-706, 1991; Mullan, M. et al., Nature Genet. 1345-347, 1992), and two genes termed presenilins 1 and 2 (Sherrington, R. et al., Nature 375:754-760, 1995; Rogaev, E. I. et al., Nature 376:775-778, 1995).
  • APP ⁇ -amyloid precursor protein
  • Presenilin-1 Presenilin-1
  • Presenilin-2 Presenilin-2
  • amyloid cascade hypothesis originally suggested that increased accumulation and deposition of the A ⁇ peptide (primarily A ⁇ 42) leads to senile plaque formation and subsequent neuronal death and dementia (Hardy et al., Science 256: 184-185, 1992).
  • this hypothesis does not explain the lack of correlation observed between the degree of dementia and the absolute number of ⁇ -amyloid deposits in the brain (Dickson et al., Neurobiol Aging 10: 402-404, 1989).
  • Wang et al. (1999) have suggested that the degree of dementia in AD patients shows a stronger correlation with the levels of soluble A ⁇ as opposed to the number of histologically determined plaques (Wang et al., Exp. Neurol. 158: 328, 1999).
  • ADDL Alzheimer derived diffusible ligands
  • sulindac sulphide and indomethacin reduced A ⁇ 42 by 80% at concentrations of 100 (Jvl without altering A ⁇ 40 in Chinese hamster ovarian cells (CHO) over-expressing APP (Weggen et al., Nature 414: 212-216, 2001).
  • Morihara et al. (2002) demonstrated that specific stereoisomers of NSAIDs, namely R-ibuprofen and R-flurbiprofen, selectively decreased A ⁇ 42 release from HEK- 293 cells over-expressing the 'Swedish mutant' form of APP.
  • NSAIDs appear to selectively decrease levels of A ⁇ 42, the concentrations employed to achieve this effect appear to be much higher than those used to inhibit COX activity in clinically relevant situations.
  • NSAIDs have IC 50 values in the nanomolar to low micromolar ranges for COX enzymes (Neupert et al., Br. J. Pharmacol. 122: 487-492, 1997), while their A ⁇ 42 lowering effects are seen at much higher concentrations (e., 100-500 ⁇ M).
  • toxicity associated with high concentrations of NSAIDs may restrain them from being used widely as AD therapeutics (Morihara et al., J. Neurochem 83: 1009-1012, 2002).
  • This invention is directed to certain pyrimidine compounds, pharmaceutical compositions containing certain pyrimidine compounds, and the use of certain pyrimidine compounds for regulating the production and/or release of ⁇ -amyloid in cells, and for alleviation and prevention of ⁇ -amyloid production, release and/or plaque development.
  • the present invention provides pyrimidine compounds of formula (1):
  • Y is selected from the group consisting of -S(O) m - and -O-;
  • W is selected from the group consisting of -OR 4 , -N(R 5 ) 2 and -NHN(R 5 ) 2 ; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R 1 and R 2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6
  • each R 3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(
  • R 4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
  • R 5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
  • R 6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl;
  • R 7 is selected from the group consisting of hydrogen, alkyl and aralkyl.
  • the invention includes these compounds as single compounds or mixtures of compounds; in pure or impure form; when stereoisomers are possible then the invention provides the compound as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; and as pharmaceutically acceptable salts.
  • the present invention also provides compositions that include at least one of these compounds.
  • the present invention provides a pharmaceutical composition comprising a pyrimidine compound of the invention.
  • the present invention provides a composition comprising a pyrimidine compound of the invention and a pharmaceutically acceptable carrier, excipient or diluent.
  • the invention provides a method for modulating the production and/or release of ⁇ -amyloid from a cell, comprising treating the cell with a compound of formula (1) or a composition comprising a compound of formula (1).
  • the invention also provides a method of treatment comprising modulating the production and/or release of ⁇ -amyloid in a non-human mammal in need of said treatment, said method comprising administering to said non-human mammal a compound that can modulate the production and/or release of ⁇ -amyloid in a human, or a composition comprising such a compound.
  • the invention also provides a method of treatment comprising modulating the production and/or release of ⁇ -amyloid in a human in need of said treatment, said method comprising administering to said human a compound that can modulate the production and/or release of ⁇ -amyloid in a human, or a composition comprising such a compound.
  • the invention yet further provides a method for preferentially reducing production and/or release of A ⁇ 42 relative to one or more other forms of A ⁇ , in a target that produces and/or releases A ⁇ 42, for instance a target selected from a cell, a human, a non-human mammal, and the brain of a human, comprising administering to the target a compound or pharmaceutical composition comprising a chemical agent as described herein.
  • This method may be used to treat, e.g., a human, wherein said human, e.g., is afflicted with Alzheimer's disease.
  • said human being treated has a genetic predisposition or environment exposure that increases the likelihood that said person will develop Alzheimer's disease.
  • said human has suffered a head injury and is treated with a compound or composition as described herein.
  • said human exhibits minimal cognitive impairment suggestive of early stage Alzheimer's disease.
  • said human has suffered a head injury and is treated with a compound or composition as described herein.
  • the invention also provides a method for delivering to the brain a compound capable of modulating A ⁇ production and/or release. This delivery system achieves specific delivery of such compounds through conjugating the compounds with a polar lipid or other carrier, achieving effective intracerebral concentration of such compounds efficiently and with specificity.
  • Figure 1 is a line graph showing the effect of Compound 1 on secreted A ⁇ 40 and A ⁇ 42 from SM-4 cells.
  • Cells were treated with vehicle (0.01 % w/v DMSO) or 50-100 ⁇ M Compound 1 for 5 hrs (A) or 50-300 ⁇ M of Compound 1 for 16 hrs (B).
  • vehicle 0.01 % w/v DMSO
  • the culture media was harvested and assayed for A ⁇ 40 and A ⁇ 42 by ELISA.
  • Secreted A ⁇ was standardized to propridium iodide fluorescence as a measure of total cell number.
  • FIG. 1 is a line graph showing the effect of Compound 2 on secreted A ⁇ 40 and A ⁇ 42 in SM-4 cells.
  • a compound refers to one or more of such compounds
  • the enzyme includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
  • Alkyl refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, ⁇ -propyl, 1-methylethyl (iso-propyl), ⁇ -butyl, n-pentyl, 1 ,1-dimethylethyl (f-butyl), and the like.
  • Alkylene chain refers to a straight or branched divalent hydrocarbon chain consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twenty carbons atoms, preferably having from one to eight carbons, e.g., methylene, ethylene, propylene, n-butylene, and the like.
  • Alkenyl refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1 ,4-dienyl, and the like.
  • Alkynyl refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, prop-1-ynyl, pent-1-ynyl, penta-1 ,4-diynyl, and the like.
  • Alkoxy refers to a radical of the formula -OR a where R a is an alkyl radical as defined above, e.g., methoxy, ethoxy, n-propoxy, 1-methylethoxy (iso-propoxy), n-butoxy, n-pentoxy, 1 ,1-dimethylethoxy (t-butoxy), and the like.
  • Aryl refers to an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi- electron system. Examples, without limitation, of aryl groups are phenyl, naphthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted.
  • the substituted group(s) is one or more selected from alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 , -N(R 6 )C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -OC(S)NR 6 , -NR 6 C(S)OR 7 , -NR 6 S(O) t R 6 (where t is t is
  • aryl or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals optionally substituted by one or more substituents selected from the group consisting of alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 , -N(R 6 )C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -S(O) t
  • Aralkyl refers to a radical of the formula -R a R b where R a is an alkyl radical as defined above and R b is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl, and the like.
  • the aryl radical may be optionally substituted as described above.
  • alkenyl refers to a radical of the formula -R e -R b where Rb is an aryl radical as defined above and R e is an alkenyl radical as defined above, e.g., 2- phenylethenyl, and the like.
  • Carboxy refers to the -C(O)OH radical.
  • Cycloalkyl refers to a stable monovalent monocyclic or bicyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having from three to ten carbon atoms, and which is saturated and attached to the rest of the molecule by a single bond, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decalinyl and the like.
  • cycloalkyl is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, halo, haloalkyl, haloalkoxy, hydroxy, amino, and carboxy.
  • Halo refers to bromo, chloro, iodo or fluoro.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl ⁇ 2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like.
  • Haloalkoxy refers to a radical of the formula -OR c where R c is an haloalkyl radical as defined above, e.g., trifluoromethoxy, difluoromethoxy, trichloromethoxy, 2,2,2-trifluoroethoxy, 1 -fluoromethyl-2-fluoroethoxy , 3-bromo-2-fluoropropoxy, 1-bromomethyl-2-bromoethoxy, and the like.
  • Heteroalkyl refers to an alkyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 , -N(R 6 )C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -OC(S)NR 6 , -NR 6 C(S)OR 7 , -NR 6 S(O) t R 6 (where t is 0 to 2), with R 6 and R 7 as defined above and the substitution
  • Heteroalkenyl refers to an alkenyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 ,
  • Heteroalkynyl refers to an alkynyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 , -N(R 6 )C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -OC(S)NR 6 , -NR 6 C(S)OR 7 , -NR 6 S(O) t R 6 (where t is 0 to 2), with R 6 and R 7 as defined
  • Heterocyclyl refers to a stable 3- to 15-membered ring radical which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quatemized; and the heterocyclyl radical may be aromatic or partially or fully saturated.
  • the heterocyclyl radical may not be attached to the rest of the molecule at any heteroatom atom.
  • heterocyclyl radicals include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzothiadiazolyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, carbazolyl, cinnolinyl, decahydroisoquinolyl, dioxolanyl, furanyl, furanonyl, isothiazolyl, imidazolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, indolizinyl, isoxazolyl,
  • heterocyclyl is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6 ) 2 , -N(R 6 )C(O)R 6 , -N(R 6 )C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -OC(S
  • “/V-heterocyclyl” refers to a heterocyclyl radical as defined above wherein the one to five heteroatoms contained therein are selected only from nitrogen, e.g., pyridinyl, tetrazolyl, pyrazolyl, isoquinolinyl, quinolinyl, and phthalazinyl and the like.
  • “Heterocyclylalkyi” refers to a radical of the formula -R a Rd where R a is an alkyl radical as defined above and R is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom.
  • heterocyclylcarbonyl refers to a radical of the formula -C(O)-Rd where R d is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the carbonyl at the nitrogen atom.
  • Hydrocarbon refers to a compound formed entirely of carbon and hydrogen (including isotopes thereof), while “hydrocarbyl” refers to a hydrocarbon radical.
  • compounds which are "commercially available” may be obtained from standard commercial sources including Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc.
  • suitable conditions for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry.
  • Prodrugs is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention.
  • prodrug refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam).
  • prodrugs as Novel Delivery Systems
  • ACS. Symposium Series Vol. 14
  • Bioreversible Carriers in Drug Design ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • prodrug is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention.
  • Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention and the like.
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Mammal” includes humans and domesticated animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like. "Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • “optionally substituted aryl” means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable salt” and “salts thereof in the compounds of the present invention refers to pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like
  • organic acids such as acetic acid, triflu
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, ⁇ /-ethylpiperidine, polyamine resins and the like.
  • basic ion exchange resins such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanol
  • organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • “Pharmaceutically acceptable excipient” as used herein is intended to include without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, emulsifier, or stabilizer which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • Treating covers the treatment of a disorder in a mammal, preferably a human, which disorder is characterized by the accumulation or deposition of ⁇ -amyloid peptide, and includes: (i) preventing the disorder from occurring in a mammal, in particular a human, when such mammal is predisposed to the disorder but has not yet been diagnosed as having it;
  • Y is selected from the group consisting of -S(O) m - and -O-;
  • W is selected from the group consisting of -OR 4 , -N(R 5 ) 2 and -NHN(R ⁇ ) 2 ; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R and R 2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -OC(O)R 7 , -C(O)N(R 6 ) 2 , -C(S)R 6 , -C(O)R 6 , -N(R 6
  • R 4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
  • R 5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
  • R 6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl;
  • R 7 is selected from the group consisting of hydrogen, alkyl and aralkyl.
  • the compound(s) may be, for example, a single stereoisomer, a mixture of stereoisomers, a racemic mixture of stereoisomers; in solvated form, as a polymorph; or as a pharmaceutically acceptable salt thereof.
  • the invention provides prodrug forms of compounds of formula (1 ).
  • one or more compounds that would otherwise be included within the scope of formula (1) are explicitly excluded.
  • the following are specific exclusions that may be applied to describe compounds of formula (1), where any two or more of the exclusions may be combined so as to create a larger set of excluded compounds.
  • the compounds of the invention exclude propanoic acid, 2- [[4-[2-chloro-5-[3,6-dihydro-3-methyl-2,6-dioxo-4-(trifluoromethyl)-1(2H)-pyrimidinyl]-4- fluorophenoxy]-2-pyrimidinyl]oxy]-, methyl ester, (having CAS Registry No. (RN) 353292-13-4).
  • the compounds of the invention additionally, or alternatively, exclude: acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2- pyrimidinyljthio]- (RN 91759-31-8); and acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)- 2-pyrimidinyl]thio]-, ethyl ester (RN 91759-33-0).
  • the compounds of the invention additionally, or alternatively, exclude any one or more of the following compounds: propanoic acid, 2- [[4-[(4-bromophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-64-6); propanoic acid, 2-[[4-[(4-chlorophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-63-5); acetic acid, [[5-methoxy-4-[[3-(trifluoromethyl)phenyl]thio]-2- pyrimidinyljthio]-, ethyl ester (RN 338423-49-7); acetic acid, [[4-[(2-chlorophenyl)thio]-5- methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423
  • excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring (where the Y group is bonded to the 1 position of the carbocyclic aromatic ring).
  • excluded are compounds having 2,3-(C C 3 alkyl) (i.e., alkyl groups having 1 , 2 or 3 carbons) substitution on the carbocyclic aromatic ring.
  • excluded are compounds having excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring when Y is oxygen.
  • provisos may be added to a claim or other statement of the invention so as to preclude (exclude) certain chemical groups from being present at positions of the compounds (where distinct provisos are separated by semicolons, and any plurality of provisos may be combined to create a larger proviso): Y is not -S-; Y is not -S(O)-; Y is not -S(O) 2 -;Y is not -S(O) m -; Y is not -O-; W is not -OR 4 , W is not -N(R 5 ) 2 ; W is not -NHN(R 5 ) 2 ; p is not 0; p is not 1 ; p is not 2; p is not 3; p is not 4; p is not 5; q is not 0; q is not 1 ; q is not 2; m is not 0; m is not 1 ;
  • R 1 is not -N(R 6 )C(O)R 6 ; R 1 is not -N(R 6 )C(O)OR 7 ; R 1 is not -S(O) t R 6 (where t is not 0 to 2); R 1 is not -S(O) t N(R 6 ) 2 (where t is not 0 to 2); R 1 is not -OC(S)NR 6 ; R 1 is not -NR 6 C(S)OR 7 ; R 1 is not -NR 6 S(O) t R 6 (where t is not 0 to 2); R 1 is not heterocyclyl; R 1 is not heterocyclylalkyi; R 2 is not alkyl; R 2 is not alkenyl; R 2 is not alkynyl; R 2 is not heteroalkyl; R 2 is not heteroalkenyl; R 2 is not heteroalkynyl; R 2 is not aryl; R 2 is not aralkyl; R 2 is not a
  • the compounds may be specified by requiring one or more of the following conditions: Y is -S-; Y is -S(O)-; Y is -S(O) 2 -;Y is -S(O) m -; Y is -O-; W is -OR 4 , W is -N(R 5 ) 2 ; W is -NHN(R 5 ) 2 ; p is 0; p is 1 ; p is 2; p is 3; p is 4; p is 5; q is 0; q is 1 ; q is 2; m is 0; m is 1 ; m is 2; n is 0; n is 1 ; n is 2; n is 3; n is 4; n is 5; R 1 is alkyl; R 1 is alkenyl; R 1 is alkynyl;
  • R 1 is heterocyclyl; R 1 is heterocyclylalkyi; R 2 is alkyl; R 2 is alkenyl; R 2 is alkynyl; R 2 is heteroalkyl; R 2 is heteroalkenyl; R 2 is heteroalkynyl; R 2 is aryl; R 2 is aralkyl; R 2 is aralkenyl; R 2 is cycloalkyl; R 2 is cycloalkylalkyl; R 2 is halo; R 2 is haloalkyl; R 2 is haloalkoxy; R 2 is nitro; R 2 is cyano; R 2 is -NHOH; R 2 is -OR 7 ; R 2 is -SR 7 ; R 2 is -C(O)OR 7 ; R 2 is -OC(O)R 7 ; R 2 is -C(O)N(R 6 ) 2 ; R 2 is cyano; R 2 is -NHOH; R 2 is -OR 7 ; R
  • the compound is in the form of a salt, particularly a pharmaceutically acceptable salt.
  • Y is -S-; Y is -S(O)-; Y is - S(O) 2 -;Y is -S(O) m -; Y is -O-; W is -OR 4 , W is -N(R 5 ) 2 ; W is -NHN(R 5 ) 2 ; p is 0; p is 1 ; p is 2; p is 3; p is 4; p is 5; q is 0; q is 1; q is 2; m is 0; m is 1; m is 2; n is 0; n is 1 ; n is 2; n is 3; n is 4; n is 5; R 1 is alkyl; R 1 is alkenyl; R 1 is alkynyl; R 1 is heteroalkyl; R 1 is heteroalkenyl; R 1 is heteroalkynyl;
  • R 1 ) p - and (R 2 ) q - are utilized herein to indicate that a number "p" of R groups are bonded to the carbocyclic aromatic ring of the compound, and a number "q" of R 2 groups are bonded to the heterocydic aromatic ring of the compound.
  • p is zero, then there are no R 1 groups present on the compound, and the carbocyclic aromatic ring is unsubstituted phenyl.
  • q is zero, then there are no R 2 groups present on the compound.
  • the compounds of formula (1) are described, in part, by the presence of various groups, e.g., Y, W, R 1 , R 2 , R 3 , etc., and various integers, e.g., m, n, p, q, etc.
  • the term "independently at each occurrence" in connection with a description of the compound and the various groups and integers thereof is intended to indicate that the selection of the identity for a particular group or integer is independent of the selection of the identity of any other group or integer.
  • the selection of any one group at one instance is independent of the selection of the same group at another instance (which will arise when a group, e.g., R 1 , appears more than once in the compound).
  • the selection of any one integer (e.g., t) at one occurrence in the compound is entirely independent of the selection of the same integer if and when it occurs an additional time in the compound.
  • the compound of formula (1) is characterized by the requirements that (where distinct requirements are separated by semicolons, and any two or more requirements may be combined): Y is S; Y is O; W is -OR 4 ; W is -OH; W is -N(R 5 ) 2 ; R 3 is -H; m is 0; n is 1 ; Y is S and W is -OR 4 ; an "R" group (e.g., R 1 , R 2 , R 3 , etc.) is a hydrocarbon, i.e., formed entirely of carbon and hydrogen; R 1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl; an "R” group (or any one or more members that comprise the R group) has a specified maximum number of carbons, where in various aspects of the invention this maximum number is 20,
  • R 2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR 7 , -SR 7 , -C(O)OR 7 , -S(O) t R 6 (where t is 0 to 2), -S(O) t N(R 6 ) 2 (where t is 0 to 2), -OC(S)NR 6 , -NR 6 C(S)OR 7 , -NR 6 S(O) t R 6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi, and R 2 has a formula weight of less than 500 and
  • composition of the present invention may be present in a composition of the present invention as described below, and may be used in any of the methods of the present invention as described below.
  • the compound of formula (1) may be described in terms of any one or more the express requirements and/or express limitations set forth herein. Likewise with descriptions of methods of the present invention.
  • Suitable protecting groups include hydroxy, amino, mercapto and carboxylic acid.
  • Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., f-butyldimethylsilyl, f-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for mercapto include -C(O)-R (where R is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or aralkyl esters.
  • Protecting groups may be added or removed in accordance with standard techniques, which are well-known to those skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and
  • the protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl chloride resin.
  • starting components may be obtained from sources such as Aldrich, or synthesized according to sources known to those of ordinary skill in the art (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th Ed., 2000 (Wiley Interscience, New York)).
  • the various R groups (e.g., R-i, R 2 , and R 3 , etc.) of the compounds of formula (1) are selected from components as indicated in the description of formula (1), and may be attached to starting components, intermediate components, and/or final products according to schemes known to those of ordinary skill in the art.
  • R 1 , R 2 , and R 3 are defined as above.
  • X is Cl or Br.
  • R' is an alkyl or an aryl group.
  • starting material of formula (a) reacts with a halogenated compound of formula (b) where X represents a leaving group, in the presence of a base such as NaHCO 3 at room temperature to afford the compound of formula (c).
  • Componds of formulae (a) and (b) are very well known in the chemical literature.
  • the conversion of the hydroxyl group to a Cl or Br group is accomplished by treating the compound of formula (c) with an agent such as POCI 3 , POBr 3 , PCI 5 or PBr 5 under reflux to afford the halogenated compound of formula (d).
  • the compound of formula (d) then reacts with a compound of formula (e) such as phenol or benzenethiol which is treated with a base such as NaOH to afford the compound of the invention having formula (f).
  • compounds of formula (e) are very well known in the chemical literature.
  • the compound of formula (f) can be oxidized by an oxidizing agent such as H 2 O 2 to afford a compound of the invention having formula (g).
  • an oxidizing agent such as H 2 O 2
  • the transformation of the compound of formula (g) to a compound of formula (1) can readily be achieved by transesterification, saponification and hydrolysis as well as by amidation of the free carboxyl group or the corresponding acid halide.
  • the halogenated starting material of formula (a') is condensed with a compound of formula (b') such as phenol and benzenethiol in an appropriate solvent under reflux.
  • a compound of formula (b') such as phenol and benzenethiol
  • This reaction yields the compound of formula (c') which is then transformed to a compound of formula (e') by reacting with a compound of formula (d') in the presence of a base such as Na 2 CO 3 under reflux.
  • a base such as Na 2 CO 3 under reflux.
  • Compounds of formula (d') are well known in the chemical literature.
  • the product is isolated and filtered, and then oxidized by an oxidizing agent such as H 2 O 2 to afford the compound of formula (g').
  • P indicates a protection group such as the BOC group or the like.
  • the method of Scheme 3 is particularly suited to prepare compounds wherein R 1 is an electron-withdrawing group, such as -NO 2 , -CF 3 and the like.
  • the amino group of a compound of formula (a") is protected by a protection group such as BOC or the like according to procedures known to one skilled in the art to yield the N-protected product (b").
  • a protection group such as BOC or the like
  • Compounds of formula (a") are well known in the chemical literature.
  • This N-protected compound is then reacted with a halogenated compound of formula (c") at room temperature in the presence of a base, such as NaHCO 3 or the like to afford the compound of formula (d”).
  • a base such as NaHCO 3 or the like
  • compounds of formula (c") are well known in the chemical literature.
  • Treatment of the compound of formula (d") with weak acid such as trifluoroacetic acid can be used to remove the protection group and thereby obtain the amino product of formula (e").
  • Compound of formula (e") is then converted into the compound of formula (f) by diazotizing the amino group with a reagent such as NaNO 2 and then treating with xanthate or water to afford the corresponding thiol or the hydroxy product of formula (f") which is subsequently treated with a compound of formula (g") in the presence of pyridine or the like under reflux to afford the compound of formula (h").
  • a compound of formula (h") can then be oxidized by an oxidizing agent such as H 2 O 2 or the like to yield the product of formula (i").
  • the transformation of the compound of formula (i") to a compound of formula (1) can readily be achieved by transesterification, saponification and hydrolysis as well as by amidation of the free carboxyl group or the corresponding acid halide.
  • a brain cell is defined herein as any cell residing within the skull bone of the head including the spinal cord.
  • Non-limiting examples of brain cells are neurons, glial cells (astrocytes, oligodendrocytes, microglia), cerebrovascular cells (muscle cells, endothelial cells), blood cells (red, white, platelets, etc.) and cells that comprise the meninges.
  • BBB blood brain barrier
  • Circulating molecules are normally able to gain access to brain cells via one of two processes: (i) lipid-mediated transport of small molecules through the BBB by free diffusion, or (ii) catalyzed transport.
  • compounds that are useful for inhibiting A ⁇ production and/or release are preferably linked to agents that will facilitate penetration of the blood brain barrier.
  • the method of the present invention will employ a naturally occurring polyamine linked to a small molecule useful at inhibiting A ⁇ production and/or release.
  • Natural cell metabolites that may be used as linkers include, but are not limited to, putrescine (PUT), spermidine (SPD), spermine (SPM), or DHA.
  • PUT putrescine
  • SPD spermidine
  • SPM spermine
  • DHA DHA
  • An alternative method to deliver a compound across the BBB is by intracerebroventricular pump.
  • the neurologic agent may also be delivered to the nasal cavity. It is preferred that the agent be delivered to the olfactory area in the upper third of the nasal cavity and particularly to the olfactory epithelium in order to promote transport of the agent into the peripheral olfactory neurons rather that the capillaries within the respiratory epithelium.
  • the transport of neurologic agents to the brain is accomplished by means of the nervous system instead of the circulatory system so that small molecules which inhibit A ⁇ production and/or release may be delivered to the appropriate areas of the brain.
  • the neurologic agent be capable of at least partially dissolving in the fluids that are secreted by the mucous membrane that surround the cilia of the olfactory receptor cells of the olfactory epithelium in order to be absorbed into the olfactory neurons.
  • the agent may be combined with a carrier and/or other substances that foster dissolution of the agent within nasal releases.
  • Potential adjuvants include GM-1 , phosphatidylserine (PS), and emulsifiers such as polysorbate 80.
  • the method of the present invention may combine the agent with substances that enhance the absorption of the agent through the olfactory epithelium. It is preferred that the additives promote the absorption of the agent into the peripheral olfactory receptor cells. Because of their role in odor detection, these peripheral neurons provide a direct connection between the brain and the outside environment.
  • the olfactory receptor cells are bipolar neurons with swellings covered by hair-like cilia which project into the nasal cavity. At the other end, axons from these cells collect into aggregates and enter the cranial cavity at the roof of the nose.
  • the neurologic agent is lipophilic in order to promote absorption into the olfactory neurons and through the olfactory epithelium.
  • neurologic agents that are lipophilic are gangliosides and phosphatidylserine (PS).
  • the neurologic agent may be combined with a carrier and/or other substances that enhance the absorption of the agent into the olfactory neurons.
  • the neurologic agent may be combined with micelles comprised of lipophilic substances. Such micelles may modify the permeability of the nasal membrane and enhance absorption of the agent.
  • the lipophilic micelles that are preferred are gangliosides, particularly GM-1 ganglioside, and phosphatidylserine (PS).
  • the neurologic agent may be combined with one or several types of micelle substances.
  • the invention further provides for transport of the neurologic agent along the olfactory neural pathway.
  • the agent may be combined with substances that possess neurotrophic or neuritogenic properties which, in turn, may assist in transporting the agent to sites of nerve cell damage.
  • Prophylactic therapies may apply the agent alone or in combination with a carrier, other agents, and/or other substances that may enhance the absorption of the agent into the olfactory neurons.
  • the agent alone or in combination with other substances as a pharmaceutical composition may be administered to the olfactory area located in the upper third of the nasal cavity.
  • the composition may be dispensed intranasally as a powdered or liquid nasal spray, nose drops, a gel or ointment, through a tube or catheter, by syringe, by packtail, by pledget, or by submucosal infusion.
  • Other modifications of the compounds described herein in order to enhance penetration of the blood brain barrier can be accomplished using methods and derivatives known in the art, including but not limited to those disclosed in the following patent publications, each of which is incorporated by reference herein:
  • U.S. Pat. No. 5,023,252 issued Jun. 11 , 1991 to Hseih discloses the use of pharmaceutical compositions comprising a neurologically active drug and a compound for facilitating transport of the drug across the blood-brain barrier including a macrocyclic ester, diester, amide, diamide, amidine, diamidine, thioester, dithioester, thioamide, ketone or lactone.
  • U.S. Pat. No. 5,039,794, issued Aug. 13, 1991 to Wier et al. discloses the use of a metastatic tumor-derived egress factor for facilitating the transport of compounds across the blood-brain barrier.
  • U.S. Pat. No. 5,124,146 issued Jun. 23, 1992 to Neuwelt discloses a method for delivery of therapeutic agents across the blood-brain barrier at sites of increased permeability associated with brain lesions.
  • U.S. Pat. No. 5,153,179, issued Oct. 6, 1992 to Eibl discloses acylated glycerol and derivatives for use in a medicament for improved penetration of cell membranes.
  • U.S. Pat. No. 5,177,064, issued Jan. 5, 1993 to Bodor discloses the use of lipoidal phosphonate derivatives of nudeoside antiviral agents for delivery across the blood-brain barrier.
  • U.S. Pat. No. 5,258,402 issued Nov. 2, 1993 to Maryanoff discloses treatment of epilepsy with imidate derivatives of anticonvulsive sulfamate.
  • U.S. Pat. No. 5,270,312, issued Dec. 14, 1993 to Glase et al. discloses substituted piperazines as central nervous system agents.
  • U.S. Pat. No. 5,284,876, issued Feb. 8, 1994 to Shashoua et al. discloses fatty acid conjugates of dopamine drugs.
  • U.S. Pat. No. 5,389,623, issued Feb. 14, 1995 to Bodor discloses the use of lipoidal dihydropyridine derivatives of anti-inflammatory steroids or steroid sex hormones for delivery across the blood-brain barrier.
  • U.S. Pat. No. 5,405,834, issued Apr. 11 , 1995 to Bundgaard et al. discloses prodrug derivatives of thyrotropin releasing hormone.
  • U.S. Pat. No. 5,413,996, issued May 9, 1995 to Bodor discloses acyloxyalkyl phosphonate conjugates of neurologically-active drugs for anionic sequestration of such drugs in brain tissue.
  • U.S. Pat. No. 5,434,137, issued Jul. 18, 1995 to Black discloses methods for the selective opening of abnormal brain tissue capillaries using bradykinin infused into the carotid artery.
  • U.S. Pat. No. 5,442,043, issued Aug. 15, 1995 to Fukuta et al. discloses a peptide conjugate between a peptide having a biological activity and incapable of crossing the blood-brain barrier and a peptide which exhibits no biological activity and is capable of passing the blood-brain barrier by receptor-mediated endocytosis.
  • U.S. Pat. No. 5,466,683, issued Nov. 14, 1995 to Sterling et al. discloses water soluble analogues of an anticonvulsant for the treatment of epilepsy.
  • U.S. Pat. No. 5,525,727, issued Jun. 11, 1996 to Bodor discloses compositions for differential uptake and retention in brain tissue comprising a conjugate of a narcotic analgesic and agonists and antagonists thereof with a lipoidal form of dihydropyridine that forms a redox salt upon uptake across the blood-brain barrier that prevents partitioning back to the systemic circulation.
  • International Pat. Application Publication Number WO85/02342 published
  • a carboxylic acid-containing compound, or a reactive equivalent thereof may be reacted with a hydroxyl-containing compound, or a reactive equivalent thereof, so as to provide the corresponding ester.
  • the following reference books and treatise provide exemplary reaction conditions to achieve such conversions: "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandier et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L.
  • the compound of formula (1) can be modified to improve its blood-brain-barrier penetration by conjugation to an organic moiety known to cross the blood-brain-barrier, e.g., docosahexaenoic acid (DHA) and the like.
  • DHA docosahexaenoic acid
  • the conjugation of DHA to the compound of formula (1) can be achieved by following the literature reported procedure. The following references are listed as the examples: Bradley et. al., J. Controlled Release, 74, 233-236, 2001; Katz et. al., US5,716,614; Bradley et. al., US5,955,459; Shashoua et.
  • the conjugation of the compound of formula (1) can be via a hydroxy or an amino group or other function groups which can form a covalent bond with DHA or the like.
  • Scheme 4 is one of the examples for the conjugation of the compound formula (1) to a DHA molecule via an amino group on the phenyl ring.
  • the compound of formula (1a) can be prepared by standard amide formation known to those skilled in the art.
  • the carboxyl group can be converted to an active ester or to an acid chloride or to an anhydride and then the intermediate reacts with the compound of formula (1) containing an amino group.
  • Transcytosis including receptor-mediated transport of compositions across the blood brain barrier, is also suitable for the compounds of the invention.
  • Transferrin receptor-mediated delivery is disclosed in U.S. Patents Nos. 5,672,683; 5,383,988; 5,527,527; 5,977,307; and 6,015,555.
  • Transferrin-mediated transport is also disclosed in Friden, P.M. et al., Pharmacol. Exp. Ther. 278:1491-1498, 1996; and Lee, H.J., J. Pharmacol. Exp. Ther. 292:1048-1052, 2000.
  • EGF receptor-mediated delivery is disclosed in Deguchi, Y. et al., Bioconjug. Chem.
  • BBB penetration of a therapeutic compound may be desired, recent evidence suggests that BBB penetrable compounds may not necessarily be required to decrease CNS ⁇ -amyloid levels.
  • Shibata et al J Clin Invest QS: 1489- 1499, 2000 demonstrate that CSF A ⁇ can be transported across the BBB into the systemic circulation, thereby decreasing A ⁇ in the CNS.
  • a ⁇ interacts with binding proteins such as ApoJ/ApoE, which results in a decrease in "free" A ⁇ in the circulation and shifts the equilibrium to facilitate further transport of A ⁇ out of the CNS.
  • the systemic circulation may act as a "sink” or pool of A ⁇ that can regulate CNS ⁇ -amyloid levels (Shibata, M et al., J Clin Invest 106: 1489-1499, 2000).
  • This "peripheral sink” hypothesis is supported by vaccination studies with anti-A ⁇ antibodies in AD transgenic mouse models. For example, vaccination of PDAPP mice with an A ⁇ antibody (m266) resulted in accumulation of CNS derived A ⁇ in the plasma (DeMattos et al., PNAS 98: 8850-8855, 1998; Holtzman et al., Adv Drug Delivery Rev 54: 1603-1613, 2002).
  • a compound of formula (1) is conjugated to another compound in order to provide an agent, where the agent has enhanced ability to cross the BBB relative to the compound of formula (1).
  • Methods of conjugating a biologically active agent to a compound, and suitable compounds that upon conjugation to a biologically active agent provide a conjugate having enhanced ability to cross the BBB are well known from the above-cited references, and these same techniques may be applied to effectively enhance the permeability of compounds of formula (1) to the BBB.
  • compositions and Administration The compounds of this invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds of the present invention can be formulated into pharmaceutical compositions by combination with appropriate pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. As such, administration of the compounds can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intracheal, efc., administration.
  • the active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.
  • the compounds may be administered in the form of their pharmaceutically acceptable salts. They may also be used in appropriate association with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • the compounds can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • the compounds can be formulated into preparations for injections by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • the compounds can be utilized in aerosol formulation to be administered via inhalation.
  • the compounds of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoro-methane, propane, nitrogen and the like.
  • the compounds can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • the compounds of the present invention can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at ambient temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more compounds of the present invention.
  • unit dosage forms for injection or intravenous administration may comprise the compound of the present invention in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier. Implants for sustained release formulations are well known in the art.
  • Implants are formulated as microspheres, slabs, etc. with biodegradable or non- biodegradable polymers.
  • biodegradable or non- biodegradable polymers For example, polymers of lactic acid and/or glycolic acid form an erodible polymer that is well tolerated by the host.
  • the implant containing the inhibitory compounds is placed in proximity to the site of the tumor, so that the local concentration of active agent is increased relative to the rest of the body.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the novel unit dosage forms of the present invention depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are readily available to the public.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • the combined use of the provided inhibitory compounds and other cytotoxic agents has the advantages that the required dosages for the individual drugs is lower, and the effect of the different drugs complementary.
  • the subject inhibitory compounds may be administered in dosages of 0.1 ⁇ g to 40 mg/kg body weight per day.
  • the range is broad, since in general the efficacy of a therapeutic effect for different mammals varies widely with doses typically being 20, 30 or even 40 times smaller (per unit body weight) in man than in the rat.
  • the mode of administration can have a large effect on dosage.
  • oral dosages in the rat may be ten times the injection dose. Higher doses may be used for localized routes of delivery.
  • a typical dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient, etc.
  • the time-release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
  • dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Some of the specific compounds are more potent than others.
  • Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given compound.
  • the subject compounds may be formulated with other pharmaceutically active agents known to one of ordinary skill in the art.
  • the compounds and pharmaceutical compositions of the invention are administered to a subject having a pathology associated with increased accumulation or deposition of the ⁇ -amyloid peptide such as but not limited to Alzheimer's disease.
  • the present compounds are useful for prophylactic or therapeutic purposes.
  • the prevention of A ⁇ accumulation and deposition is accomplished by administration of a compound of formula (1) prior to development of overt disease, e.g., to prevent ⁇ -amyloid production, release and/or accumulation in the form of plaques, etc.
  • the compounds are used to treat ongoing disease, by stabilizing or improving the clinical symptoms of the patient.
  • the host, or patient may be from any mammalian species, e.g., primate sp., particularly humans; rodents, including mice, rats and hamsters; guinea pigs; rabbits; equines, bovines, canines, felines; efc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease.
  • One method to identify a subject in need of treatment according to the present invention is to measure cognitive, behavioural and/or memory abilities of the subject. If a subject displays impairment in cognitive functioning, particularly if the subject's cognitive ability declines over time, then the subject may benefit from treatment according to the present invention.
  • the subject is a human, then cognitive function and impairment indicative of probable Alzheimer's disease can be assessed using psychological and other tests known to those skilled in the art. If the human subject displays characteristics consistent with a disease caused by increased accumulation and/or deposition of the ⁇ -amyloid peptide, such as but not limited to Alzheimer's disease, then the subject may benefit from the treatment according to the present invention.
  • the susceptibility of a particular cell to treatment with the subject compounds may be determined by in vitro testing.
  • a culture of the cell is combined with a compound of formula (1) at varying concentrations for a period of time sufficient to allow the active agents to decrease production and/or release of A ⁇ , usually between about one hour and one week.
  • cultured cells from a biopsy sample may be used.
  • the dose will vary depending on the specific compound utilized, specific disorder, patient status, etc. Typically a therapeutic dose will be sufficient to produce a substantial decrease ⁇ -amyloid production and/or release in the targeted tissue, while maintaining patient viability. Treatment will generally be continued until there is a substantial reduction, e.g., at least 10%, in ⁇ -amyloid levels and may be continued chronically.
  • the present invention provides a method for modulating the production and/or release of ⁇ -amyloid in a cell, comprising treating said cell with a compound of formula (1).
  • the cell is a brain cell; and/or the ⁇ -amyloid is ⁇ -amyloid 42; and/or ⁇ -amyloid production and/or release in the cell is reduced; and/or the cell is treated in vitro.
  • the present invention provides a method of treatment comprising modulating the production and/or release of ⁇ -amyloid in a non-human mammal in need of said treatment, said method comprising administering to said non- human mammal an effective amount of a compound of formula (1).
  • the cell is a brain cell; and/or ⁇ -amyloid is ⁇ -amyloid 42; and/or the non- human mammal is a mouse, rat, cat, dog or guinea pig; and/or ⁇ amyloid production and/or release is reduced.
  • the present invention provides a method of treatment wherein the production and/or release of ⁇ -amyloid is modulated in a human in need of said treatment, said method comprising administering to said human an effective amount of a compound of formula (1).
  • the human is afflicted with Alzheimer's disease; and/or the human has suffered a head injury; and/or the human has a genetic predisposition or environment exposure that increases the likelihood that said person will develop Alzheimer's disease; and/or the human exhibits minimal cognitive impairment suggestive of early stage Alzheimer's disease; and/or the production and/or release of the ⁇ -amyloid is a brain cell is modulated; and/or the ⁇ - amyloid is ⁇ -amyloid 42; and/or ⁇ -amyloid production and/or release is reduced.
  • the method of the present invention may preferentially reduce production and/or release of A ⁇ 42 relative to one or more other forms of A ⁇ , in a target that produces and/or releases A ⁇ 42, for instance a target selected from a cell, a human, a non-human mammal, and the brain of a human. Tests to identify the selectively of such a compound are disclosed herein.
  • the present invention provides that a subject in need to selective reduction of A ⁇ 42 relative to one or more other forms of A ⁇ , is administered a compound of formu-a (1) that affords such selectively.
  • 2,3-Dimethylphenol (0.61 g, 5 mmol) was added to a solution of NaOH (0.20 g, 5 mmol) in ethanol (EtOH, 20 mL) which was prepared by refluxing the mixture until NaOH disappeared. The mixture was kept stirring for about 5 minutes. (4,6- Dichloropyrimidin-2-ylsulfanyl)acetic acid ethyl ester (1.34 g, 5 mmol) was then added to the mixture with stirring. The resulting solution was kept at reflux for 18 hours and then concentrated in vacuo. The residue was redissolved in 80 mL of Et 2 O.
  • SM4 cells Swedish mutant ⁇ -Amyloid Precursor Protein -695 (SM4 cells) were routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10% fetal bovine serum. Cells were seeded into poly-D-Lysine (SIGMA) coated 6-well plates at a density of 5-7 X 10 5 cells per well. Subsequently, the cells were rinsed in 1 ml of PBS and treated with 50-100 ⁇ M Compound 1 for 5 hrs or 50-300 ⁇ M Compound 1 for 16 hrs in serum-free/phenol red-free DMEM.
  • SIGMA poly-D-Lysine
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide
  • the cells were lysed in 0.1 %> Triton X-100 in PBS supplemented with 5 ⁇ M propridium iodide (Molecular Probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence.
  • a ⁇ 40 and A ⁇ 42 were standardized against propridium iodide fluorescence as a measure of total cell number.
  • Compound 1 induced a significant decrease in A ⁇ 42 production and/or release from SM-4 cells after 5 and 16 hrs.
  • 100 ⁇ M Compound 1 reduced A ⁇ 42 for 92% (p ⁇ 0.001) relative to the control after 5 hrs.
  • a 16 hr treatment at 300 ⁇ M resulted in a 98%. decrease in A ⁇ 42 (p ⁇ 0.001).
  • these concentrations of Compound 1 also resulted in a significant decrease in A ⁇ 40 at 5 and 16 hrs.
  • SM4 cells Swedish mutant ⁇ -Amyloid Precursor Protein -695 (SM4 cells) were routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10% fetal bovine serum. Cells were seeded into poly-D-Lysine (SIGMA) coated 6-well plates at a density of 5-7 X 10 5 cells per well. Subsequently, the cells were rinsed in 1 ml of PBS and treated with 50-300 ⁇ M Compound 2 for 16 hrs in serum-free/phenol red-free DMEM.
  • SIGMA poly-D-Lysine
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide
  • the cells were lysed in 0.1 % Triton X-100 in PBS supplemented with 5 ⁇ M propridium iodide (Molecular Probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence.
  • a ⁇ 40 and A ⁇ 42 were standardized against propridium iodide fluorescence as a measure of total cell number.
  • 293 EBNA cells stably transfected with Swedish mutant ⁇ -Amyloid Precursor Protein -695 are maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10% ⁇ fetal bovine serum.
  • Cells are seeded into Poly-D-Lysine coated 6-well plates at a density of 5-7 X 10 5 cells per well. Subsequently, the cells are rinsed in 1 ml of PBS and treated with various concentrations of APP01643 analogs in serum- free/phenol red-free DMEM for various times.
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide
  • the cells are lysed in 0.1%) Triton X-100 in PBS supplemented with 5 ⁇ M propridium iodide (Molecular Probes, Eugene, Oregon) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Secreted A ⁇ 40 and A ⁇ 42 are standardized against propridium iodide fluorescence as a measure of total cell number.
  • EXAMPLE 8 SCREENING AGENTS FOR ABILITY TO DECREASE ⁇ -AMYLoiD PRODUCTION FROM HUMAN NEUROBLASTOMA CELLS
  • Human neuroblastoma cells (hDAT; SK-N-MC stably overexpressing human dopamine transporter) are routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10%) fetal bovine serum. Cells are seeded into 6-well plates at a density of 2.5 X 10 5 cells per well and transiently transfected with APPsw (Swedish mutant ⁇ -amyloid precursor protein-695) using lipofectamine (Life Technologies, Rockville, Maryland) as per the manufacturer's suggested protocol. Subsequently, 48 hours post-transfection the cells are rinsed with PBS and treated with vehicle (0.1%> DMSO) or various concentrations of APP01643 analogs in serum free/phenol free DMEM for various times.
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide
  • sample treatment buffer 40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide
  • the cells are lysed in 0.1% Triton X-100 in PBS supplemented with 5 ⁇ M propridium iodide (Molecular probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Secreted A ⁇ 40 and A ⁇ 42 levels are standardized against propridium iodide fluorescence as a measure of total cell number.
  • the cDNA coding for human APP695 is cloned in the Smal site of pSFV-1 as described previously (Simons et al., J. Neurosci. 16:899-908, 1996; Tienari et al., Embo. J. 15:5218-29, 1996).
  • PSFV-1 /huAPP695 constructs are linearized with Spel and run-off transcription using SP6 polymerase is performed to produce mRNA.
  • the transcribed mix of APP and pSFV-helper are cotransfected into BHK cells by electroporation to yield recombinant SFV (Olkkonen et al., J. Neurosci. Res. 35:445-51 , 1993).
  • BHK cells are grown in DMEM/F12 supplemented with 5% fetal calf serum,
  • Cortical neurons are incubated with increasing concentrations of compounds of formula (1) (stock solution 400 mM in DMSO).
  • stock solution 400 mM in DMSO a concentrated dilution series is prepared in DMSO comprising 4, 20, 40 and 200 mM compound. From each of these solutions, 2.5 ⁇ l is added to the neuronal cultures in 2 ml of neurobasal medium (dilution 1/800) resulting in 5, 25, 50 and 250 ⁇ M final concentrations.
  • 2.5 ⁇ l of DMSO is added to one dish.
  • the medium is replaced by 1.2 ml neurobasal medium and cultures are transduced by adding recombinant pSFV- humAPP695wt (dilution 1/10) for 1 hour to allow viral entry.
  • cultures are metabolically labeled using methionine-free neurobasal medium containing 100 ⁇ Ci [35S]-methionine (ICN).
  • ICN methionine-free neurobasal medium containing 100 ⁇ Ci [35S]-methionine
  • the cleared fractions are subject to immunoprecipitation with antibodies on protein G-Sepharose (Pharmacia). A ⁇ total is examined from the cleared conditioned media by immunoprecipitation using pab B7, directed against the first 17 amino acids of A ⁇ (De Strooper et al., Embo. J. 14:4932-8, 1995). After overnight rotation, the immunoprecipitates are washed 5 times in extraction buffer and once in TBS. The bound material is denatured in sample buffer and subject to gel electrophoresis on precast 4-12%> Nupage gels. Densitometric analysis is conducted using a Phosphoimager (Molecular Dynamics) and ImagQuant 5.0. A ⁇ total levels are normalized to APP levels to control for plate-to-plate variation.
  • a ⁇ 42 Quantification of A ⁇ 42 by ELISA
  • the levels of the longer A ⁇ 42 peptide are quantified in both the conditioned media and cell extracts using a sandwich ELISA test (De Strooper et al., Nature 391 :387-90, 1998; Vanderstichele et al., Amyloid. 7:245-58, 2000).
  • 800 ⁇ l of conditioned medium or cell extract is lyophilized (Savant Speedvac concentrator), dried pellets are dissolved in 400 ⁇ l of sample diluent and applied on a 96-well ELISA plate precoated with the capturing anti-A ⁇ 42 mab 21F12. This antibody only recognizes the final two amino acids of the A ⁇ 42 sequence.
  • adult guinea pigs Upon arrival of the animals from the vendor, adult guinea pigs are housed under alternating 12 hr light/dark cycles with free access to water and food (standard laboratory chow diet). After 5-6 days adjustment to the new environment guinea pigs are anaesthetized with sodium pentobarbital and using standard sterotaxic surgical procedures, the left lateral cerebral ventricle is cannulated. After the minor surgery, the guinea pigs are given an analgesic (Bupivaicane), allowed to recover and monitored to ensure normal behavior (i.e., regular food and water intake, regular rest/activity cycles etc.).
  • CSF A ⁇ 40 and A ⁇ 42 levels are analyzed by a colorimetric ELISA as per the manufacturers protocol (Biosource International Inc, California).
  • the guinea pig animal model is only one of several models known in the art that could be used. Other examples include but are not limited to, AD transgenic mice models expressing various forms of APP (Tg2576; TgAPP/Sw/1 TgAPP/Ld/2, PDAPP), presenilins or combinations of both (Tg2576 plus mutant PS1 , Tg Hu/MoAPP plus PS1) (Games, D. et al., Nature 373:523-527, 1995; Hsiao, K. H. et al., Science 214: 99-102, 1996; Moechars, D. et al., J. Biol Chem. 21 A: 6483-6492, 1999; Holcomb, L. et al., Nature Medicine 4: 97-100, 1998; Borchelt, D. R. et al., Neuron 19: 939-945, 1997).
  • AD transgenic mice models expressing various forms of APP Tg2576; TgAPP/Sw/1 Tg
  • the in vitro model uses a PBEC (porcine brain microvessel endothelial cell) monolayer which is arranged so that the ability of substances to pass from a donor compartment to an acceptor compartment can be measured.
  • PBEC neurotrophic factor-containing cell
  • This model reflects the in vivo situation wherein substances reach the brain compartment from a brain microvessel.
  • Permeation properties of an agent of the invention are measured by radiolabeling the agent, for example with 3 H, and adding it to the donor compartment. Samples are collected from the donor and acceptor compartments at routine intervals and permeability is calculated as described in Franke, H. et al., (2000).
  • the in vivo models measure the brain influx index or the measure of the passage of a substance through the blood brain barrier.
  • the agent is radiolabeled or fluorescently labeled and administered peripherally by intravenous injection (Pan, W., et al., Neuropharmacol. 37:1553-1561 , 1998), orally (Shulkin, B. L. et al., J. Neurochem. 64:1252-1257, 1995) or nasally (Thorne, R.G. et al., Brain Res. 692:278-282, 1995) and the concentration of the agent in the blood as compared to the brain is monitored.
  • the systemic circulation may act as a "sink” or pool of A ⁇ that can regulate CNS ⁇ -amybid levels (Shibata, M et al., J Clin Invest 106: 1489-1499, 2000).
  • This "peripheral sink" hypothesis is supported by vaccination studies with anti-A ⁇ antibodies in AD transgenic mouse models. For example, vaccination of PDAPP mice with an A ⁇ antibody (m266) resulted in accumulation of CNS derived A ⁇ in the plasma (DeMattos et al., PNAS 98: 8850-8855, 2001 ; Holtzman et al., Adv Drug Delivery Rev 54: 1603-1613, 2002).
  • compositions of the invention can be examined for their effects on systemic and CNS ⁇ -amyloid levels.
  • Compounds can be injected into the animal of interest followed by repeated sampling and measurement of plasma ⁇ -amyloid levels over time.
  • An increase in plasma A ⁇ levels coupled with a decrease in CNS levels would indicate that the compound is shifting the A ⁇ equilibrium.
  • the ability of the compound to cross the blood brain barrier in vivo can be measured by standard analytical chemistry techniques (e.g., mass spectroscopy).

Abstract

Pyrimidine compounds, compositions containing pyrimidine compounds and methods of using pyrimidine compounds for regulating the production and/or release of ß-amyloid in cells are provided. The compounds provide for alleviation and prevention of ß-amyloid production, release and/or plaque development as occurs in, e.g., Alzheimer's disease.

Description

ARYLTHIOETHERPYRIMIDINE AND ARYLOXYETHERPYRIMIDINE DERIVATIVES
AND THEIR THERAPEUTIC USES
CROSS-REFERENCE TO RELATED APPLICATION
This application claims the benefit of U.S. Provisional Patent Application No. 60/434,030 filed December 16, 2002, where this Provisional Application is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
Field of the Invention
The invention relates to compounds, compositions and methods for regulating the production and/or release of β-amyloid in cells, and provides for alleviation and prevention of β-amyloid production, release and/or plaque development such as occurs in, e.g., Alzheimer's disease.
Description of the Related Art
Alzheimer's disease (AD) is a common brain disorder of the elderly and is associated with progressive dementia. The key features of the disease include progressive memory impairment, loss of language and visuospatial skills, and behavior deficits. These changes in cognitive function are the result of degeneration of neurons in the cerebral cortex, hippocampus, basal forebrain, and other regions of the brain. Neuropathological analyses of postmortem Alzheimer's diseased brains consistently reveal the presence of large numbers of neurofibrillary tangles in degenerated neurons and neuritic plaques in the extracellular space and walls of the cerebral microvasculature. The neurofibrillary tangles are composed of bundles of paired helical filaments containing hyperphosphorylated tau protein (Lee, V. M and Trojanowski, J. Q. Curr. Opin. Neurobiol. 2:653, 1992). The neuritic plaques consist of deposits of proteinaceous material surrounding a β-amyloid core (Selkoe, D. J., Annu. Rev. Neurosci. 77:489-517, 1994).
Evidence suggests that accumulation and deposition of β-amyloid peptide (Aβ) plays a significant role in the etiology of Alzheimer's disease. This evidence is based upon 1) biophysical and biochemical properties of Aβ, 2) genetic analysis based on patients with familial forms of Alzheimer's disease (FAD), and 3) in vitro and in vivo AD models.
The 39-43 amino acid Aβ peptide is produced by sequential proteolytic cleavage of the β-amyloid precursor protein (APP). Alternative splicing generates several different isoforms of APP; in neurons, the predominant isoform is 695 amino acids in length (APP695). As APP traverses the endoplasmic reticulum (ER) and trans- Golgi network (TGN), it becomes N- and O-glycosylated and tyrosine-sulfated. Mature holoprotein can be catabolized in several compartments to produce both non- and amyloidogenic APP fragments. APP is expressed and constitutively catabolized in most cells. The dominant catabolic pathway appears to be cleavage of APP within the Aβ sequence by an enzyme provisionally termed α-secretase, leading to release of a soluble ectodomain fragment known as APPsα. In contrast to this non-amyloidogenic pathway, APP can also be cleaved by enzymes known as β- and γ-secretase at the N- and C-termini of the Aβ, respectively, followed by release of Aβ into the extracellular space. To date, BACE1 (β-site APP cleaving enzyme) and to a lesser extent BACE2, have been identified as β-secretase (Vassar et al., Science 286:735-741 , 1999; Farzan M et al., Proc. Natl. Acad. Sci 97: 9712-9713, 2000), and presenilins have been implicated in γ- secretase activity (De Strooper et al., Nature 391 :387-390, 1998). Although Aβ40 is the predominant form of Aβ produced by sequential cleavage of APP, 5-7% of total Aβ exists as Aβ42 (Cappai et al., Int. J. Biochem. Cell Biol. 37:885-889, 1999). The length of the Aβ peptide appears to dramatically alter its biochemical and biophysical properties. Specifically, the additional two amino acids at the C-terminus of Aβ42 are hydrophobic, presumably increasing the propensity of Aβ42 to aggregate. For example, Jarrett et al. demonstrated that Aβ42 aggregates very rapidly in vitro compared to Aβ40, suggesting that the longer forms of Aβ may be the important pathological proteins that are involved in the initial seeding of the neuritic plaques in AD (Jarrett et al., Biochemistry 32:4693-4697, 1993; Jarrett et al., Ann. NY Acad. Sci. 695: 144-148, 1993).
The role of Aβ (specifically Aβ42) has been further substantiated by the recent analysis of familial forms of AD (FAD). To date, this aggressive form of Alzheimer's disease has been shown to be caused by missense mutations in (at least) three genes: the β-amyloid precursor protein (APP) gene itself (Goate, A. et al., Nature 349:704-706, 1991; Mullan, M. et al., Nature Genet. 1345-347, 1992), and two genes termed presenilins 1 and 2 (Sherrington, R. et al., Nature 375:754-760, 1995; Rogaev, E. I. et al., Nature 376:775-778, 1995).
The "London" mutant form of APP (APPV717I) linked to FAD selectively increases the production of Aβ42/43 forms versus Aβ40 (Suzuki et al., Science 264:1336-1340, 1994) while the "Swedish" mutant form of APP (APPK670N/M671L) increases levels of both Aβ40 andAβ42/43 (Citron et al., Nature 360:672-674, 1992; Cai et al., Science 259:514-516, 1993). Also, it has been observed that FAD-linked mutations in the Presenilin-1 (PS1) or Presenilin-2 (PS2) genes will lead to a selective increase in Aβ42/43 production bit not Aβ40 (Borchelt et al., Neuron 77:1005-1013, 1996). This finding was corroborated in transgenic mouse models expressing PS mutants that demonstrate a selective increase in brain Aβ42 (Borchelt et al., Neuron 77:1005-1013, 1996; Duff et al., Neurodegeneration 5(4j:293-298, 1996).
Collectively the wealth of data derived from 1) biophysical and biochemical properties of Aβ, 2) genetic analysis based on patients with familial forms of Alzheimer's disease (FAD) and 3) in vitro and in vivo AD models - all point to Aβ42 as the key pathogenic peptide in AD. This notion led to the formulation of the 'amyloid cascade hypothesis' (Hardy et al., Science 256: 184-185, 1992; Hardy and Selkoe, Science 297: 353-356, 2002). The amyloid cascade hypothesis originally suggested that increased accumulation and deposition of the Aβ peptide (primarily Aβ42) leads to senile plaque formation and subsequent neuronal death and dementia (Hardy et al., Science 256: 184-185, 1992). However, this hypothesis does not explain the lack of correlation observed between the degree of dementia and the absolute number of β-amyloid deposits in the brain (Dickson et al., Neurobiol Aging 10: 402-404, 1989). In fact, Wang et al. (1999) have suggested that the degree of dementia in AD patients shows a stronger correlation with the levels of soluble Aβ as opposed to the number of histologically determined plaques (Wang et al., Exp. Neurol. 158: 328, 1999). Along these lines, other studies have suggested that soluble Aβ42 oligomers (Aβ derived diffusible ligands; ADDL) are toxic to neurons and may be responsible for the impaired synaptic plasticity and memory dysfunction observed in early stage AD (Lambert et al., Proc. Natl. Acad. Sci 95: 6448-6453, 1998). Thus, more recently the original amyloid cascade hypothesis has been expanded to encompass the toxic nature of soluble Aβ oligomers (Hardy and Selkoe, Science 297: 353-356, 2002).
Nevertheless, based on the evidence to date there is a need for treatments which selectively inhibit the production and/or release of Aβ42. Such treatments may prove to be extremely valuable in the treatment of both familial and/or sporadic cases of AD. In fact, recent in vitro studies have demonstrated that certain non-steroidal anti-inflammatory drugs (NSAIDs) selectively decrease Aβ42 (Weggen et al., Nature 414: 212-216, 2001).
Epidemiological studies have indicated a decreased prevalence of AD amongst patients who take NSAIDs (McGeer et al., Neurology 47: 425-432, 1996). Although the mechanism of this effect is unknown, Weggen et al. (2001) have demonstrated that certain NSAIDs selectively decrease Aβ42 in froand that this effect does not appear to be mediated by inhibition of cyclooxygenase (COX) activity; the primary target for NSAIDs. For example, sulindac sulphide and indomethacin reduced Aβ42 by 80% at concentrations of 100 (Jvl without altering Aβ40 in Chinese hamster ovarian cells (CHO) over-expressing APP (Weggen et al., Nature 414: 212-216, 2001). Similarly, Morihara et al. (2002) demonstrated that specific stereoisomers of NSAIDs, namely R-ibuprofen and R-flurbiprofen, selectively decreased Aβ42 release from HEK- 293 cells over-expressing the 'Swedish mutant' form of APP. Interestingly, R- flurbiprofen reduced Aβ42 by only 40% at a concentration of 2® μM relative to the control (Morihara et al., J. Neurochem 83: 1009-1012, 2002). Although NSAIDs appear to selectively decrease levels of Aβ42, the concentrations employed to achieve this effect appear to be much higher than those used to inhibit COX activity in clinically relevant situations. For example, NSAIDs have IC50 values in the nanomolar to low micromolar ranges for COX enzymes (Neupert et al., Br. J. Pharmacol. 122: 487-492, 1997), while their Aβ42 lowering effects are seen at much higher concentrations (e., 100-500 μM). As a result, toxicity associated with high concentrations of NSAIDs may restrain them from being used widely as AD therapeutics (Morihara et al., J. Neurochem 83: 1009-1012, 2002).
BRIEF SUMMARY OF THE INVENTION This invention is directed to certain pyrimidine compounds, pharmaceutical compositions containing certain pyrimidine compounds, and the use of certain pyrimidine compounds for regulating the production and/or release of β-amyloid in cells, and for alleviation and prevention of β-amyloid production, release and/or plaque development. In one aspect, the present invention provides pyrimidine compounds of formula (1):
Figure imgf000007_0001
wherein, independently at each occurrence, Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6,
-N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl.
The invention includes these compounds as single compounds or mixtures of compounds; in pure or impure form; when stereoisomers are possible then the invention provides the compound as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; and as pharmaceutically acceptable salts.
The present invention also provides compositions that include at least one of these compounds. For example, in one aspect the present invention provides a pharmaceutical composition comprising a pyrimidine compound of the invention. As another example, the present invention provides a composition comprising a pyrimidine compound of the invention and a pharmaceutically acceptable carrier, excipient or diluent.
The invention provides a method for modulating the production and/or release of β-amyloid from a cell, comprising treating the cell with a compound of formula (1) or a composition comprising a compound of formula (1).
The invention also provides a method of treatment comprising modulating the production and/or release of β-amyloid in a non-human mammal in need of said treatment, said method comprising administering to said non-human mammal a compound that can modulate the production and/or release of β-amyloid in a human, or a composition comprising such a compound.
The invention also provides a method of treatment comprising modulating the production and/or release of β-amyloid in a human in need of said treatment, said method comprising administering to said human a compound that can modulate the production and/or release of β-amyloid in a human, or a composition comprising such a compound.
The invention yet further provides a method for preferentially reducing production and/or release of Aβ42 relative to one or more other forms of Aβ, in a target that produces and/or releases Aβ42, for instance a target selected from a cell, a human, a non-human mammal, and the brain of a human, comprising administering to the target a compound or pharmaceutical composition comprising a chemical agent as described herein. This method may be used to treat, e.g., a human, wherein said human, e.g., is afflicted with Alzheimer's disease. In another embodiment, said human being treated has a genetic predisposition or environment exposure that increases the likelihood that said person will develop Alzheimer's disease. For example, said human has suffered a head injury and is treated with a compound or composition as described herein. In one embodiment, said human exhibits minimal cognitive impairment suggestive of early stage Alzheimer's disease. In another embodiment, said human has suffered a head injury and is treated with a compound or composition as described herein. Towards this end, the invention also provides a method for delivering to the brain a compound capable of modulating Aβ production and/or release. This delivery system achieves specific delivery of such compounds through conjugating the compounds with a polar lipid or other carrier, achieving effective intracerebral concentration of such compounds efficiently and with specificity.
These and other aspects of the present invention will be decribed in detail below.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a line graph showing the effect of Compound 1 on secreted Aβ40 and Aβ42 from SM-4 cells. Cells were treated with vehicle (0.01 % w/v DMSO) or 50-100 μM Compound 1 for 5 hrs (A) or 50-300 μM of Compound 1 for 16 hrs (B). After the treatment, the culture media was harvested and assayed for Aβ40 and Aβ42 by ELISA. Secreted Aβ was standardized to propridium iodide fluorescence as a measure of total cell number. Data are expressed as mean ± SEM with n = 3-4 and statistical significance determined by ANOVA with Tukey's post hoc test at **p<0.01 , ***p<0.001 Figure 2 is a line graph showing the effect of Compound 2 on secreted Aβ40 and Aβ42 in SM-4 cells. Cells were treated with vehicle (0.01% w/v DMSO) or 50- 300 μM Compound 2. After 16 hrs, the culture media was harvested and assayed for Aβ40 and Aβ42 by ELISA. Secreted Aβ was standardized to propridium iodide fluorescence as a measure of total cell number. Data are expressed as mean ± SEM with π = 4 and statistical significance determined by ANOVA with Tukey's post hoc test. DETAILED DESCRIPTION OF THE INVENTION
A. Definitions
In general, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs, unless clearly indicated otherwise. For clarification, listed below are definitions for certain terms used herein to describe the present invention. These definitions apply to the terms as they are used throughout this specification, unless otherwise clearly indicated.
As used herein the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. For example, "a compound" refers to one or more of such compounds, while "the enzyme" includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
"Alkyl" refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, π-propyl, 1-methylethyl (iso-propyl), π-butyl, n-pentyl, 1 ,1-dimethylethyl (f-butyl), and the like. "Alkylene chain" refers to a straight or branched divalent hydrocarbon chain consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twenty carbons atoms, preferably having from one to eight carbons, e.g., methylene, ethylene, propylene, n-butylene, and the like.
"Alkenyl" refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1 ,4-dienyl, and the like. "Alkynyl" refers to a straight or branched monovalent hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from one to twenty carbon atoms, preferably from one to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, prop-1-ynyl, pent-1-ynyl, penta-1 ,4-diynyl, and the like.
"Alkoxy" refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above, e.g., methoxy, ethoxy, n-propoxy, 1-methylethoxy (iso-propoxy), n-butoxy, n-pentoxy, 1 ,1-dimethylethoxy (t-butoxy), and the like.
"Aryl" refers to an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi- electron system. Examples, without limitation, of aryl groups are phenyl, naphthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted. When substituted, the substituted group(s) is one or more selected from alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl, heterocyclylalkyi with R6 and R7 as defined above. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals optionally substituted by one or more substituents selected from the group consisting of alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl, heterocyclylalkyi with R6 and R as defined above. "Aryloxy" refers to a radical of the formula -ORb where Rb is an aryl radical as defined above, e.g., phenoxy and the like.
"Aralkyl" refers to a radical of the formula -RaRb where Ra is an alkyl radical as defined above and Rb is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl, and the like. The aryl radical may be optionally substituted as described above.
"Aralkenyl" refers to a radical of the formula -Re-Rb where Rb is an aryl radical as defined above and Re is an alkenyl radical as defined above, e.g., 2- phenylethenyl, and the like.
"Carboxy" refers to the -C(O)OH radical.
"Cycloalkyl" refers to a stable monovalent monocyclic or bicyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having from three to ten carbon atoms, and which is saturated and attached to the rest of the molecule by a single bond, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decalinyl and the like. Unless otherwise stated specifically in the specification, the term "cycloalkyl" is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents independently selected from the group consisting of alkyl, alkoxy, halo, haloalkyl, haloalkoxy, hydroxy, amino, and carboxy. "Halo" refers to bromo, chloro, iodo or fluoro.
"Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl~2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like. "Haloalkoxy" refers to a radical of the formula -ORc where Rc is an haloalkyl radical as defined above, e.g., trifluoromethoxy, difluoromethoxy, trichloromethoxy, 2,2,2-trifluoroethoxy, 1 -fluoromethyl-2-fluoroethoxy , 3-bromo-2-fluoropropoxy, 1-bromomethyl-2-bromoethoxy, and the like.
"Heteroalkyl" refers to an alkyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), with R6 and R7 as defined above and the substitution can occur on any carbon of the alkyl group, e.g., -CH2CH(CH3)CH2NH2, -CH2CH2OH, -CH2CH(F)CH2NH2, and the like.
"Heteroalkenyl" refers to an alkenyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6,
-N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), with R6 and R7 as defined above and the substitution can occur on any carbon of the alkenyl group, e.g., -CH=CH(CH3)CH2NH2, -CH=CH2CH2OH, and the like. "Heteroalkynyl" refers to an alkynyl radical as defined above substituted with one or more individually selected from halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), with R6 and R7 as defined above and the substitution can occur on any carbon of the alkynyl group, e.g., -C≡CCH2NH2, -C≡CCH2OH, and the like.
"Heterocyclyl" refers to a stable 3- to 15-membered ring radical which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this invention, the heterocyclyl radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quatemized; and the heterocyclyl radical may be aromatic or partially or fully saturated. The heterocyclyl radical may not be attached to the rest of the molecule at any heteroatom atom. Examples of such heterocyclyl radicals include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzothiadiazolyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, carbazolyl, cinnolinyl, decahydroisoquinolyl, dioxolanyl, furanyl, furanonyl, isothiazolyl, imidazolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, indolizinyl, isoxazolyl, isoxazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxoazepinyl, oxazolyl, oxazolidinyl, oxiranyl, piperidinyl, piperazinyl, 4-piperidonyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrrolidinyl, pyrazolyl, pyrazolidinyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, thiazolyl, thiazolidinyl, thiadiazolyl, triazolyl, tetrazolyl, tetrahydrofuryl, triazinyl, tetrahydropyranyl, thienyl, thiamorpholinyl, thiamorpholinyl sulfoxide, and thiamorpholinyl sulfone. Unless stated otherwise specifically in the specification, the term "heterocyclyl" is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, heteroalkyl, haloalkyl, haloalkoxy, aryl, halo, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl, heterocyclylalkyi with R6 and R7 as defined above.
"/V-heterocyclyl" refers to a heterocyclyl radical as defined above wherein the one to five heteroatoms contained therein are selected only from nitrogen, e.g., pyridinyl, tetrazolyl, pyrazolyl, isoquinolinyl, quinolinyl, and phthalazinyl and the like. "Heterocyclylalkyi" refers to a radical of the formula -RaRd where Ra is an alkyl radical as defined above and R is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. The heterocyclyl radical may be optionally substituted as defined above. "Heterocyclylcarbonyl" refers to a radical of the formula -C(O)-Rd where Rd is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the carbonyl at the nitrogen atom. "Hydrocarbon" refers to a compound formed entirely of carbon and hydrogen (including isotopes thereof), while "hydrocarbyl" refers to a hydrocarbon radical.
As used herein, compounds which are "commercially available" may be obtained from standard commercial sources including Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc. (Costa Mesa CA), Key Organics (Cornwall U.K.), Lancaster Synthesis (Windham NH), Maybridge Chemical Co. Ltd. (Cornwall U.K.), Parish Chemical Co. (Orem UT), Pfaltz & Bauer, Inc. (Waterbury CN), Polyorganix (Houston TX), Pierce Chemical Co. (Rockford IL), Riedel de Haen AG (Hannover, Germany), Spectrum Quality Product, Inc. (New Brunswick, NJ), TCI America (Portland OR), Trans World Chemicals, Inc. (Rockville MD), and Wako Chemicals USA, Inc. (Richmond VA).
As used herein, "suitable conditions" for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry. The reference books and treatise set forth above that detail the synthesis of reactants useful in the preparation of compounds of the present invention, will also provide suitable conditions for carrying out a synthetic step according to the present invention.
As used herein, "methods known to one of ordinary skill in the art" may be identified though various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present invention, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandier et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 5th Ed., Wiley-lnterscience, New York, 2000. Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C., www.acs.org may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
"Prodrugs" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam).
A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," ACS. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
The term "prodrug" is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention and the like. "Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
"Mammal" includes humans and domesticated animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like. "Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
"Pharmaceutically acceptable salt" and "salts thereof in the compounds of the present invention refers to pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, Λ/-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. "Pharmaceutically acceptable excipient" as used herein is intended to include without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, emulsifier, or stabilizer which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Treating" or "treatment" as used herein covers the treatment of a disorder in a mammal, preferably a human, which disorder is characterized by the accumulation or deposition of β-amyloid peptide, and includes: (i) preventing the disorder from occurring in a mammal, in particular a human, when such mammal is predisposed to the disorder but has not yet been diagnosed as having it;
(ii) inhibiting the disorder, i.e., arresting its development; or (iii) relieving the disorder, i.e., causing regression of the condition.
B. Compounds
In this invention, compounds of formula (1) are defined as follows:
Figure imgf000020_0001
wherein, independently at each occurrence, Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(Rδ)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl.
The compound(s) may be, for example, a single stereoisomer, a mixture of stereoisomers, a racemic mixture of stereoisomers; in solvated form, as a polymorph; or as a pharmaceutically acceptable salt thereof. In one aspect, the invention provides prodrug forms of compounds of formula (1 ).
In various aspects of the invention, one or more compounds that would otherwise be included within the scope of formula (1) are explicitly excluded. The following are specific exclusions that may be applied to describe compounds of formula (1), where any two or more of the exclusions may be combined so as to create a larger set of excluded compounds.
In one aspect, the compounds of the invention exclude propanoic acid, 2- [[4-[2-chloro-5-[3,6-dihydro-3-methyl-2,6-dioxo-4-(trifluoromethyl)-1(2H)-pyrimidinyl]-4- fluorophenoxy]-2-pyrimidinyl]oxy]-, methyl ester, (having CAS Registry No. (RN) 353292-13-4). In another aspect, the compounds of the invention additionally, or alternatively, exclude: acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2- pyrimidinyljthio]- (RN 91759-31-8); and acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)- 2-pyrimidinyl]thio]-, ethyl ester (RN 91759-33-0). In another aspect, the compounds of the invention additionally, or alternatively, exclude any one or more of the following compounds: propanoic acid, 2- [[4-[(4-bromophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-64-6); propanoic acid, 2-[[4-[(4-chlorophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-63-5); acetic acid, [[5-methoxy-4-[[3-(trifluoromethyl)phenyl]thio]-2- pyrimidinyljthio]-, ethyl ester (RN 338423-49-7); acetic acid, [[4-[(2-chlorophenyl)thio]-5- methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-48-6); acetic acid, [[5-methoxy-4- (4-methoxyphenoxy)-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-47-5); acetic acid, [[5- methoxy-4-(3-nitrophenoxy)-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-44-2); acetic acid, [[5-methoxy-4-(phenylthio)-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-38-4); acetic acid, [[4-(3,5-dichIorophenoxy)-5-methoxy-2-pyrimidinyl]thio]-; ethyl ester (RN 338423-37-3); acetic acid, [[4-(3,4-dichlorophenoxy)-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-36-2); acetic acid, [[5-methoxy-4-[3-(trifluoromethyl)phenoxy]-2- pyrimidinyljthio]-, ethyl ester (RN 338423-35-1); acetic acid, [[5-methoxy-4-[(4- methoxyphenyl)thio]-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-26-0); and acetic acid, [[4-[(4-chlorophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester (RN 338423-25-9).
In other aspects of the compounds of the invention, excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring (where the Y group is bonded to the 1 position of the carbocyclic aromatic ring). In another aspect of the invention, excluded are compounds having 2,3-(C C3 alkyl) (i.e., alkyl groups having 1 , 2 or 3 carbons) substitution on the carbocyclic aromatic ring. In another aspect of the invention, excluded are compounds having excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring when Y is oxygen. In another aspect of the invention, excluded are compounds having 2,3-(CrC3 alkyl) (i.e., alkyl groups having 1, 2 or 3 carbons) substitution on the carbocyclic aromatic ring when Y is oxygen. In another aspect of the invention, excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring when W is OH or OC-i-C2 alkyl). In another aspect of the invention, excluded are compounds having 2,3-(C1-C3 alkyl) (λe., alkyl groups having 1 , 2 or 3 carbons) substitution on the carbocyclic aromatic ring when W is OH or OC-ι-C2 alkyl). In another aspect of the invention, excluded are compounds having 2,3-dimethyl substitution on the carbocyclic aromatic ring when W is OH or OCι-C2 alkyl) and Y is oxygen. In another aspect of the invention, excluded are compounds having 2,3-(CrC3 alkyl) (i.e., alkyl groups having 1, 2 or 3 carbons) substitution on the carbocyclic aromatic ring when W is OH or OCι-C2 alkyl) and Y is oxygen.
In various aspects of the invention, one or any combination of the following provisos may be added to a claim or other statement of the invention so as to preclude (exclude) certain chemical groups from being present at positions of the compounds (where distinct provisos are separated by semicolons, and any plurality of provisos may be combined to create a larger proviso): Y is not -S-; Y is not -S(O)-; Y is not -S(O)2-;Y is not -S(O)m-; Y is not -O-; W is not -OR4, W is not -N(R5)2; W is not -NHN(R5)2; p is not 0; p is not 1 ; p is not 2; p is not 3; p is not 4; p is not 5; q is not 0; q is not 1 ; q is not 2; m is not 0; m is not 1 ; m is not 2; n is not 0; n is not 1 ; n is not 2; n is not 3; n is not 4; n is not 5; R1 is not alkyl; R1 is not alkenyl; R1 is not alkynyl; R1 is not heteroalkyl; R1 is not heteroalkenyl; R1 is not heteroalkynyl; R is not aryl; R1 is not aralkyl; R1 is not aralkenyl; R is not cycloalkyl; R1 is not cycloalkylalkyl; R1 is not halo; R1 is not haloalkyl; R1 is not haloalkoxy; R1 is not nitro; R1 is not cyano; R1 is not -NHOH; R1 is not -OR7; R1 is not -SR7; R1 is not -C(O)OR7; R1 is not -OC(O)R7; R1 is not -C(O)N(R6)2; R1 is not -C(S)R6; R1 is not -C(O)R6; R1 is not -N(R6)2; R1 is not
-N(R6)C(O)R6; R1 is not -N(R6)C(O)OR7; R1 is not -S(O)tR6 (where t is not 0 to 2); R1 is not -S(O)tN(R6)2 (where t is not 0 to 2); R1 is not -OC(S)NR6; R1 is not -NR6C(S)OR7; R1 is not -NR6S(O)tR6 (where t is not 0 to 2); R1 is not heterocyclyl; R1 is not heterocyclylalkyi; R2 is not alkyl; R2 is not alkenyl; R2 is not alkynyl; R2 is not heteroalkyl; R2 is not heteroalkenyl; R2 is not heteroalkynyl; R2 is not aryl; R2 is not aralkyl; R2 is not aralkenyl; R2 is not cycloalkyl; R2 is not cycloalkylalkyl; R2 is not halo; R2 is not haloalkyl; R2 is not haloalkoxy; R2 is not nitro; R2 is not cyano; R2 is not -NHOH; R2 is not -OR7; R2 is not -SR7; R2 is not -C(O)OR7; R2 is not -OC(O)R7; R2 is not -C(O)N(R6)2; R2 is not -C(S)R6; R2 is not -C(O)R6; R2 is not -N(R6)2; R2 is not -N(R6)C(O)R6; R2 is not -N(R6)C(O)OR7; R2 is not -S(O)tR6 (where t is not 0 to 2); R2 is not -S(O)tN(R6)2 (where t is not 0 to 2); R2 is not -OC(S)NR6; R2 is not -NR6C(S)OR7; R2 is not -NR6S(O)tR6 (where t is not 0 to 2); R2 is not heterocyclyl; R2 is not heterocyclylalkyi; R3 is not hydrogen; R3 is not alkyl; R3 is not alkenyl; R3 is not aryl; R3 is not aralkyl; R3 is not aralkenyl; R3 is not cycloalkyl; R3 is not cycloalkylalkyl; R3 is not halo; R3 is not haloalkyl; R3 is not haloalkoxy; R3 is not nitro; R3 is not cyano; R3 is not -NHOH; R3 is not -OR7; R3 is not -SR7; R3 is not -C(O)OR7; R3 is not -OC(O)R7; R3 is not -C(O)N(R6)2; R3 is not -C(S)R6; R3 is not -C(O)R6; R3 is not -N(R6)2; R3 is not -N(R6)C(O)R6; R3 is not -N(R6)C(O)OR7; R3 is not -OC(S)NR6; R3 is not -NR6C(S)OR7; R3 is not -OR7; R3 is not -C(O)OR7; R3 is not -OC(O)R7; R3 is not -C(O)N(R6)2; R3 is not heterocyclyl; R3 is not heterocyclylalkyi; R4 is not hydrogen, R4 is not alkyl; R4 is not alkenyl; R4 is not alkynyl; R4 is not heteroalkyl; R4 is not heteroalkenyl; R4 is not heteroalkynyl; R4 is not aryl; R4 is not aralkyl; R4 is not aralkenyl; R4 is not cycloalkyl; R4 is not cycloalkylalkyl; R4 is not haloalkyl; R4 is not haloalkoxy; R4 is not heterocyclyl; R4 is not heterocyclylalkyi; R5 is not hydrogen; R5 is not alkyl; R5 is not alkenyl; R5 is not alkynyl; R5 is not heteroalkyl; R5 is not heteroalkenyl; R5 is not heteroalkynyl; R5 is not aryl; R5 is not aralkyl; R5 is not aralkenyl; R5 is not cycloalkyl; R5 is not cycloalkylalkyl; R5 is not haloalkyl; R5 is not haloalkoxy; R5 is not heterocyclyl; R5 is not heterocyclylalkyi; R6 is not hydrogen; R6 is not alkyl; R6 is not alkenyl; R6 is not cycloalkyl; R6 is not cycloalkylalkyl; R6 is not aralkyl; R6 is not aryl; R7 is not hydrogen; R7 is not alkyl; R7 is not aralkyl.
While the foregoing has provided various optional embodiments of the invention whereby particular compounds and/or groups of compounds within the scope of formula (1) may be excluded from the invention, the present invention also provides that compounds of formula (1) may be described by limiting the scope of a parameter that characterizes the compounds of formula (1). The term "parameter" is intended to refer to the various R groups (e.g., R , R2, etc.), integers (m, n, etc.), forms (salt, steroisomer, etc.) or any other characterizing feature of the compounds of formula (1).
Thus, in various aspects of the compounds, compositions and methods of the invention (where separate and distinct aspects are separated by semicolons) the compounds may be specified by requiring one or more of the following conditions: Y is -S-; Y is -S(O)-; Y is -S(O)2-;Y is -S(O)m-; Y is -O-; W is -OR4, W is -N(R5)2; W is -NHN(R5)2; p is 0; p is 1 ; p is 2; p is 3; p is 4; p is 5; q is 0; q is 1 ; q is 2; m is 0; m is 1 ; m is 2; n is 0; n is 1 ; n is 2; n is 3; n is 4; n is 5; R1 is alkyl; R1 is alkenyl; R1 is alkynyl; R1 is heteroalkyl; R is heteroalkenyl; R1 is heteroalkynyl; R1 is aryl; R1 is aralkyl; R1 is aralkenyl; R1 is cycloalkyl; R1 is cycloalkylalkyl; R1 is halo; R1 is haloalkyl; R1 is haloalkoxy; R1 is nitro; R1 is cyano; R1 is -NHOH; R is -OR7; R1 is -SR7; R1 is -C(O)OR7; R1 is -OC(O)R7; R1 is -C(O)N(R6)2; R1 is -C(S)R6; R1 is -C(O)R6; R1 is -N(R6)2; R1 is -N(R6)C(O)R6; R1 is -N(R6)C(O)OR7; R1 is -S(O)tR6 (where t is 0 to 2); R1 is -S(O)tN(R6)2 (where t is 0 to 2); R is -OC(S)NR6; R1 is -NR6C(S)OR7; R1 is
-NR6S(O)tR6 (where t is 0 to 2); R1 is heterocyclyl; R1 is heterocyclylalkyi; R2 is alkyl; R2 is alkenyl; R2 is alkynyl; R2 is heteroalkyl; R2 is heteroalkenyl; R2 is heteroalkynyl; R2 is aryl; R2 is aralkyl; R2 is aralkenyl; R2 is cycloalkyl; R2 is cycloalkylalkyl; R2 is halo; R2 is haloalkyl; R2 is haloalkoxy; R2 is nitro; R2 is cyano; R2 is -NHOH; R2 is -OR7; R2 is -SR7; R2 is -C(O)OR7; R2 is -OC(O)R7; R2 is -C(O)N(R6)2; R2 is -C(S)R6; R2 is -C(O)R6; R2 is -N(R6)2; R2 is -N(R6)C(O)R6; R2 is -N(R6)C(O)OR7; R2 is -S(O)tR6 (where t is 0 to 2); R2 is -S(O)tN(R6)2 (where t is 0 to 2); R2 is -OC(S)NR6; R2 is -NR6C(S)OR7; R2 is -NR6S(O)tR6 (where t is 0 to 2); R2 is heterocyclyl; R2 is heterocyclylalkyi; R3 is hydrogen; R3 is alkyl; R3 is alkenyl; R3 is aryl; R3 is aralkyl; R3 is aralkenyl; R3 is cycloalkyl; R3 is cycloalkylalkyl; R3 is halo; R3 is haloalkyl; R3 is haloalkoxy; R3 is nitro; R3 is cyano; R3 is -NHOH; R3 is -OR7; R3 is -SR7; R3 is -C(O)OR7; R3 is -OC(O)R7; R3 is -C(O)N(R6)2; R3 is -C(S)R6; R3 is -C(O)R6; R3 is -N(R6)2; R3 is -N(R6)C(O)R6; R3 is -N(R6)C(O)OR7; R3 is -OC(S)NR6; R3 is -NR6C(S)OR7; R3 is -OR7; R3 is -C(O)OR7; R3 is -OC(O)R7; R3 is -C(O)N(R6)2; R3 is heterocyclyl; R3 is heterocyclylalkyi; R4 is hydrogen; R4 is alkyl; R4 is alkenyl; R4 is alkynyl; R4 is heteroalkyl; R4 is heteroalkenyl; R4 is heteroalkynyl; R4 is aryl; R4 is aralkyl; R4 is aralkenyl; R4 is cycloalkyl; R4 is cycloalkylalkyl; R4 is haloalkyl; R4 is haloalkoxy; R4 is heterocyclyl; R4 is heterocyclylalkyi; R5 is hydrogen; R5 is alkyl; R5 is alkenyl; R5 is alkynyl; R6 is heteroalkyl; R5 is heteroalkenyl; R5 is heteroalkynyl; R5 is aryl; R5 is aralkyl; R5 is aralkenyl; R5 is cycloalkyl; R5 is cycloalkylalkyl; R5 is haloalkyl; R5 is haloalkoxy; R5 is heterocyclyl; R5 is heterocyclylalkyi; R6 is hydrogen; R6 is alkyl; R6 is alkenyl; R6 is cycloalkyl; R6 is cycloalkylalkyl; R6 is aralkyl; R6 is aryl; R7 is hydrogen; R7 is alkyl; R7 is aralkyl. In one aspect of the invention, the compound is in the form of a salt, particularly a pharmaceutically acceptable salt. In one aspect, the salt is the combination of the anionic form of -C(=O)-W where W is ionized -OH (i.e., -O"), and a pharmaceutically acceptable cation. In various embodiments of this aspect of the invention, where these embodiments are exemplary only: Y is -S-; Y is -S(O)-; Y is - S(O)2-;Y is -S(O)m-; Y is -O-; W is -OR4, W is -N(R5)2; W is -NHN(R5)2; p is 0; p is 1 ; p is 2; p is 3; p is 4; p is 5; q is 0; q is 1; q is 2; m is 0; m is 1; m is 2; n is 0; n is 1 ; n is 2; n is 3; n is 4; n is 5; R1 is alkyl; R1 is alkenyl; R1 is alkynyl; R1 is heteroalkyl; R1 is heteroalkenyl; R1 is heteroalkynyl; R1 is aryl; R1 is aralkyl; R1 is aralkenyl; R1 is cycloalkyl; R1 is cycloalkylalkyl; R1 is halo; R1 is haloalkyl; R1 is haloalkoxy; R1 is nitro; R1 is cyano; R1 is -NHOH; R1 is -OR7; R1 is -SR7; R1 is -C(O)OR7; R1 is -OC(O)R7; R1 is -C(O)N(R6)2; R1 is -C(S)R6; R1 is -C(O)R6; R1 is -N(R6)2; R1 is -N(R6)C(O)R6; R1 is -N(R6)C(O)OR7; R1 is -S(O)tR6 (where t is 0 to 2); R1 is -S(O)tN(R6)2 (where t is 0 to 2); R1 is -OC(S)NR6; R1 is -NR6C(S)OR7; R1 is -NR6S(O)tR6 (where t is 0 to 2); R1 is heterocyclyl; R1 is heterocyclylalkyi; R2 is alkyl; R2 is alkenyl; R2 is alkynyl; R2 is heteroalkyl; R2 is heteroalkenyl; R2 is heteroalkynyl; R2 is aryl; R2 is aralkyl; R2 is aralkenyl; R2 is cycloalkyl; R2 is cycloalkylalkyl; R2 is halo; R2 is haloalkyl; R2 is haloalkoxy; R2 is nitro; R2 is cyano; R2 is -NHOH; R2 is -OR7; R2 is -SR7; R2 is -C(O)OR7; R2 is -OC(O)R7; R2 is -C(O)N(R6)2; R2 is -C(S)R6; R2 is -C(O)R6; R2 is -N(R6)2; R2 is -N(R6)C(O)R6; R2 is -N(R6)C(O)OR7; R2 is -S(O)tR6 (where t is 0 to 2); R2 is -S(O)tN(R6)2 (where t is 0 to 2); R2 is -OC(S)NR6; R2 is -NR6C(S)OR7; R2 is -NR6S(O)tR6 (where t is 0 to 2); R2 is heterocyclyl; R2 is heterocyclylalkyi; R3 is hydrogen; R3 is alkyl; R3 is alkenyl; R3 is aryl; R3 is aralkyl; R3 is aralkenyl; R3 is cycloalkyl; R3 is cycloalkylalkyl; R3 is halo; R3 is haloalkyl; R3 is haloalkoxy; R3 is nitro; R3 is cyano; R3 is -NHOH; R3 is -OR7; R3 is -SR7; R3 is -C(O)OR7; R3 is -OC(O)R7; R3 is -C(O)N(R6)2; R3 is -C(S)R6; R3 is -C(O)R6; R3 is -N(R6)2; R3 is -N(R6)C(O)R6; R3 is -N(R6)C(O)OR7; R3 is -OC(S)NR6; R3 is -NR6C(S)OR7; R3 is -OR7; R3 is -C(O)OR7; R3 is -OC(O)R7; R3 is -C(O)N(R6)2; R3 is heterocyclyl; R3 is heterocyclylalkyi; R4 is hydrogen; R4 is alkyl; R4 is alkenyl; R4 is alkynyl; R4 is heteroalkyl; R4 is heteroalkenyl; R4 is heteroalkynyl; R4 is aryl; R4 is aralkyl; R4 is aralkenyl; R4 is cycloalkyl; R4 is cycloalkylalkyl; R4 is haloalkyl; R4 is haloalkoxy; R4 is heterocyclyl; R4 is heterocyclylalkyi; R5 is hydrogen; R5 is alkyl; R5 is alkenyl; R5 is alkynyl; R5 is heteroalkyl; R5 is heteroalkenyl; R5 is heteroalkynyl; R5 is aryl; R5 is aralkyl; R5 is aralkenyl; R5 is cycloalkyl; R5 is cycloalkylalkyl; R5 is haloalkyl; R5 is haloalkoxy; R5 is heterocyclyl; R5 is heterocyclylalkyi; R6 is hydrogen; R6 is alkyl; R6 is alkenyl; R6 is cycloalkyl; R6 is cycloalkylalkyl; R6 is aralkyl; R6 is aryl; R7 is hydrogen; R7 is alkyl; R7 is aralkyl.
The terms (R1)p- and (R2)q- are utilized herein to indicate that a number "p" of R groups are bonded to the carbocyclic aromatic ring of the compound, and a number "q" of R2 groups are bonded to the heterocydic aromatic ring of the compound. When p is zero, then there are no R1 groups present on the compound, and the carbocyclic aromatic ring is unsubstituted phenyl. Likewise, when q is zero, then there are no R2 groups present on the compound. However, when p is greater than zero, then "p" R1 groups are bonded to the carbon atoms of the carbocyclic aromatic ring of the compound, and likewise when q is greater than zero, then "q" R2 groups are bonded to the carbon atoms of the heterocydic ring of the compound. In each case when an R1 or R2 group is present in the compound, the R1 and/or R2 group replaces a hydrogen atom that would otherwise be bonded to the ring carbon.
The compounds of formula (1) are described, in part, by the presence of various groups, e.g., Y, W, R1, R2, R3, etc., and various integers, e.g., m, n, p, q, etc. The term "independently at each occurrence" in connection with a description of the compound and the various groups and integers thereof is intended to indicate that the selection of the identity for a particular group or integer is independent of the selection of the identity of any other group or integer. Furthermore the selection of any one group at one instance is independent of the selection of the same group at another instance (which will arise when a group, e.g., R1, appears more than once in the compound). Furthermore, the selection of any one integer (e.g., t) at one occurrence in the compound is entirely independent of the selection of the same integer if and when it occurs an additional time in the compound.
For example, in various aspects of the invention, the compound of formula (1) is characterized by the requirements that (where distinct requirements are separated by semicolons, and any two or more requirements may be combined): Y is S; Y is O; W is -OR4; W is -OH; W is -N(R5)2; R3 is -H; m is 0; n is 1 ; Y is S and W is -OR4; an "R" group (e.g., R1, R2, R3, etc.) is a hydrocarbon, i.e., formed entirely of carbon and hydrogen; R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl; an "R" group (or any one or more members that comprise the R group) has a specified maximum number of carbons, where in various aspects of the invention this maximum number is 20, 15, 10; R1 has 1- 10 carbons; an "R" group (or any one or more members thereof) has a specified maximum number of non-hydrogen atoms, where in various aspects of the invention this maximum number is 20, 15, 10; R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl, where optionally the the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 non- hydrogen atoms; R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2); R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi; R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl; R2 has 1-10 carbons; R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl; R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 non-hydrogen atoms; R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano; R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(Re)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2); R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi; R3 is selected from hydrogen, lower alkyl and lower haloalkyl; an "R" group has a specified maximum formula weight, where in various aspects of the invention that maximum formula weight is 500, or 400, or 300, or 200; Y is selected from the group consisting of -S- and -O-, and W is selected from the group consisting of -OR4 and -N(R5)2, and p is 1 to 4, and q is 1 to 2, and n is 1 to 5, and R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2l -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6,
-N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi, and R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), and each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2, and R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi, and R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi, and R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl, and R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl.
The compounds as set forth above, including any express requirements or express limitations, and any combinations thereof, may be present in a composition of the present invention as described below, and may be used in any of the methods of the present invention as described below. In other words, in describing a composition of the present invention that contains a compound of formula (1), the compound of formula (1) may be described in terms of any one or more the express requirements and/or express limitations set forth herein. Likewise with descriptions of methods of the present invention.
C. Compound Synthesis
Compounds of formula (1) may be prepared according to methods known to one skilled in the art, or by the methods similar to those disclosed in US 3,814,761 , US 4,559,345 and Gaetano d'Atri, et. al., J. Med. Chem., (1984) 27, 1621-1629, all of which are incorporated in full by reference herein, or by methods similar to the method described below.
It is understood that in the following description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.
It will also be appreciated by those skilled in the art that in the process described below the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., f-butyldimethylsilyl, f-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(O)-R (where R is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or aralkyl esters.
Protecting groups may be added or removed in accordance with standard techniques, which are well-known to those skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and
P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley-lnterscience. The protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl chloride resin.
It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of formula (1), as described above in the Summary of the Invention, may not possess pharmacological activity as such, they may be administered to a mammal in need of treatment according to the present invention and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of formula (1 ) are included within the scope of the invention. The following Reaction Schemes illustrate methods to make compounds of formula (1). It is understood that one of ordinary skill in the art would be able to make the compounds of formula (1) by similar methods or by methods known to one skilled in the art. In general, starting components may be obtained from sources such as Aldrich, or synthesized according to sources known to those of ordinary skill in the art (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th Ed., 2000 (Wiley Interscience, New York)). Moreover, the various R groups (e.g., R-i, R2, and R3, etc.) of the compounds of formula (1) are selected from components as indicated in the description of formula (1), and may be attached to starting components, intermediate components, and/or final products according to schemes known to those of ordinary skill in the art. R1, R2, and R3 are defined as above. X is Cl or Br. R' is an alkyl or an aryl group.
Compounds of formula (1) may be prepared according to the Scheme 1 depicted below.
Scheme 1
Figure imgf000033_0001
(a) (b) (c)
Figure imgf000033_0002
( )
Figure imgf000033_0003
(f)
Figure imgf000033_0004
(g) (1)
In general, starting material of formula (a) reacts with a halogenated compound of formula (b) where X represents a leaving group, in the presence of a base such as NaHCO3 at room temperature to afford the compound of formula (c).
Componds of formulae (a) and (b) are very well known in the chemical literature. The conversion of the hydroxyl group to a Cl or Br group is accomplished by treating the compound of formula (c) with an agent such as POCI3, POBr3, PCI5 or PBr5 under reflux to afford the halogenated compound of formula (d). The compound of formula (d) then reacts with a compound of formula (e) such as phenol or benzenethiol which is treated with a base such as NaOH to afford the compound of the invention having formula (f). Again, compounds of formula (e) are very well known in the chemical literature. The compound of formula (f) can be oxidized by an oxidizing agent such as H2O2 to afford a compound of the invention having formula (g). The transformation of the compound of formula (g) to a compound of formula (1) can readily be achieved by transesterification, saponification and hydrolysis as well as by amidation of the free carboxyl group or the corresponding acid halide.
Alternatively, compounds of formula (1) can be prepared as illustrated in Scheme 2.
Scheme 2
Figure imgf000034_0001
(c") + R'02C-(C(R3)2)nSH-
Figure imgf000034_0002
Figure imgf000034_0003
(rf) (1)
In general, the halogenated starting material of formula (a') is condensed with a compound of formula (b') such as phenol and benzenethiol in an appropriate solvent under reflux. Compounds of formula (a') and formula (b') are well known in the chemical literature. This reaction yields the compound of formula (c') which is then transformed to a compound of formula (e') by reacting with a compound of formula (d') in the presence of a base such as Na2CO3 under reflux. Compounds of formula (d') are well known in the chemical literature. The product is isolated and filtered, and then oxidized by an oxidizing agent such as H2O2 to afford the compound of formula (g'). The transformation of the compound of formula (g') to a compound of formula (1) can readily be achieved by transesterification, saponification and hydrolysis as well as by amidation of the free carboxyl group or the corresponding acid halide. Alternatively, compound of formula (1) can be prepared as illustrated in
Scheme 3. P indicates a protection group such as the BOC group or the like. The method of Scheme 3 is particularly suited to prepare compounds wherein R1 is an electron-withdrawing group, such as -NO2, -CF3 and the like.
Scheme 3
Figure imgf000036_0001
(a") (b")
Figure imgf000036_0002
(d")
Figure imgf000036_0003
(e") (f)
Figure imgf000036_0004
(g") (h")
Figure imgf000036_0005
(i")
Figure imgf000036_0006
In general, the amino group of a compound of formula (a") is protected by a protection group such as BOC or the like according to procedures known to one skilled in the art to yield the N-protected product (b"). Compounds of formula (a") are well known in the chemical literature. This N-protected compound is then reacted with a halogenated compound of formula (c") at room temperature in the presence of a base, such as NaHCO3 or the like to afford the compound of formula (d"). Again, compounds of formula (c") are well known in the chemical literature. Treatment of the compound of formula (d") with weak acid such as trifluoroacetic acid can be used to remove the protection group and thereby obtain the amino product of formula (e"). Compound of formula (e") is then converted into the compound of formula (f) by diazotizing the amino group with a reagent such as NaNO2 and then treating with xanthate or water to afford the corresponding thiol or the hydroxy product of formula (f") which is subsequently treated with a compound of formula (g") in the presence of pyridine or the like under reflux to afford the compound of formula (h"). A compound of formula (h") can then be oxidized by an oxidizing agent such as H2O2 or the like to yield the product of formula (i"). The transformation of the compound of formula (i") to a compound of formula (1) can readily be achieved by transesterification, saponification and hydrolysis as well as by amidation of the free carboxyl group or the corresponding acid halide.
D. Enhanced Penetration of Blood Brain Barrier Compounds that may be useful in vitro or in vivo for inhibiting Aβ production and/or release from cells will typically be more effective in alleviating or preventing Aβ production and/or release in the brain if they can gain access to target cells in the brain. A brain cell is defined herein as any cell residing within the skull bone of the head including the spinal cord. Non-limiting examples of brain cells are neurons, glial cells (astrocytes, oligodendrocytes, microglia), cerebrovascular cells (muscle cells, endothelial cells), blood cells (red, white, platelets, etc.) and cells that comprise the meninges. However, access is restricted due to the blood brain barrier (BBB), a physical and functional blockade which separates the brain parenchyma from the systemic circulation (reviewed in Pardridge et al., J Neurovirol 5(6):556-569, 1999; Rubin and Staddon, Rev. Neurosci 22Λ 1-28, 1999). Circulating molecules are normally able to gain access to brain cells via one of two processes: (i) lipid-mediated transport of small molecules through the BBB by free diffusion, or (ii) catalyzed transport. Thus, compounds that are useful for inhibiting Aβ production and/or release are preferably linked to agents that will facilitate penetration of the blood brain barrier. In one embodiment, the method of the present invention will employ a naturally occurring polyamine linked to a small molecule useful at inhibiting Aβ production and/or release. Natural cell metabolites that may be used as linkers, include, but are not limited to, putrescine (PUT), spermidine (SPD), spermine (SPM), or DHA. An alternative method to deliver a compound across the BBB is by intracerebroventricular pump.
The neurologic agent may also be delivered to the nasal cavity. It is preferred that the agent be delivered to the olfactory area in the upper third of the nasal cavity and particularly to the olfactory epithelium in order to promote transport of the agent into the peripheral olfactory neurons rather that the capillaries within the respiratory epithelium. In a preferred embodiment the transport of neurologic agents to the brain is accomplished by means of the nervous system instead of the circulatory system so that small molecules which inhibit Aβ production and/or release may be delivered to the appropriate areas of the brain. It is preferable that the neurologic agent be capable of at least partially dissolving in the fluids that are secreted by the mucous membrane that surround the cilia of the olfactory receptor cells of the olfactory epithelium in order to be absorbed into the olfactory neurons. Alternatively, the agent may be combined with a carrier and/or other substances that foster dissolution of the agent within nasal releases. Potential adjuvants include GM-1 , phosphatidylserine (PS), and emulsifiers such as polysorbate 80.
To further facilitate the transport of the neurologic agent into the olfactory system, the method of the present invention may combine the agent with substances that enhance the absorption of the agent through the olfactory epithelium. It is preferred that the additives promote the absorption of the agent into the peripheral olfactory receptor cells. Because of their role in odor detection, these peripheral neurons provide a direct connection between the brain and the outside environment.
The olfactory receptor cells are bipolar neurons with swellings covered by hair-like cilia which project into the nasal cavity. At the other end, axons from these cells collect into aggregates and enter the cranial cavity at the roof of the nose. It is preferred that the neurologic agent is lipophilic in order to promote absorption into the olfactory neurons and through the olfactory epithelium. Among those neurologic agents that are lipophilic are gangliosides and phosphatidylserine (PS). Alternatively, the neurologic agent may be combined with a carrier and/or other substances that enhance the absorption of the agent into the olfactory neurons. Among the supplementary substances that are preferred are lipophilic substances such as gangliosides and phosphatidylserine (PS). Uptake of non-lipophilic neurologic agents such as nerve growth factor (NGF) may be enhanced by the combination with a lipophilic substance. In one embodiment of the method of the invention, the neurologic agent may be combined with micelles comprised of lipophilic substances. Such micelles may modify the permeability of the nasal membrane and enhance absorption of the agent. Among the lipophilic micelles that are preferred are gangliosides, particularly GM-1 ganglioside, and phosphatidylserine (PS). The neurologic agent may be combined with one or several types of micelle substances.
Once the agent has crossed the nasal epithelium, the invention further provides for transport of the neurologic agent along the olfactory neural pathway. The agent may be combined with substances that possess neurotrophic or neuritogenic properties which, in turn, may assist in transporting the agent to sites of nerve cell damage. Prophylactic therapies may apply the agent alone or in combination with a carrier, other agents, and/or other substances that may enhance the absorption of the agent into the olfactory neurons.
To deliver the agent to the olfactory neurons, the agent alone or in combination with other substances as a pharmaceutical composition may be administered to the olfactory area located in the upper third of the nasal cavity. The composition may be dispensed intranasally as a powdered or liquid nasal spray, nose drops, a gel or ointment, through a tube or catheter, by syringe, by packtail, by pledget, or by submucosal infusion. Other modifications of the compounds described herein in order to enhance penetration of the blood brain barrier can be accomplished using methods and derivatives known in the art, including but not limited to those disclosed in the following patent publications, each of which is incorporated by reference herein:
U.S. Patent No. 6,024,977, issued February 15, 2000 to Yatvin, discloses covalent polar lipid conjugates for targeting to brain and central nervous system.
U.S. Pat. No. 5,017,566, issued May 21 , 1991 to Bodor discloses β and γ cyclodextrin derivatives comprising inclusion complexes of lipoidal forms of dihydropyridine redox targeting moieties.
U.S. Pat. No. 5,023,252, issued Jun. 11 , 1991 to Hseih discloses the use of pharmaceutical compositions comprising a neurologically active drug and a compound for facilitating transport of the drug across the blood-brain barrier including a macrocyclic ester, diester, amide, diamide, amidine, diamidine, thioester, dithioester, thioamide, ketone or lactone.
U.S. Pat. No. 5,024,998, issued Jun. 18, 1991 to Bodor discloses parenteral solutions of aqueous-insoluble drugs with β andγ cyclodextrin derivatives.
U.S. Pat. No. 5,039,794, issued Aug. 13, 1991 to Wier et al. discloses the use of a metastatic tumor-derived egress factor for facilitating the transport of compounds across the blood-brain barrier.
U.S. Pat. No. 5,112,863, issued May 12, 1992 to Hashimoto et al. discloses the use of N-acyl amino acid derivatives as antipsychotic drugs for delivery across the blood-brain barrier.
U.S. Pat. No. 5,124,146, issued Jun. 23, 1992 to Neuwelt discloses a method for delivery of therapeutic agents across the blood-brain barrier at sites of increased permeability associated with brain lesions. U.S. Pat. No. 5,153,179, issued Oct. 6, 1992 to Eibl discloses acylated glycerol and derivatives for use in a medicament for improved penetration of cell membranes.
U.S. Pat. No. 5,177,064, issued Jan. 5, 1993 to Bodor discloses the use of lipoidal phosphonate derivatives of nudeoside antiviral agents for delivery across the blood-brain barrier.
U.S. Pat. No. 5,254,342, issued Oct. 19, 1993 to Shen et al. discloses receptor-mediated transcytosis of the blood-brain barrier using the transferrin receptor in combination with pharmaceutical compounds that enhance or accelerate this process.
U.S. Pat. No. 5,258,402, issued Nov. 2, 1993 to Maryanoff discloses treatment of epilepsy with imidate derivatives of anticonvulsive sulfamate.
U.S. Pat. No. 5,270,312, issued Dec. 14, 1993 to Glase et al. discloses substituted piperazines as central nervous system agents. U.S. Pat. No. 5,284,876, issued Feb. 8, 1994 to Shashoua et al., discloses fatty acid conjugates of dopamine drugs.
U.S. Pat. No. 5,389,623, issued Feb. 14, 1995 to Bodor discloses the use of lipoidal dihydropyridine derivatives of anti-inflammatory steroids or steroid sex hormones for delivery across the blood-brain barrier. U.S. Pat. No. 5,405,834, issued Apr. 11 , 1995 to Bundgaard et al. discloses prodrug derivatives of thyrotropin releasing hormone.
U.S. Pat. No. 5,413,996, issued May 9, 1995 to Bodor discloses acyloxyalkyl phosphonate conjugates of neurologically-active drugs for anionic sequestration of such drugs in brain tissue. U.S. Pat. No. 5,434,137, issued Jul. 18, 1995 to Black discloses methods for the selective opening of abnormal brain tissue capillaries using bradykinin infused into the carotid artery.
U.S. Pat. No. 5,442,043, issued Aug. 15, 1995 to Fukuta et al. discloses a peptide conjugate between a peptide having a biological activity and incapable of crossing the blood-brain barrier and a peptide which exhibits no biological activity and is capable of passing the blood-brain barrier by receptor-mediated endocytosis.
U.S. Pat. No. 5,466,683, issued Nov. 14, 1995 to Sterling et al. discloses water soluble analogues of an anticonvulsant for the treatment of epilepsy. U.S. Pat. No. 5,525,727, issued Jun. 11, 1996 to Bodor discloses compositions for differential uptake and retention in brain tissue comprising a conjugate of a narcotic analgesic and agonists and antagonists thereof with a lipoidal form of dihydropyridine that forms a redox salt upon uptake across the blood-brain barrier that prevents partitioning back to the systemic circulation. International Pat. Application Publication Number WO85/02342, published
Jun. 6, 1985 for Max-Planck Institute discloses a drug composition comprising a glycerolipid or derivative thereof.
International Patent Application Publication Number WO089/11299, published Nov. 30, 1989 for State of Oregon discloses a chemical conjugate of an antibody with an enzyme which is delivered specifically to a brain lesion site for activating a separately-administered neurologically-active prodrug.
International Patent Application Publication Number WO91/04014, published Apr. 4, 1991 for Synergen, Inc. discloses methods for delivering therapeutic and diagnostic agents across the blood-brain barrier by encapsulating the drugs in liposomes targeted to brain tissue using transport-specific receptor ligands or antibodies.
International Patent Application Publication Number WO91/04745, published Apr. 18, 1991 for Athena Neurosciences, Inc. discloses transport across the blood-brain barrier using cell adhesion molecules and fragments thereof to increase the permeability of tight junctions in vascular endothelium.
International Patent Application Publication Number WO91/14438, published Oct. 3, 1991 for Columbia University discloses the use of a modified, chimeric monoclonal antibody for facilitating transport of substances across the blood-brain barrier. International Pat. Application Publication Number WO94/01131 , published Jan. 20, 1994 for Eukarion, Inc. discloses lipidized proteins, including antibodies.
International Pat. Application Publication Number WO94/03424, published Feb. 17, 1994 for Ishikira et al. discloses the use of amino acid derivatives as drug conjugates for facilitating transport across the blood-brain barrier.
International Patent Application Publication Number WO94/06450, published Mar. 31 , 1994 for the University of Florida discloses conjugates of neurologically-active drugs with a dihydropyridine-type redox targeting moiety and comprising an amino acid linkage and an aliphatic residue. International Patent Application Publication Number WO94/02178, published Feb. 3, 1994 for the U.S. Government, Department of Health and Human Services discloses antibody-targeted liposomes for delivery across the blood-brain barrier.
International Patent Application Publication Number WO95/07092, published Mar. 16, 1995 for the University of Medicine and Dentistry of New Jersey discloses the use of drug-growth factor conjugates for delivering drugs across the blood-brain barrier.
International Patent Application Publication Number WO96/00537, published Jan. 11 , 1996 for Southern Research Institute discloses polymeric microspheres as injectable drug-delivery vehicles for delivering bioactive agents to sites within the central nervous system.
International Patent Application Publication Number WO96/04001 , published Feb. 15, 1996 for Molecular/Structural Biotechnologies, Inc. discloses omega- 3-fatty acid conjugates of neurologically-active drugs for brain tissue delivery. International Patent Application Publication Number WO96/22303, published Jul. 25, 1996 for the Commonwealth Scientific and Industrial Research Organization discloses fatty acid and glycerolipid conjugates of neurologically-active drugs for brain tissue delivery. In general, it is well within the ordinary skill in the art to prepare an ester, amide or hydrazide derivative from the corresponding carboxylic acid and a suitable reagent. For instance, a carboxylic acid-containing compound, or a reactive equivalent thereof, may be reacted with a hydroxyl-containing compound, or a reactive equivalent thereof, so as to provide the corresponding ester. The following reference books and treatise provide exemplary reaction conditions to achieve such conversions: "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandier et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocydic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 5th Ed., Wiley-lnterscience, New York, 2000.
One of skill in the art can readily modify any of the compounds discussed above and test them for the desired activity and ability to penetrate the blood brain barrier. For example, the compound of formula (1) can be modified to improve its blood-brain-barrier penetration by conjugation to an organic moiety known to cross the blood-brain-barrier, e.g., docosahexaenoic acid (DHA) and the like. The conjugation of DHA to the compound of formula (1) can be achieved by following the literature reported procedure. The following references are listed as the examples: Bradley et. al., J. Controlled Release, 74, 233-236, 2001; Katz et. al., US5,716,614; Bradley et. al., US5,955,459; Shashoua et. al., US5,795,909; Shashoua, US6,225,444 and US 6,258,836. The conjugation of the compound of formula (1) can be via a hydroxy or an amino group or other function groups which can form a covalent bond with DHA or the like. Scheme 4 is one of the examples for the conjugation of the compound formula (1) to a DHA molecule via an amino group on the phenyl ring. Scheme 4
DHACOH
Figure imgf000045_0002
Figure imgf000045_0001
(1a)
In general, the compound of formula (1a) can be prepared by standard amide formation known to those skilled in the art. The carboxyl group can be converted to an active ester or to an acid chloride or to an anhydride and then the intermediate reacts with the compound of formula (1) containing an amino group.
Transcytosis, including receptor-mediated transport of compositions across the blood brain barrier, is also suitable for the compounds of the invention. Transferrin receptor-mediated delivery is disclosed in U.S. Patents Nos. 5,672,683; 5,383,988; 5,527,527; 5,977,307; and 6,015,555. Transferrin-mediated transport is also disclosed in Friden, P.M. et al., Pharmacol. Exp. Ther. 278:1491-1498, 1996; and Lee, H.J., J. Pharmacol. Exp. Ther. 292:1048-1052, 2000. EGF receptor-mediated delivery is disclosed in Deguchi, Y. et al., Bioconjug. Chem. 10:32-37, 1999, and transcytosis is described in Cerletti, A. et al., J. Drug Target. 8:435-446, 2000. The use of insulin fragments as carriers for delivery across the bloodbrain barrier is discussed by Fukuta, M. et al., Pharm. Res. 11 :1681-1688, 1994. Delivery of compounds via a conjugate of neutral avidin and cationized human albumin is described by Kang, Y.S. et al., Pharm. Res. 1 :1257-1264, 1994.
Although BBB penetration of a therapeutic compound may be desired, recent evidence suggests that BBB penetrable compounds may not necessarily be required to decrease CNS β-amyloid levels. Shibata et al (J Clin Invest QS: 1489- 1499, 2000) demonstrate that CSF Aβ can be transported across the BBB into the systemic circulation, thereby decreasing Aβ in the CNS. Once in the systemic circulation, Aβ interacts with binding proteins such as ApoJ/ApoE, which results in a decrease in "free" Aβ in the circulation and shifts the equilibrium to facilitate further transport of Aβ out of the CNS. Thus, the systemic circulation may act as a "sink" or pool of Aβ that can regulate CNS β-amyloid levels (Shibata, M et al., J Clin Invest 106: 1489-1499, 2000). This "peripheral sink" hypothesis is supported by vaccination studies with anti-Aβ antibodies in AD transgenic mouse models. For example, vaccination of PDAPP mice with an Aβ antibody (m266) resulted in accumulation of CNS derived Aβ in the plasma (DeMattos et al., PNAS 98: 8850-8855, 1998; Holtzman et al., Adv Drug Delivery Rev 54: 1603-1613, 2002). Therefore, if compounds can systemically decrease Aβ levels, the peripheral sink hypothesis indicates that this may shift the Aβ equilibrium between the CNS and plasma resulting in a decreased β-amyloid burden in the CNS. Therefore, pharmaceutical agents of the invention can act systemically and may not be required to cross the BBB.
Nevertheless, in one aspect of the invention, a compound of formula (1) is conjugated to another compound in order to provide an agent, where the agent has enhanced ability to cross the BBB relative to the compound of formula (1). Methods of conjugating a biologically active agent to a compound, and suitable compounds that upon conjugation to a biologically active agent provide a conjugate having enhanced ability to cross the BBB, are well known from the above-cited references, and these same techniques may be applied to effectively enhance the permeability of compounds of formula (1) to the BBB.
E. Pharmaceutical Compositions and Administration The compounds of this invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds of the present invention can be formulated into pharmaceutical compositions by combination with appropriate pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. As such, administration of the compounds can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intracheal, efc., administration. The active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.
In pharmaceutical dosage forms, the compounds may be administered in the form of their pharmaceutically acceptable salts. They may also be used in appropriate association with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.
For oral preparations, the compounds can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
The compounds can be formulated into preparations for injections by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
The compounds can be utilized in aerosol formulation to be administered via inhalation. The compounds of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoro-methane, propane, nitrogen and the like.
Furthermore, the compounds can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds of the present invention can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at ambient temperature.
Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more compounds of the present invention. Similarly, unit dosage forms for injection or intravenous administration may comprise the compound of the present invention in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier. Implants for sustained release formulations are well known in the art.
Implants are formulated as microspheres, slabs, etc. with biodegradable or non- biodegradable polymers. For example, polymers of lactic acid and/or glycolic acid form an erodible polymer that is well tolerated by the host. The implant containing the inhibitory compounds is placed in proximity to the site of the tumor, so that the local concentration of active agent is increased relative to the rest of the body.
The term "unit dosage form", as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for the novel unit dosage forms of the present invention depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host. The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
The combined use of the provided inhibitory compounds and other cytotoxic agents has the advantages that the required dosages for the individual drugs is lower, and the effect of the different drugs complementary. Depending on the patient and condition being treated and on the administration route, the subject inhibitory compounds may be administered in dosages of 0.1 μg to 40 mg/kg body weight per day. The range is broad, since in general the efficacy of a therapeutic effect for different mammals varies widely with doses typically being 20, 30 or even 40 times smaller (per unit body weight) in man than in the rat. Similarly the mode of administration can have a large effect on dosage. Thus for example oral dosages in the rat may be ten times the injection dose. Higher doses may be used for localized routes of delivery.
A typical dosage may be a solution suitable for intravenous administration; a tablet taken from two to six times daily, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient, etc. The time-release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release. Those of skill will readily appreciate that dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Some of the specific compounds are more potent than others. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given compound.
For use in the subject methods, the subject compounds may be formulated with other pharmaceutically active agents known to one of ordinary skill in the art.
F. Methods of Use The compounds and pharmaceutical compositions of the invention are administered to a subject having a pathology associated with increased accumulation or deposition of the β-amyloid peptide such as but not limited to Alzheimer's disease. The present compounds are useful for prophylactic or therapeutic purposes. The prevention of Aβ accumulation and deposition is accomplished by administration of a compound of formula (1) prior to development of overt disease, e.g., to prevent β-amyloid production, release and/or accumulation in the form of plaques, etc. Alternatively the compounds are used to treat ongoing disease, by stabilizing or improving the clinical symptoms of the patient.
The host, or patient, may be from any mammalian species, e.g., primate sp., particularly humans; rodents, including mice, rats and hamsters; guinea pigs; rabbits; equines, bovines, canines, felines; efc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease. One method to identify a subject in need of treatment according to the present invention is to measure cognitive, behavioural and/or memory abilities of the subject. If a subject displays impairment in cognitive functioning, particularly if the subject's cognitive ability declines over time, then the subject may benefit from treatment according to the present invention. If the subject is a human, then cognitive function and impairment indicative of probable Alzheimer's disease can be assessed using psychological and other tests known to those skilled in the art. If the human subject displays characteristics consistent with a disease caused by increased accumulation and/or deposition of the β-amyloid peptide, such as but not limited to Alzheimer's disease, then the subject may benefit from the treatment according to the present invention.
The susceptibility of a particular cell to treatment with the subject compounds may be determined by in vitro testing. Typically a culture of the cell is combined with a compound of formula (1) at varying concentrations for a period of time sufficient to allow the active agents to decrease production and/or release of Aβ, usually between about one hour and one week. For in vitro testing, cultured cells from a biopsy sample may be used.
The dose will vary depending on the specific compound utilized, specific disorder, patient status, etc. Typically a therapeutic dose will be sufficient to produce a substantial decrease β-amyloid production and/or release in the targeted tissue, while maintaining patient viability. Treatment will generally be continued until there is a substantial reduction, e.g., at least 10%, in β-amyloid levels and may be continued chronically.
Thus, in one aspect, the present invention provides a method for modulating the production and/or release of β-amyloid in a cell, comprising treating said cell with a compound of formula (1). In optional embodiments: the cell is a brain cell; and/or the β-amyloid is β-amyloid 42; and/or β-amyloid production and/or release in the cell is reduced; and/or the cell is treated in vitro.
In another aspect, the present invention provides a method of treatment comprising modulating the production and/or release of β-amyloid in a non-human mammal in need of said treatment, said method comprising administering to said non- human mammal an effective amount of a compound of formula (1). In optional embodiments: the cell is a brain cell; and/or β-amyloid is β-amyloid 42; and/or the non- human mammal is a mouse, rat, cat, dog or guinea pig; and/or βamyloid production and/or release is reduced.
In another aspect, the present invention provides a method of treatment wherein the production and/or release of β-amyloid is modulated in a human in need of said treatment, said method comprising administering to said human an effective amount of a compound of formula (1). In optional embodiments: the human is afflicted with Alzheimer's disease; and/or the human has suffered a head injury; and/or the human has a genetic predisposition or environment exposure that increases the likelihood that said person will develop Alzheimer's disease; and/or the human exhibits minimal cognitive impairment suggestive of early stage Alzheimer's disease; and/or the production and/or release of the β-amyloid is a brain cell is modulated; and/or the β- amyloid is β-amyloid 42; and/or β-amyloid production and/or release is reduced. As mentioned above, the method of the present invention may preferentially reduce production and/or release of Aβ42 relative to one or more other forms of Aβ, in a target that produces and/or releases Aβ42, for instance a target selected from a cell, a human, a non-human mammal, and the brain of a human. Tests to identify the selectively of such a compound are disclosed herein. Thus, in one aspect, the present invention provides that a subject in need to selective reduction of Aβ42 relative to one or more other forms of Aβ, is administered a compound of formu-a (1) that affords such selectively. The present invention will now be illustrated with the reference to the following non-limiting examples.
EXAMPLES
PREPARATION 1 SYNTHESIS OF (4,6-DICHLOROPYRIMIDIN-2-YLSULFANYL)ACETIC ACID ETHYL ESTER
To a solution of NaHCO3 (8.4 g, 0.1 mol) in 500 mL of water was added 2- thiobarbituric acid (14.4 g, 0.1 mol) with stirring. Ethyl bromoacetate (11.1 mL, 0.1 mol) was then added, followed by the addition of 400 mL of ethanol (EtOH) to obtain a clear solution. This mixture was kept stirring at room temperature for 3 hours and then the solvent was removed in vacuum and a precipitate was formed. The solid was collected by filtration, washed with the mother liquor, and then dried in vacuo over P2O5 for 3 days to yield 17.9 g (78%) of the white solid product which was used in the next step without further purification.
To a mixture of the white solid obtained above (17.9 g, 77.7 mmol) in 120 mL of POCI3 was slowly added Λ/,/V-diethylaniline (11.6 g, 77.7 mmol) at 5°C over 10 minutes. The mixture was stirred at 10-15°C for 15 minutes and then refluxed for 5 hours. The excess POCI3 was removed in vacuo. The residue was treated with cold water (500 mL) and the mixture was stirred for 3 days and then filtered. The solid collected was recrystallized from hexanes yielding 6.73 g (32%) of the product which was used as described below without further purification. PREPARATION 2
SYNTHESIS OF 2,3-DIMETHYLBENZENETHIOL
To a stirred mixture of 2,3-dimethylaniline (15.0 g, 0.12 mol) and crushed ice (50 mL) was added 23 mL of HCI (cone.) dropwise over 45 minutes, followed by the addition of NaNO2 (9.0 g, 0.13 mol) in 15 mL of water over 15 minutes. The reaction mixture was kept at 0°C for 25 minutes. The cold diazonium solution was added dropwise to a solution of potassium ethyl xanthate (30 g, 0.13 mol) in 40 mL of water. This mixture was then warmed to 40°C and kept at this temperature for 45 minutes. After cooling down to room temperature, the reaction mixture was extracted with Et2O (3X100 mL). The combined organic extracts were washed with 25 mL of 10% NaHCO3 solution, water (2X50 mL) and then dried over Na2SO . The solution was filtered and the solvent was removed. The crude product was used in the next step reaction without further purification.
To a slurry of LiAIH4 in Et2O (60 mL) was added dropwise the solution of the product obtained above (6.52 g, 28.8 mmol) in 40 mL of Et2O at such a rate that the ether refluxed gently without external cooling. The mixture was kept stirring at room temperature for about 3.5 hours, followed by the slow addition of 10 mL of water. Stirring was kept for another 10 minutes and then 90 mL of 20% HCI solution and 120 mL of ice-cold water were added sequentially. The aqueous layer was extracted with 100 mL of Et2O and the combined organic layers were washed with water (2X50 mL) and dried over Na2SO4. The solvent was removed and the residue was purified by flash column chromatography (eluted with hexanes:Et2O = 4:1) to afford the product which was used as described below without further purification.
EXAMPLE 1
SYNTHESIS OF [4-CHLORO-6-(2,4-DIMETHYLPHENYLSULFANYL)PYRIMIDIN-2-YLSULFANYL]-
ACETIC ACID (COMPOUND 1)
Sodium hydride (0.16 g, 4 mmol) was added to a solution of 2,4- dimethylbenzenethiol (0.55 g, 4 mmol) in 10 mL of dimethyl formamide (DMF) in portions. This mixture was then added to a DMF (10 mL) solution of (4,6- dichloropyrimidin-2-ylsulfanyl)-acetic acid ethyl ester (1.34 g, 5 mmol) dropwise at room temperature over about 2 hours. The resulting mixture was stirred at room temperature for 2 days. The solvent was removed in vacuo and the residue was purified by flash column chromatography eluted with hexanes:Et2O = 19:1. The product was obtained in 42%o yield (0.78 g) and used in the next reaction without further purification.
The product obtained above (0.67 g, 1.82 mmol) was dissolved in 20 mL of iso-propanol and the solution was heated to reflux. NaOH solution (1 N, 1.82 mL) was added to the hot solution and the mixture was kept under reflux for 5 minutes and then cooled to room temperature. The solvent was removed in vacuo. The residue was dissolved in 30 mL of water and then washed with diethylether (Et2O). The aqueous layer was acidified with 6 N HCI (to pH ~1) and then extracted with methylene chloride (CH2CI2, 3 times). The organic layers were combined and dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography eluted with CH2CI2:MeOH = 99:1.5 to yield a white product (0.57 g, 92%). H NMR (ppm, CDCI3): 2.36 (s, 3H), 2.41 (s, 3H), 3.83 (s, 2H), 6.41 (s, 1 H), 7.10-7.12 (m. 1 H), 7.23 (m, 1H), 7.41-7.43 (m, 1 H). MS (m/z, ES+): 341.0 (100%, M+1).
EXAMPLE 2
SYNTHESIS OF [4-CHLORO-6-(2,3-DIMETHYLPHENOXY)PYRIMIDIN-2-YLSULFANYL]ACETIC ACID (COMPOUND 2)
2,3-Dimethylphenol (0.61 g, 5 mmol) was added to a solution of NaOH (0.20 g, 5 mmol) in ethanol (EtOH, 20 mL) which was prepared by refluxing the mixture until NaOH disappeared. The mixture was kept stirring for about 5 minutes. (4,6- Dichloropyrimidin-2-ylsulfanyl)acetic acid ethyl ester (1.34 g, 5 mmol) was then added to the mixture with stirring. The resulting solution was kept at reflux for 18 hours and then concentrated in vacuo. The residue was redissolved in 80 mL of Et2O. The Et2O solution was washed with water twice, then dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The oily residue was purified by flash column chromatography eluted with hexanes:Et2O = 19:1 to afford a white solid in 52%> yield. The product obtained was used in the next step without further purification.
The product obtained above (0.70 g, 1.98 mmol) was dissolved in 25 mL of iso-propanol and the solution was heated to reflux. NaOH solution (1 N, 1.82 mL) was added to the hot solution and the mixture was kept under reflux.for 5 minutes and then cooled to room temperature. The solvent was removed in vacuo. The residue was dissolved in 20 mL of water and then washed with Et2O. The aqueous layer was acidified with 6 N HCI (to pH ~1) and then extracted with CH2CI2 (3x). The organic layers were combined and dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography eluted with CH2CI2:MeOH = 200:1 to yield a white product (0.39 g, 61%). 1H NMR (ppm, CDCI3): 2.03 (s, 3H), 2.32 (s, 3H), 3.73 (s, 2H), 6.53 (s, 1 H), 6.84-6.86 (m. 1H), 7.09-7.14 (m, 2H). MS (m/z, ES+): 324.8 (100%, M+1).
EXAMPLE 3
SYNTHESIS OF [4-CHLORO-6-(2,4-DIMETHYLPHENYLSULFANYL)PYRIMIDIN-2-YLSULFANYL]-
ACETIC ACID
The title compound was synthesized similarly to the procedure described in Example 1. 1H NMR (ppm, DMSO-d6): 2.30 (s, 3H), 2.35 (s, 3H), 3.79 (s, 2H), 6.61 (s, 1 H), 7.20-7.27 (m. 1H), 7.38-7.46 (m, 2H). MS (m/z, ES+): 341.0 (100%, M+1). EXAMPLE 4
EFFECT OF COMPOUND 1 TREATMENT ON β-AMYLOiD PRODUCTION AND/OR RELEASE FROM CELLS
Cell Lines and Pharmacological Treatments 293 EBNA cells (InVitrogen, Carlsbad, CA) stably transfected with
Swedish mutant β-Amyloid Precursor Protein -695 (SM4 cells) were routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10% fetal bovine serum. Cells were seeded into poly-D-Lysine (SIGMA) coated 6-well plates at a density of 5-7 X 105 cells per well. Subsequently, the cells were rinsed in 1 ml of PBS and treated with 50-100 μM Compound 1 for 5 hrs or 50-300 μM Compound 1 for 16 hrs in serum-free/phenol red-free DMEM.
Aβ Detection and Standardization
After the pharmacological treatment, the exposure media was collected and supplemented with 10%) sample treatment buffer (40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide), and assayed for either Aβ40 or Aβ42 by a colorimetric ELISA as per the manufacturer's protocol (Biosource International Inc, California). The cells were lysed in 0.1 %> Triton X-100 in PBS supplemented with 5 μM propridium iodide (Molecular Probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Aβ40 and Aβ42 were standardized against propridium iodide fluorescence as a measure of total cell number.
As seen in Figure 1 , Compound 1 induced a significant decrease in Aβ42 production and/or release from SM-4 cells after 5 and 16 hrs. For example, 100 μM Compound 1 reduced Aβ42 for 92% (p<0.001) relative to the control after 5 hrs. Similarly, a 16 hr treatment at 300 μM resulted in a 98%. decrease in Aβ42 (p<0.001). Furthermore, these concentrations of Compound 1 also resulted in a significant decrease in Aβ40 at 5 and 16 hrs. EXAMPLE 5
EFFECT OF COMPOUND 2 TREATMENT ON -AMYLOID PRODUCTION
AND/OR RELEASE FROM CELLS
Cell Lines and Pharmacological Treatments 293 EBNA cells (InVitrogen, Carlsbad, CA) stably transfected with
Swedish mutant β-Amyloid Precursor Protein -695 (SM4 cells) were routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10% fetal bovine serum. Cells were seeded into poly-D-Lysine (SIGMA) coated 6-well plates at a density of 5-7 X 105 cells per well. Subsequently, the cells were rinsed in 1 ml of PBS and treated with 50-300 μM Compound 2 for 16 hrs in serum-free/phenol red-free DMEM.
Aβ Detection and Standardization.
After the pharmacological treatment, the exposure media was collected and supplemented with 10%) sample treatment buffer (40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% Sodium azide), and assayed for either Aβ40 or Aβ42 by a colorimetric ELISA as per the manufacturer's protocol (Biosource International Inc, California). The cells were lysed in 0.1 % Triton X-100 in PBS supplemented with 5 μM propridium iodide (Molecular Probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Aβ40 and Aβ42 were standardized against propridium iodide fluorescence as a measure of total cell number.
As seen in Figure 2, Compound 2 decreased the mean Aβ42 levels without altering mean Aβ40 suggesting a selective action on Aβ42. EXAMPLE 6
SCREENING AGENTS FOR ABILITY TO DECREASE β-AMYLoiD PRODUCTION
USING AN IN VITRO GAMMA SECRETASE ASSAY
Several assays have been described in the literature which measure the formation of various Aβ species using an in vitro γ-secretase assay (Tian et al., 8th Intl Conference on Alzheimer's Disease and Related Disorders, Abstract 653, Stockholm, Sweden, 2002; Golde et al., 32nd Annual Society for Neuroscience Conference, Abstract 722.6, Orlando, USA, 2002Eriksen et al., 32nd Annual Society for Neuroscience Conference, Abstract 722.7, Orlando, USA, 2002). These in vitro assays measure proteolytic activity due to the activity of the γ-secretase complex and are known to those skilled in the art. Compounds of formula (1) may be screened using such assays in order to identify their relative ablility to modulate β-amyloid formation.
EXAMPLE 7 SCREENING AGENTS FOR ABILITY TO DECREASE P-AMYLOID PRODUCTION AND/OR RELEASE FROM CELLS
Cell Lines and Pharmacological Treatments
293 EBNA cells stably transfected with Swedish mutant β-Amyloid Precursor Protein -695 are maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10%ι fetal bovine serum. Cells are seeded into Poly-D-Lysine coated 6-well plates at a density of 5-7 X 105 cells per well. Subsequently, the cells are rinsed in 1 ml of PBS and treated with various concentrations of APP01643 analogs in serum- free/phenol red-free DMEM for various times.
Aβ Detection and Standardization
After the pharmacological treatment, the exposure media is collected and supplemented with 10%> sample treatment buffer (40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1 % Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide), and assayed for either Aβ40 or Aβ42 by a colorimetric ELISA as per the manufacturer's protocol (Biosource International, Inc., California). The cells are lysed in 0.1%) Triton X-100 in PBS supplemented with 5 μM propridium iodide (Molecular Probes, Eugene, Oregon) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Secreted Aβ40 and Aβ42 are standardized against propridium iodide fluorescence as a measure of total cell number.
EXAMPLE 8 SCREENING AGENTS FOR ABILITY TO DECREASE β-AMYLoiD PRODUCTION FROM HUMAN NEUROBLASTOMA CELLS
Cell Lines and Pharmacological Treatment
Human neuroblastoma cells (hDAT; SK-N-MC stably overexpressing human dopamine transporter) are routinely maintained in DMEM supplemented with sodium pyruvate (1 mM) and 10%) fetal bovine serum. Cells are seeded into 6-well plates at a density of 2.5 X 105 cells per well and transiently transfected with APPsw (Swedish mutant β-amyloid precursor protein-695) using lipofectamine (Life Technologies, Rockville, Maryland) as per the manufacturer's suggested protocol. Subsequently, 48 hours post-transfection the cells are rinsed with PBS and treated with vehicle (0.1%> DMSO) or various concentrations of APP01643 analogs in serum free/phenol free DMEM for various times.
Aβ Detection and Standardization
After the pharmacological treatment, the exposure media is collected and supplemented with 10%> sample treatment buffer (40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2 mM EGTA, 0.1% sodium azide), and assayed for either Aβ40 or Aβ42 by a colorimetric ELISA as per the manufacturer's protocol (Biosource International Inc, California). The cells are lysed in 0.1% Triton X-100 in PBS supplemented with 5 μM propridium iodide (Molecular probes, Eugene, OR) and incubated at 37°C for 30 minutes prior to measuring fluorescence. Secreted Aβ40 and Aβ42 levels are standardized against propridium iodide fluorescence as a measure of total cell number.
EXAMPLE 9
SCREENING AGENTS FOR ABILITY TO DECREASE β-AMYLoiD PRODUCTION FROM PRIMARY
MURINE CORTICAL NEURONS
Semliki Forest Virus (SFV) stocks
The cDNA coding for human APP695 is cloned in the Smal site of pSFV-1 as described previously (Simons et al., J. Neurosci. 16:899-908, 1996; Tienari et al., Embo. J. 15:5218-29, 1996). PSFV-1 /huAPP695 constructs are linearized with Spel and run-off transcription using SP6 polymerase is performed to produce mRNA. The transcribed mix of APP and pSFV-helper are cotransfected into BHK cells by electroporation to yield recombinant SFV (Olkkonen et al., J. Neurosci. Res. 35:445-51 , 1993). BHK cells are grown in DMEM/F12 supplemented with 5% fetal calf serum,
2 mM L-glutamine, 100 U/ml penicillin, and 100 mg/ml streptomycin. Twenty-four hours after transfection, the culture supernatant containing infective recombinant SFV is collected. Aliquots are snap-frozen in liquid nitrogen and stored at -70°C until use.
Neuronal Culture All experiments are conducted on murine primary cortical neurons derived from E14 embryos according to established procedures (Annaert et al., J. Cell Biol. 147:277-294, 1999; Cupers et al., J. Cell Biol. 154:731-40, 2001 ; De Strooper et al., Nature 391 :387-90, 1998). Briefly, cortices of 14-day-old murine embryos are dissected, transferred to Hanks' Balanced Salt Solution (HBSS, Gibco BRL, Rockville, MD) and trypsinized for 15 minutes at 37°C. Dissociated cell suspensions are routinely plated on poly-L-lysine (I mg/ml, Sigma, St. Louis, MO) coated dishes (Nunc, Naperville, IL) in Minimal Essential Medium (MEM; Gibco BRL) supplemented with 10% horse serum and transferred to a CO2 incubator. After 3 hours, the culture medium is replaced by serum-free neurobasal medium with B27 supplement (Gibco BRL). After 24 hours, cytosine arabinoside (5 μM) was added to each dish to prevent nonneuronal (glial) cell proliferation. Three to four days post-plating, mixed cortical neuron cultures are used for drug testing.
Semliki Forest Virus Infection
Cortical neurons are incubated with increasing concentrations of compounds of formula (1) (stock solution 400 mM in DMSO). First, a concentrated dilution series is prepared in DMSO comprising 4, 20, 40 and 200 mM compound. From each of these solutions, 2.5 μl is added to the neuronal cultures in 2 ml of neurobasal medium (dilution 1/800) resulting in 5, 25, 50 and 250 μM final concentrations. As a control, 2.5 μl of DMSO is added to one dish.
After various incubation times at 37°C, the medium is replaced by 1.2 ml neurobasal medium and cultures are transduced by adding recombinant pSFV- humAPP695wt (dilution 1/10) for 1 hour to allow viral entry. Following a 2-hour incubation in the absence of virus, cultures are metabolically labeled using methionine-free neurobasal medium containing 100 μCi [35S]-methionine (ICN). After 4 hours at 37°C, the conditioned medium and the cell extracts are collected and centrifuged (14,000 rpm, 15 min).
Detection of Aβtotal from Conditioned Media
The cleared fractions are subject to immunoprecipitation with antibodies on protein G-Sepharose (Pharmacia). Aβtotal is examined from the cleared conditioned media by immunoprecipitation using pab B7, directed against the first 17 amino acids of Aβ (De Strooper et al., Embo. J. 14:4932-8, 1995). After overnight rotation, the immunoprecipitates are washed 5 times in extraction buffer and once in TBS. The bound material is denatured in sample buffer and subject to gel electrophoresis on precast 4-12%> Nupage gels. Densitometric analysis is conducted using a Phosphoimager (Molecular Dynamics) and ImagQuant 5.0. Aβtotal levels are normalized to APP levels to control for plate-to-plate variation.
Quantification of Aβ42 by ELISA The levels of the longer Aβ42 peptide are quantified in both the conditioned media and cell extracts using a sandwich ELISA test (De Strooper et al., Nature 391 :387-90, 1998; Vanderstichele et al., Amyloid. 7:245-58, 2000). In summary, 800 μl of conditioned medium or cell extract is lyophilized (Savant Speedvac concentrator), dried pellets are dissolved in 400 μl of sample diluent and applied on a 96-well ELISA plate precoated with the capturing anti-Aβ42 mab 21F12. This antibody only recognizes the final two amino acids of the Aβ42 sequence. After washing, the wells are incubated with biotin-labeled mAb 3D6 directed against the first 7 amino acids of Aβ, followed by streptavidin-HRP. Finally HRP substrate is added and the colorimetric reaction is quantitated spectrophotometrically using a Victor 2 (Wallac) equipped with a 450 nm filter. For each experiment a duplicate standard curve for Aβ42 is included. The Aβ42 concentrations in the samples are finally calculated based on the Aβ42 standards nonlinear regression equation and using Mathematica 4.1 software package (Wolfram Research, Champaign, lL).
EXAMPLE 10 SCREENING AGENTS FOR ABILITY TO DECREASE β-AMYLoiD PRODUCTION
AND/OR RELEASE IN VIVO
Upon arrival of the animals from the vendor, adult guinea pigs are housed under alternating 12 hr light/dark cycles with free access to water and food (standard laboratory chow diet). After 5-6 days adjustment to the new environment guinea pigs are anaesthetized with sodium pentobarbital and using standard sterotaxic surgical procedures, the left lateral cerebral ventricle is cannulated. After the minor surgery, the guinea pigs are given an analgesic (Bupivaicane), allowed to recover and monitored to ensure normal behavior (i.e., regular food and water intake, regular rest/activity cycles etc.). One day post-surgery, 25 μl of various doses of compounds of formula (1) diluted in phenol free DMEM supplemented with 6 % DMSO are injected into the cannula. Control animals are injected with 25 μl of phenol-free DMEM supplemented with 6% DMSO. Subsequently, at various time points post-injection, CSF is extracted through standard cisterna magna puncture and supplemented with 10%) sample treatment buffer (40 mM sodium phosphate (pH 7.4), 40 mM triethanolamine, 0.1% Triton X-100, 200 mM NaCI, 2mM EGTA, 0.1%o sodium azide), prior to freezing. After the protocol has been completed, the guinea pigs are euthanized using lethal injection of sodium pentobarbital. CSF Aβ40 and Aβ42 levels are analyzed by a colorimetric ELISA as per the manufacturers protocol (Biosource International Inc, California).
The guinea pig animal model is only one of several models known in the art that could be used. Other examples include but are not limited to, AD transgenic mice models expressing various forms of APP (Tg2576; TgAPP/Sw/1 TgAPP/Ld/2, PDAPP), presenilins or combinations of both (Tg2576 plus mutant PS1 , Tg Hu/MoAPP plus PS1) (Games, D. et al., Nature 373:523-527, 1995; Hsiao, K. H. et al., Science 214: 99-102, 1996; Moechars, D. et al., J. Biol Chem. 21 A: 6483-6492, 1999; Holcomb, L. et al., Nature Medicine 4: 97-100, 1998; Borchelt, D. R. et al., Neuron 19: 939-945, 1997).
EXAMPLE 11 SCREENING AGENTS FOR ABILITY TO PENETRATE BLOOD BRAIN BARRIER (BBB)
Using an in vitro model such as that disclosed in Franke, H. et al., Brain Res. Prot. 5:248-256, 2000, or an in vivo model such as those described by Shulkin, B. L et al., J. Neurochem. 64:1252-1257, 1995; Thorne, R.G. et al., Brain Res. 692:278- 282, 1995; Pan, W., et al., Neuropharmacol. 37:1553-1561, 1998, pharmaceutical agents of the invention can be routinely tested for their ability to penetrate the blood brain barrier. The in vitro model uses a PBEC (porcine brain microvessel endothelial cell) monolayer which is arranged so that the ability of substances to pass from a donor compartment to an acceptor compartment can be measured. This model reflects the in vivo situation wherein substances reach the brain compartment from a brain microvessel. Permeation properties of an agent of the invention are measured by radiolabeling the agent, for example with 3H, and adding it to the donor compartment. Samples are collected from the donor and acceptor compartments at routine intervals and permeability is calculated as described in Franke, H. et al., (2000).
The in vivo models measure the brain influx index or the measure of the passage of a substance through the blood brain barrier. The agent is radiolabeled or fluorescently labeled and administered peripherally by intravenous injection (Pan, W., et al., Neuropharmacol. 37:1553-1561 , 1998), orally (Shulkin, B. L. et al., J. Neurochem. 64:1252-1257, 1995) or nasally (Thorne, R.G. et al., Brain Res. 692:278-282, 1995) and the concentration of the agent in the blood as compared to the brain is monitored.
EXAMPLE 12
SCREENING AGENTS ADMINISTERED SYSTEMICALLY THAT DECREASE CNS β-AMYLoiD LEVELS
Recent evidence indicates that BBB penetrable compounds may not be required to decrease CNS β-amyloid levels. Shibata et al (J Clin Investλ 06: 1489- 1499, 2000) demonstrate that CSF Aβ can be transported across the BBB into the systemic circulation, thereby decreasing Aβ in the CNS. Once in the systemic circulation, Aβ interacts with binding proteins such as ApoJ/ApoE, which results in a decrease in "free" Aβ in the circulation and shifts the equilibrium to facilitate further transport of Aβ out of the CNS. Thus, the systemic circulation may act as a "sink" or pool of Aβ that can regulate CNS β-amybid levels (Shibata, M et al., J Clin Invest 106: 1489-1499, 2000). This "peripheral sink" hypothesis is supported by vaccination studies with anti-Aβ antibodies in AD transgenic mouse models. For example, vaccination of PDAPP mice with an Aβ antibody (m266) resulted in accumulation of CNS derived Aβ in the plasma (DeMattos et al., PNAS 98: 8850-8855, 2001 ; Holtzman et al., Adv Drug Delivery Rev 54: 1603-1613, 2002). Therefore, if compounds can systemically decrease Aβ levels, the peripheral sink hypothesis indicates that this may shift the Aβ equilibrium between the CNS and plasma resulting in a decreased β-amyloid burden in the CNS. Therefore, pharmaceutical agents of the invention can act systemically and may not be required to cross the BBB.
Using transgenic animal models described above pharmaceutical agents of the invention can be examined for their effects on systemic and CNS β-amyloid levels. Compounds can be injected into the animal of interest followed by repeated sampling and measurement of plasma β-amyloid levels over time. An increase in plasma Aβ levels coupled with a decrease in CNS levels would indicate that the compound is shifting the Aβ equilibrium. Furthermore, the ability of the compound to cross the blood brain barrier in vivo can be measured by standard analytical chemistry techniques (e.g., mass spectroscopy).
All of the above U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, are incorporated herein by reference, in their entirety. From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

A compound of formula (1):
Figure imgf000066_0001
wherein, independently at each occurrence,
Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(Re)2l heterocyclyl and heterocyclylalkyi; R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof; excluding the following compounds: propanoic acid, 2-[[4-[2-chloro-5-[3,6-dihydro-3-methyl-2,6-dioxo-4-(trifluoromethyl)-
1 (2H)-pyrimidinyl]-4-fluorophenoxy]-2-pyrimidinyl]oxy]-, methyl ester; acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2-pyrimidinyl]thio]-; and acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2-pyrimidinyl]thio]-, ethyl ester.
2. The compounds of claim 1 , further excluding: propanoic acid, 2-[[4-[(4-bromophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester; propanoic acid, 2-[[4-[(4-chlorophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[5-methoxy-4-[[3-(trifluoromethyl)phenyl]thio]-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[4-[(2-chlorophenyl)thio]-5-methoxy-2-pyrimidinyljthio]-, ethyl ester; acetic acid, [[5-methoxy-4-(4-methoxyphenoxy)-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[5-methoxy-4-(3-nitrophenoxy)-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[5-methoxy-4-(phenylthio)-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[4-(3,5-dichlorophenoxy)-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[4-(3,4-dichlorophenoxy)-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[5-methoxy-4-[3-(trifluoromethyl)phenoxy]-2-pyrimidinyl]thio]-, ethyl ester; acetic acid, [[5-methoxy-4-[(4-methoxyphenyl)thio]-2-pyrimidinyl]thio]-, ethyl ester; and acetic acid, [[4-[(4-chlorophenyl)thio]-5-methoxy-2-pyrimidinyl]thio]-, ethyl ester.
3. A compound in claim 1, wherein Y is S.
4. A compound in claim 1, wherein Y is O.
5. A compound in claim 1 , wherein W is -OR4.
6. A compound in claim 1 , wherein W is -OH.
7. A compound of claim 1 , wherein W is -N(R5)2.
8. A compound in claim 1 , wherein R3 is -H.
9. A compound in claim 1 , wherein m is 0.
10. A compound in claim 1 , wherein n is 1.
11. A compound of claim 1 , wherein Y is S and W is -OR4.
12. A compound of claim 1 , wherein R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
13. A compound of claim 1 wherein R1 has 1-10 carbons.
14. A compound of claim 1 wherein R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
15. A compound of claim 14 wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
16. A compound of claim 1 wherein R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
17. A compound of claim 1 wherein R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
18. A compound of claim 1 , wherein R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
19. A compound of claim 1 wherein R2 has 1-10 carbons.
20. A compound of claim 1 wherein R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
21. A compound of claim 20 wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 non-hydrogen atoms.
22. A compound of claim 1 wherein R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano.
23. A compound of claim 1 wherein R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
24. A compound of claim 1 wherein R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
25. A compound of claim 1 wherein R3 is selected from hydrogen, lower alkyl and lower haloalkyl.
26. A compound of claim 1 wherein, independently at each occurrence, Y is selected from the group consisting of -S- and -O-;
W is selected from the group consisting of -OR4 and -N(R5)2; p is 1 to 4; q is 1 to 2; n is 1 to 5;
R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi;
R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2); each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2l -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2;
R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
27. A composition comprising a pharmaceutically acceptable carrier, excipient or diluent and a compound of formula (1):
Figure imgf000071_0001
wherein, independently at each occurrence,
Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof; excluding the following compounds: acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2-pyrimidinyl]thio]-; and acetic acid, [[4-chloro-6-(2,3-dimethylphenoxy)-2-pyrimidinyl]thio]-, ethyl ester.
28. The composition of claim 27, wherein Y is S.
29. The composition of claim 27, wherein Y is O.
30. The composition of claim 27,wherein W is -OR4.
31. The composition of claim 27, wherein W is -OH.
32. The composition of claim 27, wherein W is -N(R5)2.
33. The composition of claim 27, wherein R3 is -H.
34. The composition of claim 27, wherein m is 0.
35. The composition of claim 27, wherein n is 1.
36. The composition of claim 27, wherein Y is S and W is -OR4.
37. The composition of claim 27, wherein R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
38. The composition of claim 27, wherein R1 has 1-10 carbons.
39. The composition of claim 27, wherein R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
40. The composition of claim 27, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
41. The composition of claim 27, wherein R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NRδC(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
42. The composition of claim 27, wherein R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
43. The composition of claim 27, wherein R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
44. The composition of claim 27, wherein R2 has 1-10 carbons.
45. The composition of claim 27, wherein R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
46. The composition of claim 45, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
47. The composition of claim 27, wherein R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano.
48. The composition of claim 27, wherein R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
49. The composition of claim 27, wherein R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
50. The composition of claim 27, wherein R3 is selected from hydrogen, lower alkyl and lower haloalkyl.
51. The composition of claim 27, wherein, independently at each occurrence,
Y is selected from the group consisting of -S- and -O-;
W is selected from the group consisting of -OR4 and -N(R5)2; p is 1 to 4; q is 1 to 2; n is 1 to 5;
R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2); each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2;
R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
52. A method for modulating the production and/or release of β-amyloid in a cell, comprising treating said cell with a compound of formula (1)
Figure imgf000077_0001
wherein, independently at each occurrence,
Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi; R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
53. A method of claim 52, wherein Y is S.
54. A method of claim 52, wherein Y is O.
55. A method of claim 52, wherein W is -OR4.
56. A method of claim 52, wherein W is -OH.
57. A method of claim 52, wherein W is -N(R5)2.
58. A method of claim 52, wherein R3 is -H.
59. A method of claim 52, wherein m is 0.
60. A method of claim 52, wherein n is 1.
61. A method of claim 52, wherein Y is S and W is -OR4.
62. A method of claim 52, wherein R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
63. A method of claim 52, wherein R1 has 1-10 carbons.
64. A method of claim 52, wherein R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
65. A method of claim 64, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
66. A method of claim 52, wherein R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
67. A method of claim 52, wherein R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
68. A method of claim 52, wherein R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
69. A method of claim 52, wherein R2 has 1-10 carbons.
70. A method of claim 52, wherein R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
71. A method of claim 52, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
72. A method of claim 52, wherein R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano.
73. A method of claim 52, wherein R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
74. A method of claim 52, wherein R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
75. A method of claim 52, wherein R3 is selected from hydrogen, lower alkyl and lower haloalkyl.
76. A method of claim 52, wherein, independently at each occurrence, Y is selected from the group consisting of -S- and -O-;
W is selected from the group consisting of -OR4 and -N(R5)2; p is 1 to 4; q is 1 to 2; n is 1 to 5;
R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi;
R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2); each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2;
R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
77. A method of any one of claims 52-76 wherein said cell is a brain cell.
78. A method of any one of claims 52-76 wherein said β-amyloid is β-amyloid 42.
79. A method of any one of claims 52-76 wherein β-amyloid production and/or release in the cell is reduced.
80. A method of any one of claims 52-76 wherein said cell is treated in vitro.
81. A method of treatment comprising modulating the production and/or release of β-amyloid in a non-human mammal in need of said treatment, said method comprising administering to said non-human mammal a compound of formula (1)
Figure imgf000082_0001
wherein, independently at each occurrence,
Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, -N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi;
R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
82. A method of claim 81 , wherein Y is S.
83. A method of claim 81 , wherein Y is O.
84. A method of claim 81 , wherein W is -OR4.
85. A method of claim 81 , wherein W is -OH.
86. A method of claim 81 , wherein W is -N(R5)2.
87. A method of claim 81 , wherein R3 is -H.
88. A method of claim 81 , wherein m is 0.
89. A method of claim 81 , wherein n is 1.
90. A method of claim 81 , wherein R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
91. A method of claim 81 , wherein R1 has 1-10 carbons.
92. A method of claim 81 , wherein R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
93. A method of claim 92, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
94. A method of claim 81 , wherein R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
95. A method of claim 81 , wherein R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
96. A method of claim 81 , wherein R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
97. A method of claim 81 , wherein R2 has 1 -10 carbons.
98. A method of claim 81 , wherein R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
99. A method of claim 81 , wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
100. A method of claim 81 , wherein R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano.
101. A method of claim 81 , wherein R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
102. A method of claim 81 , wherein R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
103. A method of claim 81 , wherein R3 is selected from hydrogen, lower alkyl and lower haloalkyl.
104. A method of claim 81 , wherein, independently at each occurrence,
Y is selected from the group consisting of -S- and -O-;
W is selected from the group consisting of -OR4 and -N(R5)2; p is 1 to 4; q is 1 to 2; n is 1 to 5;
R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi;
R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O),N(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2); each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2;
R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi; R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
105. A method of any one of claims 81-104 wherein β-amyloid production and/or release in a brain cell is modulated.
106. A method of any one of claims 81 -104 wherein said β-amyloid is β-amyloid 42.
107. A method of any one of claims 81 -104 wherein said non-human mammal is a mouse, dog or guinea pig.
108. A method of any one of claims 81 -104 wherein β-amyloid production and/or release in a cell is reduced.
109. A method of treatment wherein the production and/or release of β- amyloid is modulated in a human in need of said treatment, said method comprising administering to said human a compound of formula (1)
Figure imgf000088_0001
wherein, independently at each occurrence,
Y is selected from the group consisting of -S(O)m- and -O-;
W is selected from the group consisting of -OR4, ~N(R5)2 and -NHN(R5)2; p is 0 to 5; q is 0 to 2; m is 0 to 2; n is 0 to 5; each R1 and R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi; each R3 is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, heterocyclyl and heterocyclylalkyi; R4 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceutically acceptable salt thereof.
110. A method of claim 109, wherein Y is S.
111. A method of claim 109, wherein Y is O.
112. A method of claim 109, wherein W is -OR4.
113. A method of claim 109, wherein W is -OH.
114. A method of claim 109, wherein W is -N(R5)2.
115. A method of claim 109, wherein R3 is -H.
116. A method of claim 109, wherein m is 0.
117. A method of claim 109, wherein n is 1.
118. A method of claim 109, wherein Y is S and W is -OR4.
119. A method of claim 109, wherein R1 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
120. A method of claim 109, wherein R1 has 1-10 carbons.
121. A method of claim 109, wherein R1 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
122. A method of claim 121 , wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
123. A method of claim 109, wherein R1 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2l -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
124. A method of claim 109, wherein R1 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
125. A method of claim 109, wherein R2 is a hydrocarbon selected from the group consisting of alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl and cycloalkylalkyl.
126. A method of claim 109, wherein R2 has 1 -10 carbons.
127. A method of claim 109, wherein R2 in at least one occurrence is selected from the group consisting of heteroalkyl, heteroalkenyl and heteroalkynyl.
128. A method of claim 109, wherein the selected heteroalkyl, heteroalkenyl and heteroalkynyl group has less than 10 atoms.
129. A method of claim 109, wherein R2 in at least one occurrence is selected from the group consisting of halo, haloalkyl, haloalkoxy, nitro and cyano.
130. A method of claim 109, wherein R2 in at least one occurrence is selected from the group consisting of -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7 and -NR6S(O)tR6 (where t is 0 to 2).
131. A method of claim 109, wherein R2 in at least one occurrence is selected from the group consisting of heterocyclyl and heterocyclylalkyi.
132. A method of claim 109, wherein R3 is selected from hydrogen, lower alkyl and lower haloalkyl.
133. A method of claim 109, wherein, independently at each occurrence, Y is selected from the group consisting of -S- and -O-;
W is selected from the group consisting of -OR4 and -N(R5)2; p is 1 to 4; q is 1 to 2; n is 1 to 5;
R1 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2), heterocyclyl and heterocyclylalkyi;
R2 has a formula weight of less than 500 and is selected from the group consisting of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -S(O)tR6 (where t is 0 to 2), -S(O)tN(R6)2 (where t is 0 to 2), -OC(S)NR6, -NR6C(S)OR7, -NR6S(O)tR6 (where t is 0 to 2); each R3 has a formula weight of less than 200 and is selected from the group consisting of hydrogen, alkyl, alkenyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, halo, haloalkyl, haloalkoxy, nitro, cyano, -NHOH, -OR7, -SR7, -C(O)OR7, -OC(O)R7, -C(O)N(R6)2, -C(S)R6, -C(O)R6, -N(R6)2, -N(R6)C(O)R6, -N(R6)C(O)OR7, -OC(S)NR6, -NR6C(S)OR7, -OR7, -C(O)OR7, -OC(O)R7, and -C(O)N(R6)2;
R4 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R5 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, haloalkyl, haloalkoxy, heterocyclyl and heterocyclylalkyi;
R6 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aralkyl and aryl; and
R7 has a formula weight of less than 500 and is selected from the group consisting of hydrogen, alkyl and aralkyl; as a single stereoisomer, a mixture of stereoisomers, as a racemic mixture of stereoisomers; as a solvate, as a polymorph; or as a pharmaceufically acceptable salt thereof.
134. A method of any one of claims 109-133 wherein said human is afflicted with Alzheimer's disease.
135. A method of any one of claims 109-133 wherein said human has suffered a head injury.
136. A method of any one of claims 109-133 wherein said human has a genetic predisposition or environment exposure that increases the likelihood that said person will develop Alzheimer's disease.
136. A method of any one of claims 109-133 wherein said human exhibits minimal cognitive impairment suggestive of early stage Alzheimer's disease.
137. A method of any one of claims 109-133 wherein β-amyloid production and/or release in a brain cell is modulated.
138. A method of any one of claims 109-133 wherein said β-amyloid is β-amyloid 42.
139. A method of any one of claims 109-133 wherein β-amyloid production in the human, or β-amyloid release from a cell in the human, is reduced.
PCT/CA2003/001962 2002-12-16 2003-12-16 Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses WO2004054988A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003287840A AU2003287840A1 (en) 2002-12-16 2003-12-16 Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43403002P 2002-12-16 2002-12-16
US60/434,030 2002-12-16

Publications (1)

Publication Number Publication Date
WO2004054988A1 true WO2004054988A1 (en) 2004-07-01

Family

ID=32595259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2003/001962 WO2004054988A1 (en) 2002-12-16 2003-12-16 Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses

Country Status (2)

Country Link
AU (1) AU2003287840A1 (en)
WO (1) WO2004054988A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007125351A1 (en) * 2006-04-27 2007-11-08 Senexis Limited Pyrimidine derivatives for the treatment of amyloid-related diseases

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002100836A2 (en) * 2001-06-12 2002-12-19 Active Pass Pharmaceuticals, Inc. Compounds, compositions and methods for modulating beta-amyloid production

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002100836A2 (en) * 2001-06-12 2002-12-19 Active Pass Pharmaceuticals, Inc. Compounds, compositions and methods for modulating beta-amyloid production

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GAETANO D'ARTI: "NOVEL PYRIMIDINE AND 1,3,5-TRIAZINE HYPOLIPIDEMIC AGENTS.", JOURNAL OF MEDICINAL CHEMISTRY., vol. 27, no. 12, 1984, AMERICAN CHEMICAL SOCIETY. WASHINGTON., US, pages 1621 - 9, XP002273042, ISSN: 0022-2623 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007125351A1 (en) * 2006-04-27 2007-11-08 Senexis Limited Pyrimidine derivatives for the treatment of amyloid-related diseases

Also Published As

Publication number Publication date
AU2003287840A1 (en) 2004-07-09

Similar Documents

Publication Publication Date Title
US8088778B2 (en) Benzisoxazole piperazine compounds and methods of use thereof
US7482460B2 (en) Doxepin analogs and methods of use thereof
US7687665B2 (en) 2-methylprop anamides and their use as pharmaceuticals
US7524864B2 (en) Methods of treating sleep disorders
US7763612B2 (en) Therapeutic amine-arylsulfonamide conjugate compounds
RU2501792C2 (en) Compounds, compositions and methods for treating beta-amyloid diseases and synucleinopathies
JPH09500883A (en) Piperazine derivatives as α1A-adrenoceptor antagonists
EP0858996A1 (en) Nerve cell protective agents
US7326721B2 (en) Doxepin analogs and methods of use thereof
US9802897B2 (en) Amidopyridinol derivative or pharmaceutically acceptable salt thereof and pharmaceutical composition comprising same as active component
US20030125338A1 (en) Compounds, compositions and methods for modulating beta-amyloid production
CA2564642A1 (en) Lxr receptor modulators
US20030191144A1 (en) Compounds, compositions and methods for modulating beta-amyloid production
WO2004054988A1 (en) Arylthioetherpyrimidine and aryloxyetherpyrimidine derivatives and their therapeutic uses
JP2005289808A (en) 3-substituted-4-pyrimidone derivative
US20070232619A1 (en) Piperazine Derivatives of Dialkyl Oxindoles
US10287265B2 (en) Enantiomers of 8-hydroxyquinoline derivatives and the synthesis thereof
US9725429B2 (en) Furanyl-imine and thiofuranyl-imine compounds for stabilizing ryanodine receptors from aberrant levels of calcium release
US8268884B2 (en) Derivatives of substituted tartaric acid and usage for preparing beta-secretase inhibitors
KR20080022940A (en) Preparation method of triethylene tetramine dihydrochloride
US20050143348A1 (en) Doxepin analogs and methods of use thereof
AU2002312673A1 (en) Compounds, compositions and methods for modulating beta-amyloid production
US10870633B2 (en) Types of C-3 substituted kynurenic acid derivatives with improved neuroprotective activity
JPH04316561A (en) New imidazole compound
Kumar et al. Newer Indolyl antiparkinsonian agents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 69(1) EPC. (EPO COMMUNICATION FORM 1205 HAS BEEN SENT ON 21/12/2005)

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP